You are on page 1of 246

BIOCHEMISTRY RESEARCH TRENDS

PALMITIC ACID
OCCURRENCE, BIOCHEMISTRY
AND HEALTH EFFECTS

No part of this digital document may be reproduced, stored in a retrieval system or transmitted in any form or
by any means. The publisher has taken reasonable care in the preparation of this digital document, but makes no
expressed or implied warranty of any kind and assumes no responsibility for any errors or omissions. No
liability is assumed for incidental or consequential damages in connection with or arising out of information
contained herein. This digital document is sold with the clear understanding that the publisher is not engaged in
rendering legal, medical or any other professional services.

BIOCHEMISTRY RESEARCH TRENDS


Additional books in this series can be found on Novas website
under the Series tab.

Additional e-books in this series can be found on Novas website


under the e-book tab.

BIOCHEMISTRY RESEARCH TRENDS

PALMITIC ACID
OCCURRENCE, BIOCHEMISTRY
AND HEALTH EFFECTS

LUCAS F. PORTO
EDITOR

New York

Copyright 2014 by Nova Science Publishers, Inc.


All rights reserved. No part of this book may be reproduced, stored in a retrieval system or
transmitted in any form or by any means: electronic, electrostatic, magnetic, tape, mechanical
photocopying, recording or otherwise without the written permission of the Publisher.
For permission to use material from this book please contact us:
Telephone 631-231-7269; Fax 631-231-8175
Web Site: http://www.novapublishers.com
NOTICE TO THE READER
The Publisher has taken reasonable care in the preparation of this book, but makes no expressed or
implied warranty of any kind and assumes no responsibility for any errors or omissions. No
liability is assumed for incidental or consequential damages in connection with or arising out of
information contained in this book. The Publisher shall not be liable for any special,
consequential, or exemplary damages resulting, in whole or in part, from the readers use of, or
reliance upon, this material. Any parts of this book based on government reports are so indicated
and copyright is claimed for those parts to the extent applicable to compilations of such works.
Independent verification should be sought for any data, advice or recommendations contained in
this book. In addition, no responsibility is assumed by the publisher for any injury and/or damage
to persons or property arising from any methods, products, instructions, ideas or otherwise
contained in this publication.
This publication is designed to provide accurate and authoritative information with regard to the
subject matter covered herein. It is sold with the clear understanding that the Publisher is not
engaged in rendering legal or any other professional services. If legal or any other expert
assistance is required, the services of a competent person should be sought. FROM A
DECLARATION OF PARTICIPANTS JOINTLY ADOPTED BY A COMMITTEE OF THE
AMERICAN BAR ASSOCIATION AND A COMMITTEE OF PUBLISHERS.
Additional color graphics may be available in the e-book version of this book.

Library of Congress Cataloging-in-Publication Data


ISBN:  (eBook)

Library of Congress Control Number: 2014944853

Published by Nova Science Publishers, Inc. New York

CONTENTS
Preface

vii

Chapter 1

Fatty Acids in Vascular Health


Reggie Hui-Chao Lee, Carl S. Wilkins, Alexandre Couto e Silva,
Stephen E. Valido, Celeste Yin-Chieh Wu and Hung Wen Lin

Chapter 2

Occurrence, Biochemical, Antimicrobial and Health Effects


of Palmitic Acid
Melissa Johnson and Daniel A. Abugri

17

Palmitic Acid: Effect of Diet Supplementation and Occurrence


in Animal Origin Food
P. G. Peiretti

45

Chapter 3

Chapter 4

General Aspects of Palmitic Acid


Deusdlia Teixeira de Almeida, Mariana Melo Costa
and Sabrina Feitosa

Chapter 5

Palmitic Acid As a Cardiometabolic Risk Factor


Danijela Risti-Medi and Vesna Vui

105

Chapter 6

Palmitic Acid in Higher Plant Lipids


R.A. Sidorov, A.V. Zhukov, V.P. Pchelkin
and V.D. Tsydendambaev

125

Chapter 7

Palmitic Acid in Infant Nutrition


Fabiana Bar-Yoseph, Yael Lifshitz, Tzafra Cohen
and Ita Litmanovitz

145

Chapter 8

Processing of Palmitic Acid and Its Derivatives Using


Supercritical Fluids
C. E. Schwarz

Chapter 9
Index

Palmitic Acid in Tunisian Olive Oil: Updating and Perspective


Ghayth Rigane and Ridha Ben Salem

63

159
211
219

PREFACE
This book discusses the occurrence, biochemistry, and health effects of palmitic acid.
Chapter 1 - Fatty acids have traditionally been described as artery clogging species that is
detrimental to overall health. The most prevalent fatty acid is palmitic acid (PA), a sixteen
carbon chain fatty acid that is ubiquitous in biological systems. PA is prevalent in most
eukaryotic cell membranes and in the mitochondria derived from the Krebs cycle utilizing
acetyl-coenzyme A as its precursor. PA is found in a variety of plants with a high amounts in
coconut oil. Many cosmetics, shampoos, and commercialized beauty products contain PA
providing structure and substance to the gel or reagent.
An emerging field of study is the esterified form of PA or methyl palmitate, as it is
involved in biological signaling in the central nervous system. More specifically, methyl
palmitate or palmitic acid methyl ester can cause arterial vasodilation and is thought to be
involved in neurotransmission, as well as modulate vascular tonicity in cerebral circulation.
Methyl palmitate has also been implicated as a neuroprotective agent in both models of focal
and global cerebral ischemia; however, the exact mechanism(s) are still unknown. The
authors will focus on the known pharmacology, biochemistry, and clinical implications of PA
and other related fatty acids (i.e. Non-esterified v. esterified fatty acids) commonly found in
daily diets. Additionally, cellular target(s) of PA will be discussed as it relates to
improvement of disease states, synthesis, and possible health implications/benefits of methyl
palmitate in biological systems.
Chapter 2 - Palmitic acid (PA), one of the most abundant saturated fatty acid (SFAs)
within plants, humans, animals, microbial (bacteria), fungal, and marine organisms,
constitutes ~16 to 45 % of the lipid profile. The impressive abundance of PA throughout
nature could be attributed partly to its critical role in membrane lipid structural functionality,
formation of subcellular cysteine residue linkages and RNA posttranslational modifications in
eukaryotic and prokaryotic cells. PA has been demonstrated to undergo -oxidation to
produce short and medium chain fatty acids to maintain homeostasis in response to
endogenous and exogenous cues. Further, non-esterified PA is known to mediate numerous
biochemical and antimicrobial pathways towards the betterment of human health. Although
the importance of PA in human health and nutrition are established, critics attest that
excessive dietary PA may be unhealthy and even detrimental. Current microbiological and
epidemiological studies suggest that PA produces specific health benefits that are not
common to all other SFAs. For example, studies suggest that PA may activate nitric oxide
and superoxide, thus functioning as an antimicrobial agent against some strains of bacteria,

viii

Lucas F. Porto

algae, and helminthes and certain foodborne pathogens. This chapter reviews the occurrence,
biochemistry, and subsequent health implications of PA.
Chapter 3 - In the last few decades, disagreement between opinions and findings
concerning the ability of palmitic acid (PA) and other saturated fatty acids (SFAs) to raise
cholesterolaemia has led to discussions on whether PA, which has been positively related to
high serum cholesterol levels, could increase the risk of cardiovascular diseases. This study
aims to review the PA content of meat, dairy products, fish, and other food of animal origin in
the human diet and discusses nutritional issues related to the occurrence of this fatty acid
(FA) in these foods due to different diet supplementation. Meat and dairy products are
considerable dietary sources of SFAs, such as PA. In most industrialized countries, a high
meat or dairy intake contributes to a higher than recommended SFA intake. Palmitic and
myristic acids are common FAs in meat and dairy products, making up about 30-40% of total
FA intake and are the main factors responsible for raising cholesterol levels; indeed, strong
evidence indicates that these two SFAs increase serum cholesterol concentrations in humans.
Stearic acid is partially converted to oleic acid in vivo and has not been shown to elevate
blood cholesterol, while lauric acid is not as potent as PA at raising concentrations of total
cholesterol and LDL cholesterol in humans. The occurrence of PA in animal origin food is
influenced by both genetic and environmental factors, such as the composition of the animals
diet, its digestive system and its biosynthetic processes. The FA profile in food of animal
origin mainly reflects dietary lipid sources and has the potential to play a valuable role in
human nutrition by manipulating the composition of animal fat through diet. In order to
explain the variability in FA composition in food of animal origin, this review examines
different nutrition trials that have studied the effects of PA supplementation on the lipid
profile of animal origin food.
Chapter 4 - Palmitic acid or hexadecanoic acids is the most abundant saturated fatty acid
in human nutrition and represents about 17.6g per day in the United Kington diet. It is the
first fatty acid produced during the lipogenesis. During this process, glucose is converted to
fatty acids, which then react with glycerol to produce triacylglycerols. Palmitic acid mainly
occurs as its ester in triglycerides, especially in palm oil (40-44 %) but also in lard (20-30 %),
dairy products (25-40 %) and cocoa butter (25-27 %). One of the main applications of
palmitic acid in the food industry has been the formulation of interesterified fats, used as a
replacement of trans fats. In breast milk, native lard, enzyme-directed and randomly
chemically interesterified plant fats, palmitic acid is predominantly esterified to
triacylglycerol, center or -position, in native palm oil and cows milk, it is mainly at the
external or -positions. A higher palmitic acid absorption is obtained with formulas rich in
palmitic acid esterified in triacylglycerol sn-2 position, than with those containing palmitic
acid predominantly esterified in the sn-1,3 positions. These specific fatty acids distributions in
triacylglycerol, determine the physical properties of the fat, which affects its absorption,
metabolism and distribution into tissues. Many authors claim that a palmitic acid intake may
promote increased risk of hypercholesterolemia, liver disease, type 2 diabetes, insulin
resistance and toxicity. However, more recent investigations on the topic seem to have
reconsidered the negative role of the dietary saturated fatty acids as a risk factor for
cardiovascular diseases and show that not only the type of fat, but also that the triglyceride
structure plays a role in these diseases.
Chapter 5 - Current dietary recommendations are based on a reduced saturated fatty acid
(SFA) consumption to prevent cardiovascular disease (CVD). The role of individual SFA in

Preface

ix

metabolic disease is not fully understandable. One type of SFA present in many common
foods (dairy, meat, palm and coconut oil) is palmitic acid (16:0). A number of
epidemiological studies have shown that the populations who consume large amounts of
atherogenic SFA (especially palmitic, myristic, lauric) have elevated levels of LDL and HDLcholesterol. Saturated fatty acid exert their atherogenic and thrombogenic effect through
increased production of LDL, very-low-density lipoproteins particles and apolipoproteins A1,
with a decrease of LDL- receptors specific activity, and an increase in platelet aggregation.
The total cholesterol/ HDL-cholesterol ratio, the best overall indication of potential effects on
coronary heart disease (CHD) risk is nonsignificantly affected by consumption of palmitic
acid (PA). Compared with lipid effects, the influence of SFA intake on inflammation markers
is less well explored. The associations between circulating and tissue PA and dietary intake of
PA are diverse and most likely reflecting endogenous metabolism. Status of PA is not in
intakeresponse relationship biomarker, probably partly due to conversion of 16:0 to 16:1 by
steaoryl-CoA-desaturase (SCD-1). Increased SFA intake has been associated with increased
SCD-1 activity in which may predict mortality. Palmitoylation is the process involved in
proteinmembrane interactions and signal transduction. Increases in dietary intake of PA
decrease fat oxidation and daily energy expenditure with slight increases in adiposity.
Evidence for the effects of SFA, particularly PA consumption on insulin resistance, vascular
function, type 2 diabetes, and stroke is various. It is considered that circulating PA, as
nonesterified fatty acids stimulate insulin resistance by decreasing phosphorylation of the
insulin receptor and insulin receptor substrate-1. In muscle cells, PA decrease oxidation of
fatty acids and glucose which elevates fatty acid and glucose levels in tissues and blood, and
decreases adiponectin production, which may both promote insulin resistance. It was shown
that 16:0 and 14:0 stimulate -cells and endothelial dysfunction. The incidence of type 2
diabetes was associated with total SFA levels of plasma cholesterol esters (also demonstrated
for 16:0 levels independently) and phospholipids (also for 16:0 and 18:0). In skeletal muscle
phospholipids, PA has been negatively associated with insulin sensitivity and diabetes type 2.
Systematic reviews on prospective cohort studies indicated that CHD risk has not been
directly associated with SFA intake, although is associated with a dietary habits, high in SFArich foods. Taken together, there is collective convincing evidence for decreased CHD risk
when replacing SFA with polyunsaturated fats. Differences in cardiometabolic risk appear
greater between food groups and overall dietary patterns rather than between separate SFA.
Chapter 6 - Palmitic acid (C16:0) is one of the major fatty acids (FAs) forming virtually all
natural lipids. Both in eu-, and prokaryotes, C16:0 forms various lipid classes, which serve
either as the lipid background of storage fats and oils, or the hydrophobic matrix of cell
membranes, or the components of cuticle waxes and polymers. Non-esterified 16:0 does not
occur in living cells, and it is present there only as an acyl residue in various lipid classes,
such as mono-, di-, and triacylglycerols, glyco-, phospho-, and sphingolipids, wax and steryl
esters etc., where it esterifies the hydroxy groups of glycerol backbone or other alcohols
(sphingosine, higher and lower aliphatic alcohols etc.). Palmitic acid is known to be a primary
higher FA synthesized in the cell, while nearly all other FAs of natural lipids are the products
of its further modification caused by elongation, desaturation, insertion of various functional
groups, such as methyl, hydroxy, oxo, epoxy, etc. As a saturated FA, C16:0 is used by the cell
for regulating its functional state by shifting the membrane fluidity under adverse
environmental conditions and thus providing a necessary molecular species composition of
the membrane polar lipids. Among the latter, such classes as phosphatidylinositols,

Lucas F. Porto

phosphatidylserines, and other highly polar lipids are particularly rich in palmitic acid. In
accordance, its content in plant lipids rises as they became less TLC-mobile, more difficultly
extractable, or tightly bound. It is evident that further screening of plant lipids as regards this
index is of considerable interest.
Chapter 7 - Human breast milk provides the optimum nutrition for infants. Designed to
provide balanced nutrition, human breast milk naturally meets the needs of growing infants in
the first months after birth. In human breast milk, and in most infant formulas, approximately
50% of the energy is supplied to newborns as fat, of which more than 98% is in the form of
triglycerides. Triglyceride synthesis occurs in the mammary gland. The fatty acids are
specifically positioned to sn1, sn2 or sn3 positions on the glycerol backbone to yield the
structure-specific triglycerides that are found in human milk. Palmitic acid (C16:0) is the
predominant saturated fatty acid, comprising 17-25% of the fatty acids in mature human milk.
Surprisingly, the positioning of palmitic acid is highly conserved across populations, and
approximately 70-75% of palmitic fatty acids are esterified to the sn2 position of the
triglyceride (sn2 palmitate).
Clinical and pre-clinical studies over the last three decades have provided increasing
evidence that this specific positioning of palmitic acid on the triglycerides in human milk has
a significant holistic effect on optimal infant development and well-being that is related to the
increased absorption of both palmitic acid and calcium: softer stools, increased bone strength,
increased beneficial gut flora, controlled intestinal health, and reduced infant crying. All of
these contribute to the benefits of infant wellbeing.
The overall aim of the current review is to expand the understanding of the role of
palmitic acid and its specific sn2 position in infant nutrition.
Chapter 8 - Palmitic acid, either in its triglyceride form or hydrolysed as a free fatty acid
or an ester, needs to be extracted from its source, processed and isolated to obtain useful
products. The objective of this work is to consider the use of SCF (supercritical fluid)
processing as a method to extract and process palmitic acid and/or its derivatives. A phase
behaviour analysis, in supercritical CO2, ethane and propane, at temperatures close to the
critical point of the solvent show moderate solubility of palmitic acid and tripalmitin at
pressures below 50 MPa and total solubility of methyl and ethyl palmitate at pressures below
25 MPa. Analysis of the phase behaviour considered and studies presented in the literature
have shown that SCFs can be widely applied to the processing of palmitic acid containing
compounds. In particular SCFs can fractionate a mixture of acids or their derivatives
according to the chain length, it can de-acidify an edible oil and it is able to fractionate a
mixture containing palmitic acid and other compounds. Additionally, SCFs can also be used
to extract palmitic acid containing triglycerides from plant material. SCFs, in particular CO2,
are thus excellent alternative solvent to traditional organic solvents and should be considered
when processing palmitic acid containing products.
Chapter 9 - In this review the major saturated fatty acid, palmitic acid, of Virgin Olive
Oil (VOO) was studied. This oil is one of the oldest known vegetable oils and it plays a
fundamental role in human nutrition around the Mediterranean basin. This nature juice is the
only edible oil of great production obtained by physical methods from the fruit Olea europaea
L. Consideration of VOO as a natural functional fat is related to the presence of palmitic acid.
Updating of its levels in Virgin olive oils throughout the Tunisian olive oil as well as
information on expecting levels in other products from olive tree establish the authors view
point. Studies on levels palmitic acid upon maturity stage in the oil are also discussed.

Preface

xi

Major analytical practices are given in brief. Palmitic acid (C16:0) is the principal
saturated fatty acid in olive oil, responsible for its figeability at low temperature.
Few are the exceptions as palmitic acid content depends heavily on the genetic factor.
Palmitic fatty acids, important for the nutritional properties of an olive oil, showed a crucial
rule in the characterization of olive oils.

In: Palmitic Acid: Occurrence, Biochemistry and Health Effects ISBN: 978-1-63321-519-1
Editor: Lucas F. Porto
2014 Nova Science Publishers, Inc.

Chapter 1

FATTY ACIDS IN VASCULAR HEALTH


Reggie Hui-Chao Lee1, Carl S. Wilkins2, Alexandre Couto e Silva1,
Stephen E. Valido1, Celeste Yin-Chieh Wu1 and Hung Wen Lin1,
1

Cerebral Vascular Disease Laboratories, Department of Neurology, University of Miami


Miller School of Medicine, Miami, FL, US
2
Florida International University Herbert Wertheim College
of Medicine, Miami, FL, US

ABSTRACT
Fatty acids have traditionally been described as artery clogging species that is
detrimental to overall health. The most prevalent fatty acid is palmitic acid (PA), a
sixteen carbon chain fatty acid that is ubiquitous in biological systems. PA is prevalent in
most eukaryotic cell membranes and in the mitochondria derived from the Krebs cycle
utilizing acetyl-coenzyme A as its precursor. PA is found in a variety of plants with a
high amounts in coconut oil. Many cosmetics, shampoos, and commercialized beauty
products contain PA providing structure and substance to the gel or reagent.
An emerging field of study is the esterified form of PA or methyl palmitate, as it is
involved in biological signaling in the central nervous system. More specifically, methyl
palmitate or palmitic acid methyl ester can cause arterial vasodilation and is thought to be
involved in neurotransmission, as well as modulate vascular tonicity in cerebral
circulation. Methyl palmitate has also been implicated as a neuroprotective agent in both
models of focal and global cerebral ischemia; however, the exact mechanism(s) are still
unknown. We will focus on the known pharmacology, biochemistry, and clinical
implications of PA and other related fatty acids (i.e. Non-esterified v. esterified fatty
acids) commonly found in daily diets. Additionally, cellular target(s) of PA will be
discussed as it relates to improvement of disease states, synthesis, and possible health
implications/benefits of methyl palmitate in biological systems.

Corresponding author: Hung Wen Lin, Ph.D. University of Miami, Miller School of Medicine, Department of
Neurology, Cerebral Vascular Disease Research Center, Two Story Laboratory (TSL), Medical Campus,
Locator: D4-5, 1420 N.W. 9th Avenue, Miami, FL 33136. E-mail: hwlin@med.miami.edu.

Reggie Hui-Chao Lee, Carl S. Wilkins, Alexandre Couto e Silva et al.

Keywords: Palmitic acid methyl ester, methyl palmitate, stearic acid, stearic acid methyl
ester, cerebral ischemia, stroke, vasodilation

INTRODUCTION
High fatty acid content has been commonly associated with an increased risk of
cardiovascular diseases in humans [1]. The most prevalent of fatty acids are palmitic (16
carbon) and stearic (18 carbon) acids, which are fatty acids ubiquitously present in biological
systems abundant in eukaryotic cell membranes. Palmitic acid (PA) is the most common
saturated fatty acid found in various organisms. Synthesis of PA is well-known and occurs
naturally in mammalian cells via acetyl-coenzyme A (CoA) and malonyl-CoA precursors.
Alternatively, the synthesis and function of the esterified form of palmitic and stearic acid,
palmitic acid methyl ester (PAME) and stearic acid methyl ester (SAME), has not been welldocumented (Figure 1) [2]. Only recently has PAME and SAME been introduced to the
forefront of fatty acid research in biology and disease. Specifically, PAME results in aortic
vasodilation and neuroprotection, while SAME causes neuroprotection all in the context of
cerebral ischemia. Thus far, PAME and SAME are thought to act as a neurotransmitter/
modulator released from a neuronal source (SCG, superior cervical ganglion SCG) [3].

Figure 1. Structures of palmitic acid, stearic acid, PAME, and SAME.

PALMITIC ACID METHYL ESTER (PAME)


PA is the most common saturated fatty acid (16:0) and the main component in many
types of cellular membranes [4]. Recent studies suggest that fatty acids may have vasoactive
properties involved in circulation. Interestingly, PA alone does not have any vasoactive
properties, but methylated PA, also known as methyl palmitate or PAME, has been reported
to be a potent vasodilator [5-7]. PAME was first discovered in the superior cervical ganglion
(SCG). The SCG is the largest of the three ganglions in the sympathetic chain, and is thought
to be the origin of innervations of cerebral blood vessel [3]. Field electrical stimulation of the
SCG caused simultaneously release of PAME and SAME [5] further enhanced by the
presence of arginine analogs such as L-arginine and nitric oxide synthase (NOS) inhibitor

Fatty Acids in Vascular Health

(N-nitro-L-arginine). Subsequent investigations have led to the discovery of PAME in other


locale such as perivascular adipose tissue (PVAT), and the retina inducing strong aortic
vasodilation [5-7] suggesting that PAME plays a crucial role in regulating systemic
circulation and cerebral blood flow (CBF) in the retina and other peripheral circulation in
vitro. Since PAME is an abundant component in PVAT and the retina, it is highly likely that
PAME is the PVAT-derived relaxing factor as well as the retina-derived relaxing factor,
respectively [6, 7]. Furthermore, the EC50 (half maximal effective concentration) for PAMEinduced aortic vasodilation is 1.92 (0.46-7.93) x10-10 M which is much lower than other
known nitric oxide (NO, one of the most potent vasodilators) donors such as sodium
nitroprusside, 1-[2-(carboxylato)pyrrolidin-1-yl]diazen-1-ium-1,2-diolate, and N-[4-[1-(3Aminopropyl)-2-hydroxy-2-nitrosohydrazino]butyl]-1,3-propanediamine [5, 8, 9] indicating
that PAME is a novel and potent vasodilator.
The exact mechanism(s) underlying PAME-induced vasodilation has not been welldefined. However, PAME-induced vasodilation is inhibited by voltage-gated potassium (Kv)
channel blockers such as tetraethylammonium and 4-aminopyridine [7] suggesting that
PAME may regulate vascular tone via opening of Kv channels hyperpolarizing vascular
smooth muscle cells (VSMCs) in blood vessels. The release of PAME from PVAT is
significantly reduced in genetically-altered hypertensive animals (spontaneously hypertensive
rats, SHR) [7] indicating that PAME may be involved in blood pressure regulation. Focal or
global ischemia-induced hypoperfusion that lasts from hours to days is one of the contributors
to neuronal cell death and neurocognitive (learning/memory) deficits after ischemia [10,
11].Administration of PAME alleviates focal and global ischemia-induced hypoperfusion
(hypoperfusion decreases CBF) to reduce cerebral injury after ischemia due to the fact that
PAME causes potent vasodilator [12].

ARACHIDONIC ACID (AA)


Besides PAME and SAME, other fatty acids have also been shown to have vasoactive
properties. For example, AA is a 20-carbon polyunsaturated -6 fatty acid (20:4 n-6), which
is an essential fatty acid found in peanut oil and eggs/meats. AA has been shown to cause
endothelium- and K+-independent vasodilation [13] metabolized by cytochrome P450
enzymes in the liver to form epoxyeicosatrienoic acid (EET) (Figure 2). EETs are produced in
the vascular endothelium and causes potent vasodilation in renal, mesenteric, coronary, and
cerebral arterioles in a variety of mammalian species [14-17]. EETs hyperpolarize VSMCs by
activating calcium-activated potassium channels (KCa) channels. Several investigators have
proposed that EET may serve as a possible candidate for endothelium-derived
hyperpolarizing factor (The exact structure and identity of EDHF is currently unknown) due
to the fact that EETs are produced in the endothelium [18]. EETs can lower systemic blood
pressure in SHR-hypertensive rats suggesting possible therapeutic validity [19, 20].

Reggie Hui-Chao Lee, Carl S. Wilkins, Alexandre Couto e Silva et al.

Figure 2. Structures of arachidonic acid, and epoxyeicosatrienoic acid.

NON-ESTERIFIED FATTY ACIDS (NEFAS)


NEFAs are a major component of triglycerides also known as, lauric (C12:0), myristic
(C14:0), palmitic (C16:0), stearic (C18:0) or linolenic (C18:3) acid (Figure 3). Some NEFAs
have been reported to inhibit endothelium-dependent vasodilation at physiological
concentrations [21] indicating that NEFAs may prove crucial in disease processes such as
atherosclerosis-mediated endothelium dysfunction [22] and also associated with major risk
factors for cardiovascular diseases and metabolic syndrome. Intravenous infusion of NEFAs
inhibits vasodilation via attenuation of both endothelium-dependent and independent
(bradykinin-mediated) vasodilation, and enhanced vascular -adrenergic receptor sensitivity
[23].
Patients with high plasma concentration of NEFAs have been associated with obesityand type II diabetes (non-insulin-dependent)-mediated hypertension [24].Interestingly, the
dihydropyridine family of calcium channel blockers (e.g. nifedipine and amlodipine) and -3
fatty acid supplement (e.g. fish oil) has been reported to either reduce plasma NEFAs
concentration or inhibit NEFAs-induced endothelial dysfunction [25, 26] used to treat
obesity- and/or type II diabetes-mediated hypertension.

-3 FATTY ACIDS
Eicosapentaenoic acid (EPA, 20:5n-3), docosahexaenoic acid (DHA, 22:6n-3) (Figure 4),
and -linolenic acid (ALA, 18:3n-3, rich in plant oils) are three major -3 essential fatty
acids involved in cardiovascular function and general circulation. Administration of EPA can
1) induce endothelium-independent aortic and mesenteric vasodilation via activation of K+ATP
channels on VSMCs via EPA-derived prostanoids [27], 2) Activation of largeconductance/Ca2+-mediated K+ channels (BK) on VSMCs by EPA metabolite, 17,18-EET
[28].
In addition, clinical research studies on the administration of EPA can inhibit the onset
and progression of atherosclerosis and decrease the prevalence of myocardial infarction [29].

Fatty Acids in Vascular Health

Figure 3. Structures of lauric, myristic, or linolenic acid.

Figure 4. Structures of -3 fatty acids, acidseicosapentaenoic acid and docosahexaenoic acid.

DHA has been shown to induce endothelium-independent vasodilation. The mechanism(s)


underlying DHA-induced vasodilation are 1) activation of ATP-sensitive K+ channels in
VSMCs by prostanoids (DHA metabolite) [30], 2) inhibition of L-type Ca2+ channel and
intracellular calcium release in VSMCs [30]. Ingestion of -3 fatty acid supplements (e.g.
fish oil and corn oil) in rats reduced norepinephrine or vasopressin-mediated aortic
vasoconstriction and enhanced endothelium-dependent vasodilation via acetylcholine.
Furthermore, DHA-mediated vasodilation was prevalent in spontaneous hypertensive rat aorta
suggesting that dietary intake of DHA is beneficial to counteract hypertension [30, 31].
Similarly, ALA causes coronary arterial vasodilation via activation of VSMC Na+/K+ATPase-mediated hyperpolarization [32]. Administration of ALA can also increase CBF and
vasodilation of rodent basilar artery (via activation of TREK-1 potassium channel) [33]
indicating that ALA may also have therapeutic value used to combat stroke/ischemia by
increasing cerebral circulation.
Dietary intake of -3 fatty acid supplements has been shown to enhance endotheliumdependent brachial arterial vasodilation in hypercholesterolemic patients [34] and reduced
forearm vascular resistance to angiotensin II (a potent vasoconstrictor) in normotensive men

Reggie Hui-Chao Lee, Carl S. Wilkins, Alexandre Couto e Silva et al.

[35]. Furthermore, dietary intake of fish oil has also shown to enhance the coronary
vasomotor activity in patients with coronary artery disease [36] and reduce the number of
deaths derived from chronic cardiac failure [29]. Altogether, these data suggest that dietary
intake of -3 fatty acid supplements are beneficial to counteract against cardiovascularrelated diseases such as hyperlipidemia, hypertension, atherosclerosis, and myocardial
infarction.

LOW-DENSITY LIPOPROTEIN (LDL)


V. HIGH-DENSITY LIPOPROTEIN (HDL)
LDL is one of the major carriers of cholesterol in circulation. LDL is also known as a
bad lipoprotein due to the fact that LDL transports cholesterol and other fat molecules to
peripheral tissues (i.e. arterial walls) and plays a crucial role in the development of several
cardiovascular-related diseases such as atherosclerosis, stroke, and myocardial infarction.
Oleic acid and lysophosphatidylcholine (Figure 5) are the major constituents of LDL.
Oxidized LDL (ox-LDL) is rapidly engulfed by macrophages to induce foam cell formation
in the arterial wall [37]. Therefore, ox-LDL is thought to be one of the major contributors to
the development of atherosclerosis. Moreover, ox-LDL can enhance coronary vasospasm
(vasoconstriction) via induce endothelium-dependent vasoconstriction consequently
preventing vasodilation and increase the activity of protein kinase C isoforms and in
VSMCs of porcine coronary arteries [38-40].
HDL is another one of the major lipoproteins in circulation known as the good
lipoprotein due to the fact that HDL transports cholesterol and fat molecules from peripheral
tissues and arterial walls to the liver for excretion. Enhanced plasma HDL-cholesterol
concentration lowers the risk of cardiovascular-associated diseases [41] providing healthy
endothelial cell function (vasodilation) [42]. HDL enhances the activity of endothelial NOS to
induce femoral arterial vasodilation in vitro [43] and enhances myocardial perfusion via NOdependent mechanisms in vivo [44]. Altogether, these data suggest that HDL may reduce the
prevalence and may serve as a novel biometric for cardiovascular disease.

FATTY ACID BIOSYNTHESIS


Fatty acids are synthesized in the cytosol from acetyl-CoA derived from the Krebs cycle.
The first step of the Krebs cycle in the mitochondria matrix is acetyl-CoA, which condenses
with oxaloacetate to form citrate via citrate synthase [45, 46]. However, citrate transferred
into the cytosol is converted to acetyl-CoA and oxaloacetate with ATP (adenosine
triphosphate) and CoA by ATP-citrate lyase (ATP-CL) [47, 48]. Lipid synthesis begins with
the irreversible carboxylation of cytosolic acetyl-CoA to malonyl-CoA. During fatty acid
synthesis, (PA or SA), saturated fatty acids are incorporated with a repeated series of
reactions. PA is the primary product during fatty acid synthesis and the major precursor to
synthesize other fatty acids by an elongation enzyme system. SA is desaturated and elongated
via a series of reactions converting to various polyunsaturated fatty acid (Figure 6). The

Fatty Acids in Vascular Health

Figure 5. Structures of oleic acid lysophosphatidylcholine.

Modified from Ratledge C. 2004; Catal A, 2013; Condary R and Yao JK, 2011.
Figure 6. Synthesis of unsaturated fatty acids.

elongation reaction occurs in the mitochondria or endoplasmic reticulum [49]. Elongation is


achieved via a four-step process. Fatty acyl-CoA adds two-carbon units in each elongation
cycle [50-52]. The four sequential enzymatic reactions are 1) fatty acyl-CoA (n) is condensed
with malonyl-CoA to generate 3-ketoacyl-CoA by FA elongase (ELOVL1-7, the rate-limiting
step) [50, 51, 53], 2) reduction of 3-ketoacyl-CoA to form 3-hydroxyacyl-CoA by 3-ketoacylCoA reductase (KAR) with NADPH as a cofactor [54], 3) 3-hydroxyacyl-CoA is converted to
generate trans-2,3-enoyl-CoA using 3-hydroxyacyl-CoA dehydratase (HADC1-4) [55], 4)
reduction of trans-2,3-enoyl-CoA to produce elongated fatty acyl-CoA (n+2) by trans-2,3enoyl-CoA reductase (TER) (Figure 7) [54].
PAME and other fatty acid methyl esters (FAMEs) are endogenous compounds [2, 56],
formed via fatty acid ethyl ester synthase (FAEES) [2]. The fatty acid esterification catalyzes
an oxygen atom from the carboxyl group, then condenses the carboxyl group of an acid and
the hydroxyl group of an alcohol. FAME synthesis can also be modulated by exogenous
methanol and inhibitors of FAEES [2, 57, 58].

Reggie Hui-Chao Lee, Carl S. Wilkins, Alexandre Couto e Silva et al.

Figure 7. Typical fatty acid elongation cycle.

FATTY ACID OXIDATION


-oxidation of fatty acids is a major metabolic process in which fatty acids are degraded
in the mitochondria and peroxisome to produce energy [59, 60]. -oxidation occurs at the carbon (C-3) of the fatty acid. However, fatty acids must be activated for degradation before
being -oxidized, because negatively charged fatty acids cannot enter the plasma membrane.
Activation of fatty acids are catalyzed by fatty acyl-CoA synthetase (FACS, or called
thiokinases) to form fatty acyl-CoA thioester [61]. The net reaction of this activation process
is ATP-dependent.
Fatty acid + ATP + HS-CoA Fatty acyl-CoA + AMP + 2 Pi
Following the catalytic reaction to form long-chain fatty acyl-CoA, the enzymatic
reaction of carnitine palmitoyltranferase I (CPT-1) replaces CoA with carnitine to form fatty
acylcarnitine [62]. This conversion allows fatty acids to be transported from the cytoplasm to
the inner mitochondrial membrane via carnitine acylcarnitine translocase. Once across the
inner mitochondria membrane, fatty acylcarnitine is reversely converted back to long-chain
fatty acyl-CoA by carnitine palmitoyltrandferase II (CPT-2) for subsequent -oxidation [63,
64]. Each -oxidation cycle removes a two carbon unit and involves four main enzymes: 1)
acyl-CoA dehydrogenase, 2) enoyl-CoA hydratase, 3) 3-hydroxyacyl-CoA dehydrogenase,
and 4) -ketothiolase [65]. The net reaction of each -oxidation pathway is:
Fatty acyl-CoA + FAD + NAD+ + CoA + H2O Fatty acyl-CoA (2C less)
+ FADH2 + NADH + H+ + acetyl-CoA
The net reaction of palmitic acid (C16 fatty acid) requires seven cycles of -oxidation to
convert one molecule of palmitic acid or palmitate into eight molecules of acetyl-CoA.

Fatty Acids in Vascular Health

Palmitate + 7FAD + 7NAD+ + 8CoA + 7H2O + ATP 8acetyl-CoA +


7FADH2 + 7NADH + AMP + 2 Pi + 7H+
The large amounts of ATP are generated from FADH2, NADH, and acetyl-CoA by
subsequent citric acid cycle (Krebs cycle) and electron transfer chain. It is evident that fatty
acid oxidation is a major source of cellular ATP.

THE ROLE OF FATTY ACIDS AND ISCHEMIA/STROKE


Ischemic stroke is a condition archetypally resulting from mechanical obstruction of a
cerebral artery by an atherosclerotic plaque, or by obstruction from a thrombus causing
obstruction. Stroke results in significant morbidity, mortality, and health care costs every year
in the United States. According to the American Heart Association, there are 800,000 victims
of stroke each year, resulting in about 1 in every 19 deaths [66]. This translates into one
stroke victim every 40 seconds and one death attributable to stroke every 4 minutes [66].
Though deaths resulting from stroke fell 35.8% from the year 2000-2010 [66], the prevalence
of ischemic stroke is still quite high, so investigations into better therapeutic interventions are
important for long-term reduction of stroke and general ischemia.
Hypoperfusion after ischemia causes decreased oxygen delivery to neurons resulting in
cell death [67]. Pharmacological intervention targeting cerebral vascular regulation after
ischemia have gained significant interest as potential therapies, while already existing
modalities such as acetylsalicylic acid, heparin, direct thrombin inhibitors (DTI, dabigatran),
or inhibitor of factor Xa (rivaroxaban) are not direct targets of cerebral vasculature. However,
previous studies have suggested that fatty acids such as PAME promote vasodilation and
neuroprotection following global cerebral ischemia as well as in models of ischemic stroke
[12]. These fatty acids are endogenously produced hydrocarbon molecules containing a
carboxyl functional group, participating in many biological processes involving cellular
metabolism, signaling, and structure [68, 69].
Early interests in vasodilation therapy against ischemia involve the use of calcium
channel blockers (CCB) to induce vasodilation; however CCBs have little to no efficacy in
ischemic stroke treatment [70, 71]. Fatty acids have gained unfair notoriety as harmful agents
though they provide many important roles in the body, including emerging data that they
could be useful as therapeutic adjuncts for treatment against ischemic stroke. Increased
interest in fatty acids is due to previous animal studies suggesting that some fatty acids are
endogenously produced and released, causing vasodilation and/or increase in CBF in vivo and
in vitro [5, 12]. Though fatty acids do not exhibit any thrombolytic effects, their vasodilation
properties offer a novel therapeutic opportunity to restore blood to the ischemic penumbra,
bypassing the obstruction resulting in increased chance of survival of hypoxic tissues. Lin et
al., 2014 demonstrated that rats pretreated with PAME or SAME can attenuate neuronal cell
death in the CA1 region of the rat hippocampus by increasing CBF in order to combat
ischemia-induced hypoperfusion after asphyxial cardiac arrest (ACA, global ischemia) and
focal ischemia [12]. These results are promising since these FAMEs are endogenously
produced suggesting the possibility of low toxicity if pharmacologically administered in vivo.

10

Reggie Hui-Chao Lee, Carl S. Wilkins, Alexandre Couto e Silva et al.

Other fatty acids such as alpha-linolenic acid (ALA), have been shown to increase CBF
and vasodilation in vivo and in vitro [33]. ALA, PA, or saline was administered onto rat
carotid or basilar arteries resulting in ALA-induced vasodilation in basilar arteries. This was
also confirmed by laser-Doppler flowmetry suggesting an increase in CBF further supporting
ALA as a vasodilator [33] thought to be activated by the TREK-1 K+ channel. Additionally,
ALA has been shown to provide some degree of neuroprotection in animal models following
transient global ischemia [72].
Interestingly, Wang et al., 2006 and 2007 suggested that treatment with SA alone has
been shown to mitigate neuronal cell death due to ischemia-induced oxidative stress [73, 74].
More research with SAME, the esterified form of SA, is necessary to prove therapeutic
efficacy under ischemic conditions. Moreover, since PAME and SAME were co-released
from a neuronal source, it would be interesting to co-administer PAME/SAME and observe
the possible effects of CBF and neuroprotection [5]. Taken together, it is important to note
that the esterified from of PA produces consistent vasodilation while, PA alone does not
possess any vasodilatory properties.These results illustrate the need for further investigation
into PAME regarding its potential receptor/binding site, synthesis, and localization of stored
PAME. Assuming that further studies of PAME, SAME, and other fatty acid signaling
molecules reinforce data already available, this class of endogenous molecules should be
highly considered for interventional therapy against stroke and general ischemia.

ACKNOWLEDGMENTS
This work was supported by National Institutes of Health grant NS073779-03, American
Heart Association-Philips grant AHA-13SDG13950014 and ASA-14BFSC17690007, and
Miami Evelyn F. McKnight Brain Institute.
Disclosure/Conflict of Interest: The authors have no conflict of interest in this
manuscript.

REFERENCES
[1]

[2]
[3]

[4]

Benatar, J. R., Gladding, P., White, H. D., Zeng, I., Stewart, R. A. (2011) Trans-fatty
acids in New Zealand patients with coronary artery disease. Eur. J. Cardiovasc. Prev.
Rehabil. 18: 615-620.
Kaphalia, B. S., Carr, J. B., Ansari, G. A. (1995) Increased endobiotic fatty acid methyl
esters following exposure to methanol. Fundam. Appl. Toxicol. 28: 264-273.
Lin, H. W., Perez-Pinzon, M. (2013) The role of fatty acids in the regulation of cerebral
vascular function and neuroprotection in ischemia. CNS Neurol. Disord. Drug Targets
12: 316-324.
Cooper, G. M. (2000) Cell Membranes. The Cell: A Molecular Approach 2nd edition.
http://www.ncbi.nlm.nih.gov/books/NBK9839/.

Fatty Acids in Vascular Health


[5]

[6]

[7]

[8]

[9]

[10]
[11]

[12]

[13]

[14]

[15]

[16]
[17]

[18]

[19]

11

Lin, H. W., Liu, C. Z., Cao, D., Chen, P. Y., Chen, M. F., et al. (2008) Endogenous
methyl palmitate modulates nicotinic receptor-mediated transmission in the superior
cervical ganglion. Proc. Natl. Acad. Sci. US 105: 19526-19531.
Lee, Y. C., Chang, H. H., Liu, C. H., Chen, M. F., Chen, P. Y., et al. (2010) Methyl
palmitate: a potent vasodilator released in the retina. Invest. Ophthalmol. Vis. Sci. 51:
4746-4753.
Lee, Y. C., Chang, H. H., Chiang, C. L., Liu, C. H., Yeh, J. I., et al. (2011) Role of
perivascular adipose tissue-derived methyl palmitate in vascular tone regulation and
pathogenesis of hypertension. Circulation 124: 1160-1171.
Feelisch, M., Kotsonis, P., Siebe, J., Clement, B., Schmidt, H. H. (1999) The soluble
guanylyl cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3,-a] quinoxalin-1-one is a
nonselective heme protein inhibitor of nitric oxide synthase and other cytochrome P450 enzymes involved in nitric oxide donor bioactivation. Mol. Pharmacol. 56: 243253.
Shelkovnikov, S., Merlic, C. A., Gonick, H. C. (2004) Influence of nitric oxide donors
and peroxynitrite on the contractile effect and concentration of norepinephrine. Life Sci.
74: 2919-2928.
Harukuni, I., Bhardwaj, A. (2006) Mechanisms of brain injury after global cerebral
ischemia. Neurol. Clin. 24: 1-21.
Sabri, M., Lass, E., Macdonald, R. L. (2013) Early brain injury: a common mechanism
in subarachnoid hemorrhage and global cerebral ischemia. Stroke Res. Treat. 2013:
394036.
Lin, H. W., Saul, I., Gresia, V. L., Neumann, J. T., Dave, K. R., et al. (2014) Fatty acid
methyl esters and Solutol HS 15 confer neuroprotection after focal and global cerebral
ischemia. Transl. Stroke Res. 5: 109-117.
Miller, A. W., Katakam, P. V., Lee, H. C., Tulbert, C. D., Busija, D. W., et al. (2003)
Arachidonic acid-induced vasodilation of rat small mesenteric arteries is lipoxygenasedependent. J. Pharmacol. Exp. Ther. 304: 139-144.
Ellis, E. F., Police, R. J., Yancey, L., McKinney, J. S., Amruthesh, S. C. (1990) Dilation
of cerebral arterioles by cytochrome P-450 metabolites of arachidonic acid. Am. J.
Physiol. 259: H1171-1177.
Campbell, W. B., Harder, D. R. (1999) Endothelium-derived hyperpolarizing factors
and vascular cytochrome P450 metabolites of arachidonic acid in the regulation of tone.
Circ. Res. 84: 484-488.
Fisslthaler, B., Popp, R., Kiss, L., Potente, M., Harder, D. R., et al. (1999) Cytochrome
P450 2C is an EDHF synthase in coronary arteries. Nature 401: 493-497.
Ye, D., Zhou, W., Lee, H. C. (2005) Activation of rat mesenteric arterial KATP
channels by 11,12-epoxyeicosatrienoic acid. Am. J. Physiol. Heart Circ. Physiol. 288:
H358-364.
Campbell, W. B., Gebremedhin, D., Pratt, P. F., Harder, D. R. (1996) Identification of
epoxyeicosatrienoic acids as endothelium-derived hyperpolarizing factors. Circ. Res.
78: 415-423.
Hye Khan, M., Pavlov, T. S., Christain, S. V., Neckar, J., Staruschenko, A., et al.
(2014) Epoxyeicosatrienoic Acid (EET) Analog Lowers Blood Pressure Through
Vasodilation And Sodium Channel Inhibition. Clin. Sci. (Lond.).

12

Reggie Hui-Chao Lee, Carl S. Wilkins, Alexandre Couto e Silva et al.

[20] Imig, J. D., Elmarakby, A., Nithipatikom, K., Wei, S., Capdevila, J. H., et al. (2010)
Development of epoxyeicosatrienoic acid analogs with in vivo anti-hypertensive
actions. Front. Physiol. 1: 157.
[21] Edirisinghe, I., McCormick Hallam, K., Kappagoda, C. T. (2006) Effect of fatty acids
on endothelium-dependent relaxation in the rabbit aorta. Clin. Sci. (Lond.) 111: 145151.
[22] Carlsson, M., Wessman, Y., Almgren, P., Groop, L. (2000) High levels of nonesterified
fatty acids are associated with increased familial risk of cardiovascular disease.
Arterioscler. Thromb. Vasc. Biol. 20: 1588-1594.
[23] De Kreutzenberg, S. V., Puato, M., Kiwanuka, E., Del Prato, S., Pauletto, P., et al.
(2003) Elevated non-esterified fatty acids impair nitric oxide independent vasodilation,
in humans: evidence for a role of inwardly rectifying potassium channels.
Atherosclerosis 169: 147-153.
[24] Egan, B. M., Greene, E. L., Goodfriend, T. L. (2001) Nonesterified fatty acids in blood
pressure control and cardiovascular complications. Curr. Hypertens. Rep. 3: 107-116.
[25] Yasu, T., Kobayashi, M., Mutoh, A., Yamakawa, K., Momomura, S., et al. (2013)
Dihydropyridine calcium channel blockers inhibit non-esterified-fatty-acid-induced
endothelial and rheological dysfunction. Clin. Sci. (Lond.) 125: 247-255.
[26] Shearer, G. C., Savinova, O. V., Harris, W. S. (2012) Fish oil -- how does it reduce
plasma triglycerides? Biochim. Biophys. Acta 1821: 843-851.
[27] Engler, M. B., Engler, M. M., Browne, A., Sun, Y. P., Sievers, R. (2000) Mechanisms
of vasorelaxation induced by eicosapentaenoic acid (20:5n-3) in WKY rat aorta. Br. J.
Pharmacol. 131: 1793-1799.
[28] Hercule, H. C., Salanova, B., Essin, K., Honeck, H., Falck, J. R., et al. (2007) The
vasodilator 17,18-epoxyeicosatetraenoic acid targets the pore-forming BK alpha
channel subunit in rodents. Exp. Physiol. 92: 1067-1076.
[29] Ohnishi, H., Saito, Y. (2013) Eicosapentaenoic acid (EPA) reduces cardiovascular
events: relationship with the EPA/arachidonic acid ratio. J. Atheroscler. Thromb. 20:
861-877.
[30] Engler, M. B., Engler, M. M. (2000) Docosahexaenoic acid--induced vasorelaxation in
hypertensive rats: mechanisms of action. Biol. Res. Nurs. 2: 85-95.
[31] Malis, C. D., Leaf, A., Varadarajan, G. S., Newell, J. B., Weber, P. C., et al. (1991)
Effects of dietary omega 3 fatty acids on vascular contractility in preanoxic and
postanoxic aortic rings. Circulation 84: 1393-1401.
[32] Pomposiello, S. I., Alva, M., Wilde, D. W., Carretero, O. A. (1998) Linoleic acid
induces relaxation and hyperpolarization of the pig coronary artery. Hypertension 31:
615-620.
[33] Blondeau, N., Petrault, O., Manta, S., Giordanengo, V., Gounon, P., et al. (2007)
Polyunsaturated fatty acids are cerebral vasodilators via the TREK-1 potassium
channel. Circ. Res. 101: 176-184.
[34] Goodfellow, J., Bellamy, M. F., Ramsey, M. W., Jones, C. J., Lewis, M. J. (2000)
Dietary supplementation with marine omega-3 fatty acids improve systemic large artery
endothelial function in subjects with hypercholesterolemia. J. Am. Coll. Cardiol. 35:
265-270.

Fatty Acids in Vascular Health

13

[35] Kenny, D., Warltier, D. C., Pleuss, J. A., Hoffmann, R. G., Goodfriend, T. L., et al.
(1992) Effect of omega-3 fatty acids on the vascular response to angiotensin in
normotensive men. Am. J. Cardiol. 70: 1347-1352.
[36] Yamamoto, H., Yoshimura, H., Noma, M., Suzuki, S., Kai, H., et al. (1995)
Improvement of coronary vasomotion with eicosapentaenoic acid does not inhibit
acetylcholine-induced coronary vasospasm in patients with variant angina. Jpn. Circ. J.
59: 608-616.
[37] Parthasarathy, S., Printz, D. J., Boyd, D., Joy, L., Steinberg, D. (1986) Macrophage
oxidation of low density lipoprotein generates a modified form recognized by the
scavenger receptor. Arteriosclerosis 6: 505-510.
[38] Murohara, T., Kugiyama, K., Ohgushi, M., Sugiyama, S., Ohta, Y., et al. (1994) LPC in
oxidized LDL elicits vasocontraction and inhibits endothelium- dependent relaxation.
Am. J. Physiol. 267: H2441-2449.
[39] Giardina, J. B., Tanner, D. J., Khalil, R. A. (2001) Oxidized-LDL enhances coronary
vasoconstriction by increasing the activity of protein kinase C isoforms alpha and
epsilon. Hypertension 37: 561-568.
[40] Niu, X. L., Liu, L. Y., Hu, M. L., Chen, X. (1995) Some similarities in vascular effects
of oleic acid and oxidized low-density lipoproteins on rabbit aorta. J. Mol. Cell.
Cardiol. 27: 531-539.
[41] Emerging Risk Factors C, Di Angelantonio, E., Sarwar, N., Perry, P., Kaptoge, S., et al.
(2009) Major lipids, apolipoproteins, and risk of vascular disease. JAMA 302: 19932000.
[42] Steer, P., Hulthe, J., Miligard, J., Sarabi, D. M., Basu, S., et al. (2002) Endothelial
vasodilatory function is predicted by circulating apolipoprotein B and HDL in healthy
humans. Lipids 37: 1135-1140.
[43] Gong, M., Wilson, M., Kelly, T., Su, W., Dressman, J., et al. (2003) HDL-associated
estradiol stimulates endothelial NO synthase and vasodilation in an SR-BI-dependent
manner. J. Clin. Invest. 111: 1579-1587.
[44] Levkau, B., Hermann, S., Theilmeier, G., van der Giet, M., Chun, J., et al. (2004) Highdensity lipoprotein stimulates myocardial perfusion in vivo. Circulation 110: 33553359.
[45] Krebs, H. A. (1940) The citric acid cycle and the Szent-Gyorgyi cycle in pigeon breast
muscle. Biochem. J. 34: 775-779.
[46] Chinopoulos, C. (2013) Which way does the citric acid cycle turn during hypoxia? The
critical role of alpha-ketoglutarate dehydrogenase complex. J. Neurosci. Res. 91: 10301043.
[47] Chypre, M., Zaidi, N., Smans, K. (2012) ATP-citrate lyase: a mini-review. Biochem.
Biophys. Res. Commun. 422: 1-4.
[48] Zaidi, N., Swinnen, J. V., Smans, K. (2012) ATP-citrate lyase: a key player in cancer
metabolism. Cancer Res. 72: 3709-3714.
[49] Ratledge, C. (2004) Fatty acid biosynthesis in microorganisms being used for Single
Cell Oil production. Biochimie 86: 807-815.
[50] Guillou, H., Zadravec, D., Martin, P. G., Jacobsson, A. (2010) The key roles of
elongases and desaturases in mammalian fatty acid metabolism: Insights from
transgenic mice. Prog. Lipid Res. 49: 186-199.

14

Reggie Hui-Chao Lee, Carl S. Wilkins, Alexandre Couto e Silva et al.

[51] Jakobsson, A., Westerberg, R., Jacobsson, A. (2006) Fatty acid elongases in mammals:
their regulation and roles in metabolism. Prog. Lipid Res. 45: 237-249.
[52] Kihara, A. (2012) Very long-chain fatty acids: elongation, physiology and related
disorders. J. Biochem. 152: 387-395.
[53] Ohno, Y., Suto, S., Yamanaka, M., Mizutani, Y., Mitsutake, S., et al. (2010) ELOVL1
production of C24 acyl-CoAs is linked to C24 sphingolipid synthesis. Proc. Natl. Acad.
Sci. US 107: 18439-18444.
[54] Moon, Y. A., Horton, J. D. (2003) Identification of two mammalian reductases involved
in the two-carbon fatty acyl elongation cascade. J. Biol. Chem. 278: 7335-7343.
[55] Ikeda, M., Kanao, Y., Yamanaka, M., Sakuraba, H., Mizutani, Y., et al. (2008)
Characterization of four mammalian 3-hydroxyacyl-CoA dehydratases involved in very
long-chain fatty acid synthesis. FEBS Lett. 582: 2435-2440.
[56] Lough, A. K., Garton, G. A. (1968) The lipids of human pancreas with special reference
to the presence of fatty acid methyl esters. Lipids 3: 321-323.
[57] Mericle, K. A., Kaphalia, B. S., Ansari, G. A. (2004) Modulation of fatty acid methyl
esters in rats pretreated with tri-o-tolyl phosphate. J. Toxicol. Environ. Health A 67:
583-593.
[58] Kaphalia, B. S., Green, S. M., Ansari, G. A. (1999) Fatty acid ethyl and methyl ester
synthases, and fatty acid anilide synthase in HepG2 and AR42J cells: interrelationships
and inhibition by tri-o-tolyl phosphate. Toxicol. Appl. Pharmacol. 159: 134-141.
[59] Eaton, S., Bartlett, K., Pourfarzam, M. (1996) Mammalian mitochondrial betaoxidation. Biochem. J. 320 (Pt 2): 345-357.
[60] Poirier, Y., Antonenkov, V. D., Glumoff, T., Hiltunen, J. K. (2006) Peroxisomal betaoxidation--a metabolic pathway with multiple functions. Biochim. Biophys. Acta 1763:
1413-1426.
[61] Watkins, P. A., Ellis, J. M. (2012) Peroxisomal acyl-CoA synthetases. Biochim.
Biophys. Acta 1822: 1411-1420.
[62] Stephens, F. B., Constantin-Teodosiu, D., Greenhaff, P. L. (2007) New insights
concerning the role of carnitine in the regulation of fuel metabolism in skeletal muscle.
J. Physiol. 581: 431-444.
[63] Jeppesen, J., Kiens, B. (2012) Regulation and limitations to fatty acid oxidation during
exercise. J. Physiol. 590: 1059-1068.
[64] Rufer, A. C., Thoma, R., Hennig, M. (2009) Structural insight into function and
regulation of carnitine palmitoyltransferase. Cell. Mol. Life Sci. 66: 2489-2501.
[65] Hiltunen, J. K., Qin, Y. (2000) beta-oxidation - strategies for the metabolism of a wide
variety of acyl-CoA esters. Biochim. Biophys. Acta 1484: 117-128.
[66] Go, A. S., Mozaffarian, D., Roger, V. L., Benjamin, E. J., Berry, J. D., et al. (2014)
Executive summary: heart disease and stroke statistics--2014 update: a report from the
American Heart Association. Circulation 129: 399-410.
[67] Niquet, J., Baldwin, R. A., Allen, S. G., Fujikawa, D. G., Wasterlain, C. G. (2003)
Hypoxic neuronal necrosis: protein synthesis-independent activation of a cell death
program. Proc. Natl. Acad. Sci. US 100: 2825-2830.
[68] Comba, A., Lin, Y. H., Eynard, A. R., Valentich, M. A., Fernandez-Zapico, M. E., et al.
(2011) Basic aspects of tumor cell fatty acid-regulated signaling and transcription
factors. Cancer Metastasis Rev. 30: 325-342.

Fatty Acids in Vascular Health

15

[69] Vrablik, T. L., Watts, J. L. (2012) Emerging roles for specific fatty acids in
developmental processes. Genes Dev. 26: 631-637.
[70] Horn, J., Limburg, M. (2001) Calcium antagonists for ischemic stroke: a systematic
review. Stroke 32: 570-576.
[71] Zhang, J., Yang, J., Zhang, C., Jiang, X., Zhou, H., et al. (2012) Calcium antagonists for
acute ischemic stroke. Cochrane Database Syst. Rev. 5: CD001928.
[72] Lauritzen, I., Blondeau, N., Heurteaux, C., Widmann, C., Romey, G., et al. (2000)
Polyunsaturated fatty acids are potent neuroprotectors. EMBO J. 19: 1784-1793.
[73] Wang, Z. J., Li, G. M., Nie, B. M., Lu, Y., Yin, M. (2006) Neuroprotective effect of the
stearic acid against oxidative stress via phosphatidylinositol 3-kinase pathway. Chem.
Biol. Interact. 160: 80-87.
[74] Wang, Z. J., Liang, C. L., Li, G. M., Yu, C. Y., Yin, M. (2007) Stearic acid protects
primary cultured cortical neurons against oxidative stress. Acta Pharmacol. Sin. 28:
315-326.

In: Palmitic Acid: Occurrence, Biochemistry and Health Effects ISBN: 978-1-63321-519-1
Editor: Lucas F. Porto
2014 Nova Science Publishers, Inc.

Chapter 2

OCCURRENCE, BIOCHEMICAL, ANTIMICROBIAL


AND HEALTH EFFECTS OF PALMITIC ACID
Melissa Johnson*1 and Daniel A. Abugri2
1

Department of Agricultural and Environmental Sciences,


College of Agriculture, Environment and Nutrition Sciences,
Tuskegee University, Tuskegee AL, US
2
Department of Chemistry, College of Arts and Sciences,
Tuskegee University, Tuskegee, AL, US

ABSTRACT
Palmitic acid (PA), one of the most abundant saturated fatty acid (SFAs) within
plants, humans, animals, microbial (bacteria), fungal, and marine organisms, constitutes
~16 to 45 % of the lipid profile. The impressive abundance of PA throughout nature
could be attributed partly to its critical role in membrane lipid structural functionality,
formation of subcellular cysteine residue linkages and RNA posttranslational
modifications in eukaryotic and prokaryotic cells. PA has been demonstrated to undergo
-oxidation to produce short and medium chain fatty acids to maintain homeostasis in
response to endogenous and exogenous cues. Further, non-esterified PA is known to
mediate numerous biochemical and antimicrobial pathways towards the betterment of
human health. Although the importance of PA in human health and nutrition are
established, critics attest that excessive dietary PA may be unhealthy and even
detrimental. Current microbiological and epidemiological studies suggest that PA
produces specific health benefits that are not common to all other SFAs. For example,
studies suggest that PA may activate nitric oxide and superoxide, thus functioning as an
antimicrobial agent against some strains of bacteria, algae, and helminthes and certain
foodborne pathogens. This chapter reviews the occurrence, biochemistry, and subsequent
health implications of PA.

Keywords: Palmitic acid, saturated fatty acid, biochemistry, health implications

mjonson@mytu.tuskegee.edu;
dabugri@mytu.tuskegee.edu; abugrigh@yahoo.com.

18

Melissa Johnson and Daniel A. Abugri

1.1. INTRODUCTION
The most common and abundant saturated fatty acid (SFA) in nature, as well as within
plants, animals and humans, palmitic acid (PA) has many critical functions (Figure 1).
Serving as the primary storage form of excess dietary carbohydrates and the precursor for
longer chain fatty acids, palmitic acid occupies a critical role in long-term energy storage, the
synthesis of other biomolecules (e.g. glycolipids, phospholipids, vitamins, prostaglandins,
prostacyclins, thromboxanes, etc.) and cellular membrane structural constituents. Palmitic
acid has also been evaluated based on its antimicrobial, antifungal, antibacterial contributions,
when bound to specific proteins. Applications within the health and beauty (e.g. cosmetics,
soap production), food (e.g. additive, texturing agent), pharmaceutical (e.g. palmate ester as a
carrier medium and releasing agent) and biotechnology industries further add credence to the
virtual diversity of this fatty acid to function as a multipurpose entity and challenges one to
consider this fatty acid beyond the conventional understanding as a fatty acid whose primarily
purpose for existence is to serve as a fuel source.
Palmitic acid and its bioactive metabolites and successors exhibit unique absorption,
transport, metabolic, site-specific distribution(s), mechanisms of action and implications
(Figure 1). These characteristics contribute to localized and generalized observed and
subsequent potential repercussions- both acute and chronic. The ability of palmitic acid to
influence protein-protein interactions, hydrophobicity, membrane association, and subcellular
trafficking within and between membrane constituents, exemplifies the capability of palmitic
acid to purposefully engage in covalent binding to specific protein residues. The occurrence,
biochemistry and health effects of palmitic acid within the micro and macro bio-arenas of
plants, animals, humans and other microorganisms accentuate the transformative nature of
this fatty acid and its influence on the lipidome, metabolome, proteome, transcriptome and
genome. This chapter seeks to integratively explore the multi-faceted nature of palmitic acid
and provide the reader with an interactive learning experience, with appropriate visual aids to
abridge concepts and aid in comprehension, which allows for the enhanced appreciation of
one of the most abundant fatty acids in nature.

1.2. DEFINITION, IMPORTANCE AND POTENTIAL OF PALMITIC ACID


1.2.1. Standard Definition of Palmitic Acid
Palmitic acid (PA) or hexadecanoic acid is one of the most plentiful fatty acids in nature
and tissues, is characterized by a 16 Carbon atom chain with the absence of a C-C double
bond (C16:0). It encompasses both hydrophobic and hydrophilic regions, containing carboxyl
acid (-COOH) and a methyl group end (-CH3) functional groups respectively. This unique
feature allows palmitic acid to exist both as a free fatty acid and in complex with other lipids
(e.g. diglyceride, triglycerides), carbohydrate (e.g. glycolipid), and protein (e.g. lipoprotein)
entities.

*Excess carbohydrates (CHOs) are converted to palmitic acid; negative feedback (inhibition) by acetyl-CoA carboxylase (Benoit et al. 2009) the enzyme
catalyzing the conversion of acetyl-CoA to malonyl-CoA; positive feedback (synthesis) catalyzing the synthesis of palmitic acid; CVD- cardiovascular
disease.
Figure 1. Summary of the occurrence, biochemical courses of action and implications of palmitic acid.

20

Melissa Johnson and Daniel A. Abugri

1.2.2. Importance of Palmitic Acid


Palmitic acid is used as the precursor of long chain fatty acids lipogenesis and can also
undergo beta oxidation to produce short and medium chain fatty acids. For instance, palmitic
acid is first used to synthesized palmitoleic acid under cellular condition using the de novo
pathways which is further use to make C18:0 (stearic acid). From the C18:0 fatty acid, a
C18:1n9 and C18:2n6 and C18:3n3 can also be synthesized from the 18 carbon saturated fatty
acid (C18:0). Palmitic acid serves as a precursor for longer chain fatty acid synthesis e.g.
Triacylglycerol (Figure 5). From these C18 carbons the omega 6 and omega 3 from C18 and
high chain fatty acids can then be made for cellular utilization. The C16:0 fatty acid presences
in the cell play a major role in posttranslational modification of mRNA (Cooper, 2000). Here,
the acid is known to bind with cysteine residues which render the cysteine residue favorable
for any modification of the protein to be expressed for cellular use (Corvi et al., 2000).
Basically, palmitic acid under cellular condition binds to sulfur atoms from the side chains of
the internal amino acid called cysteine residues during posttranslational modification. In
addition, palmitoylation, palmitic acid has the ability to contribute greatly to lipid rafts
associated with cytosolic and plasmic cell signaling mechanisms (Dykstra et al., 2003). This
is very important in understanding the pathogenesis of diseases and their causative agents
(Corvi et al., 2012). This type of phenomena is unique to palmitic acid due to thioester bond
formation between the cysteine amino acid. This in turn, permits soluble proteins to interact
with cellular membranes, mediate and initiate localization at the surface of mammalian cell
membranes (Resh, 2006). It has been proposed that the ability of palmitic acid to dictates
protein function is due to its reversibility property on like other lipid modification that cannot
be reversible (Blanc, Blaskovic, & Goot, 2013). It is important to point out that most protein
hydrophobicity among most fatty acids modification (e.g. myristic) is not as efficient as the
one formed by palmitic acid. When palmitic acid is involved, it increases membrane proteins
interaction in the cell. Also aside the, palmitoylation, they are good sources for generation of
ATP during exogenous and endogenous activities in the cell. Palmitic acid ability to under
modification creates several opportunities for soluble and integral membrane proteins such as
signaling proteins, enzymes, scaffolding proteins, ion channels, cell adhesion molecules and
neurotransmitter receptors (Blanc, Blaskovic and Goot, 2013).

1.2.3. Potentials of Palmitic Acid


In addition to exhibiting vast importance to biological systems, palmitic acid, palmitic
acid is of importance to the food, cosmetics, pharmaceutical, biotechnology and biodiesel
industries (Carmo Jr et al., 2009; Houde, Kademi, & Leblanc, 2004; Jaeger & Reetz, 1998;
Keng et al., 2009; Metzger & Bornscheuer, 2006; Pitto et al., 2002; Quintavalla & Vicini,
2002; Schmelzle et al., 2003). As outlined, in the figure 2 below palmitic acid have many
functions, extending beyond the obvious cellular functions of serving as a precursor for
longer chain fatty acid synthesis and as an energy source during -oxidation.

Occurrence, Biochemical, Antimicrobial and Health Effects of Palmitic Acid

21

Figure 2. Functions and contributions of palmitic acid. (Image Sources: en.wikipedia.org; Microsoft office
clipart).

1.3. OCCURRENCE OF PALMITIC ACID


1.3.1. Distribution in Nature (Plant Sources, Algae, Fungus, Etc.)
In plants, algae, fungus, yeast and bacteria have also shown similar predominant
saturated fatty acid to be palmitic. Palmitic acid distribution varies both within species and
among species (Table 1). In recent reports PA content can be influence by the environment in
which such plant, fungus, bacteria and yeast are found (Griffiths et al. 2003). Most of the
factors contributing to this variation are soil pH, nutrient-ion interaction, age, water and host
interaction. For instance variation has been observed in mushrooms, yeast, algae, viruses and
other plants PA content (Table 1). It is important that in considering chemotaxonomy these
above factors need to be critical considered to avoid biasness in data.

1.3.2. Distribution in Human Tissue


Palmitic acid was found to be the second most abundant fatty acid in abdominal adipose
tissue following oleic acid (Garaulet et al., 2001). With increasing concentrations of palmitic
acid, increases in storage within visceral adipocytes have been observed; palmitic acid was
able to significantly increase linoleic acid accumulation in visceral fat (adipocytes) and to a
lesser degree in subcutaneous adipocytes (Sabin et al., 2007). Further, in free fatty acid form,

22

Melissa Johnson and Daniel A. Abugri

lipid levels increased in both visceral and subcutaneous adipocytes, in comparison to primary
accumulation in visceral adipocytes when in triglyceride form.

1.3.3. Distribution in Animal Tissue


PA is the most abundant saturated fatty acid found in most animal products, for example
meat. Palmitic acid has been reported to account for approximately 27% of the saturated fatty
acid content of beef (Whetsell et al., 2003). PA occurrences in animals tissues are likely to
be influence by environmental and nutritional factors. This unique feature makes PA an
essential chemotaxonomic tool for identification of species of animals and poultry and even
plant related species.
Table 1. Approximate palmitic acid distribution in selected human, animal, plant,
fungal, viral and bacterial sources
Source
Human
Milk
Sebum (from back)
Hair
Animal
Adipose tissue
Plasma (mice)
Brain (mice)
Brain (rat)

Liver (SHR)
Liver (mice)
Fungal
Agaricus bisporus
Cortinarius glaucopus
Hygrophoropsis
aurantiaca
Hypholoma capnoides
Laccaria laccata
Lactarius salmonicolor
Lespista inversa
Turkey tail

% PA

References

20-25%
17.6-30.1%
36.0%

(Innis, Dyer, & Nelson, 1994)


(Boughton & Wheatley, 1959)
(Weitkamp, Smiljanic, & Rothman,
1947)

23.4-23.8%
19.5-19.6%
19.8-21.5%
69% (Phosphoglycerides)
3% (Monoacylglycerides)
8% (Cholesterol)
4% (Free fatty acids)
6% (1,2-Diacylglyceride)
3% (1,3-Diacylglyceride)
2%(Cholesterol ester)
6% (Triglyceride)
16-21%
21.0-21.5%

(Rule, 1997)
(Shirai, Suzuki, & Wada, 2005)
(Shirai, Suzuki, & Wada, 2005)

13-14%

(Abugri, McElhenney, & Willian,


2012)
(Heleno et al., 2009)
(Heleno et al., 2009)

12%
10%
16%
12%
7.4%
16.4%
23%

(Johnson et al., 2013)


(Shirai, Suzuki, & Wada, 2005)

(Heleno et al., 2009)


(Heleno et al., 2009)
(Heleno et al., 2009)
(Heleno et al., 2009)
(Abugri, McElhenney, & Willian,
2012)

Occurrence, Biochemical, Antimicrobial and Health Effects of Palmitic Acid


Source
Tinder polypore

% PA
14%-15.1%

Artis Conk

17.3% -17.4%

Algae
Phaeodactylum
tricornutum
Thalassiosira weissflogii

14.7-26.8%
28.8-36.6%

Dunaliella primolecta

21.8-26.0%

Nannochloris sp.

15.1-17.8%

Parietochloris incisa

10.0-19.8%

Nostoc commune

25.3-43.5%

Synechocystis sp.

18.8-26.5%

Pavlova lutheri
Emiliana huxleyi

11.1-23.6%
10.3-17.7%

Heterosigma akashiwo

40.0-46.3%

Yeast
Nadsonia fulvescens and
N. commutata
R. bisporidii and R.
dibovatum, R.Toruloides
Bacteria
Lactobacillus arabinosus

References
(Abugri, McElhenney, & Willian,
2012)
(Abugri, McElhenney, & Willian,
2012)
(Lang et al., 2011; Tonon et al., 2002;
Viso & Marty, 1993)
(Lang et al., 2011; Viso & Marty,
1993)
(Lang et al., 2011; Viso & Marty,
1993)
(Lang et al., 2011; Viso & Marty,
1993)
(Bigogno et al., 2002; Lang et al.,
2011)
(Lang et al., 2011; Temina et al.,
2007)
(Lang et al., 2011; Viso & Marty,
1993)
(Lang et al., 2011; Tonon et al., 2002)
(Lang et al., 2011; Viso & Marty,
1993)
(Lang et al., 2011; Viso & Marty,
1993)

14-15%

Botha and Kock,1993

14-19.0%

Westhuizen et al., 1987

18.7%

(Hofmann et al., 1955; Hofmann,


Lucas, & Sax, 1952)
(Hofmann et al., 1955; Hofmann &
Sax, 1953)
(Hofmann, Henis, & Panos, 1957)

Lactobacillus casei

24.3%

Lactobacillus
delbrueckii
Streptococcus sp

27.3%

Clostridium butyricum
Escherichia coli
Argrobacterium
tumefaciens
Plant
Malva sylvestris
(leaves,flowers,immature
fruits, leafy flowered
stems)
Sweet potatoes (Ipomoea
batatasi) leaves
Capsicum chinnese

49.0%
25-38.6%
8.2%

26.6%

(Hofmann et al., 1955; Hofmann &


Tausig, 1955a; Hofmann & Tausig,
1955b)
(Goldfine & Bloch, 1961)
(Shaw & Ingraham, 1965)
O Leary, 1959

10% (leaves)
17.2% (flowers)
20%(immature fruits)
13% (leafy flowers stems)
10-21%%
16-21%

(Barros, Carvalho, & Ferreira, 2010)

(Almazan & Adeyeye, 1998;


Karmakar, Muslim, & Rahman)
Abugri et al., 2014 unpublished data

23

24

Melissa Johnson and Daniel A. Abugri


Table 1. (Continued)
Source
Sorghum bicolor red
leaves
Rice seeds (Hassawi and
CV. Hassa No.2)

% PA
16.2%

References
(Abugri et al., 2013)

16 %-29.20%

(Abdulaziz & Ai-Bahrany, 2002)

1.4. PHYSICAL, CHEMICAL, STUCTURAL AND FUNCTIONAL


PROPERTIES OF PALMITIC ACID
PA, written as, CH3CH2CH2CH2CH2CH2CH2CH2CH2CH2CH2CH2CH2CH2CH2COOH,
is solid at room temperature and has a melting point of approximately 63 oC, boiling point of
about 351 oC. The fatty acid has a density of about 853 kg/m3. Palmitic acid, the most
common fatty acid within plants, humans, animals and microorganisms, is a 16 carbon
unsaturated fatty acid (Figures 3a, 3b) that appears as white crystalline scales or colorless
needles. Palmitic acid has a molecular mass and melting point of 256.42 g mol-1 (O'Neil,
2013). Furthermore, palmitic acid has a low fluidity as compared to unsaturated fatty acid and
other long chain saturated fatty acids, but has high fluidity than other saturated fatty acid
ranged from C3.0 to C15:0. The bonds are mostly carbon-carbon and carbon-hydrogen single
bonds. There is also carbon-oxygen double bond. It is important to point out that PA has
negative feed backs inhibition on acetyl-CoA carboxylase (Benoit et al. 2009) enzyme which
is responsible for converting acetyl-ACP to malonyl-ACP on the growing acyl chain, thus
resulting in the further generation of palmitate. The physical and chemical properties of
palmitic acid facilitate the biochemical actions of palmitic acid (Figure 4)

Source: http://en.wikipedia.org/wiki/File:Palmitic-acid-3D-balls.png.
Figure 3a. Ball-and-stick 3D structure of palmitic acid molecule.

Source: http://en.wikipedia.org/wiki/File:Palmitic_acid.svg.
Figure 3b. Linear structure of palmitic acid molecule.

Occurrence, Biochemical, Antimicrobial and Health Effects of Palmitic Acid

25

Source: http://ercfre86.wordpress.com/2012/03/19/applications-of-palmitic-acid/
Figure 3c. Physical appearance of palmitic acid.

Figure 4. Summary of biochemical actions of palmitic acid.

1.4.3. Antimicrobial, Antibacterial and Antifungal Properties and


Mechanisms Action of Palmitic acid
Antimicrobial Agents
Palmitic acid inhibits and kills bacteria, virus and other pathogens (i.e. fungus) directly or
via indirect mechanism (Yff et al., 2002; McGraw et al. 2002; Seidal and Taylor, 2004; Fluhr
et al., 2005; Takigawa et al., 2005; Orhan et al. 2011). If PA is in its free form it makes
condition not favorable for growth of some type of fungus and bacteria on the surface of the
skin because of its acidity property. This conjecture is supported by (Fluhr et al., 2001;
Takigawa et al., 2005). Another route use by fatty acids to inhibit pathogens is their ability for
interaction with cell membrane. For instance, in fungus, the cell membrane is expected to
maintain the cell organelles in order as well as the cell integrity (Pohl, Kock, & Thibane,
2011). Furthermore, fatty acids can either interacts with the membrane directly resulting in
the insertion into the lipid bio-layers of the fungal and any pathogens membranes which cause
a physical disturbance of the membrane integrity (Pohl et al. 2011). This consequently,
creates high fluidity of the membrane. The higher the fluidity within the cell, the greater the

26

Melissa Johnson and Daniel A. Abugri

cell will experience disruption in the cell membranes which could lead to greater
conformational changes within membrane proteins and their functionality (Liu et al. 2008;
Avis & Belanger, 2001; Altieri et al., 2007; Pohl et al. 2011). Other effects meditated by PA
at high fluidity could be the release of intracellular components, cytoplasmic disorder and
eventually cell disintegration and apoptosis (Liu et al. 2008; Avis & Belanger, 2001; Altieri et
al., 2007; Pohl et al. 2011).
Fatty acids are known in nature to function as anionic surface agents and these roles
make them nonfunctional at certain physiological pH conditions (Armstrong, 1957; Kabara et
al., 1972; Scharff & Beck, 1959). Palmitic acid acts as antimicrobial agents against bacteria
(Kabara et al., 1972), fungal and other pathogens. For instance, palmitic acid has been
demonstrated to greatly inhibit Alernaria solani (Liu et al., 2008), Aspergillus niger (Altieri
et al., 2007), Aspergillus terreus (Altieri et al., 2007), Cucumerinum lagenarium (Liu et al.,
2008), Emericella nidulans (Altieri et al., 2007) and Fusarium oxysporum (Liu et al., 2008).
With this knowledge about palmitic acid, its biochemical pathways of inhibition of the
enzymatic secretory pathways used by such microorganisms are speculated to be either
disruption of mitochondrial machinery resulting in much electron transport influx or
disruption of the cell membrane integrity (Kabara et al., 1972; Desbois & Smith, 2010; Pohl
et al., 2011).

Selected Possible Mechanisms of Action of Palmitic Acid


Generally, fatty acid either in a free form or in an esterified form has antimicrobial
properties. These properties are depended on several factors such as the temperature, ions of
fatty acids, pH, functional groups (COOCH3, COOH, and NCOCH2), carbon chain length and
the degree of unsaturation (Kabara et al., 1972; Sikkema, De Bont, & Poolman, 1995).
Furthermore, lipid classes (phospholipids and sphingolipids) also have influence on the
degree of inhibitory, biostatic, killing, DNA and protein synthesis disruption, cell membrane
permeability, influx of ion concentration, electron transfer and mitochondria energy
expenditure production ( Desbois and Smith, 2010; Liu et al. 2008; Avis & Belanger, 2001;
Altieri et al., 2007; Pohl et al., 2011 ). Willett and Morse (Willett & Morse, 1966) have
demonstrated the unique potency of PA against certain group of bacteria.
The target organelle of palmitic acid is possibly the cell membrane of the bacteria, fungi,
virus or any parasite (Pohl et al., 2011; Liu et al. 2008; Avis & Belanger, 2001; Altieri et al.,
2007). When the cell membrane is disrupted as a result of the detergent permeability
properties of palmitic acid, it might result in disruption of the intracellular organelles
functionality. The acid might be able to perform this role due to its amphipathic nature. The
good thing is that the amphipathic property of palmitic acid will permit it to interact with the
cell wall and cell membranes resulting in the creation of small pores throughout the cell
membranes. This consequently affects the cell delimiting ability to prevent the introduction of
foreign materials into the inner contents or organelles of the microorganism. Hence, no
regularly movements of substances into and out of the cell will exist, resulting in osmotic
shock in which the parasite is kill internally. The different processes that palmitic acid might
be using to achieve this antimicrobial activity include cell lysis, inhibition of enzymes
activity, impartment of nutrient uptake and release, release of toxic peroxidation and
autoxidation products (Desbois & Smith, 2010; Pohl et al.2011).

Occurrence, Biochemical, Antimicrobial and Health Effects of Palmitic Acid

27

A. Cell Lysis
Fluidity of fatty acid depends on the chain length and the degree of unsaturation,
therefore because of these unique property, fatty acids have the ability to insert themselves
into the inner membranes of pathogens resulting in more fluidity and permeability
(Chamberlain et al., 1991; Greenway & Dyke, 1979). PA mechanism of cell lysising may be
due to high permeability of membrane due to the insertion of PA, and has inner consequence
such as allowing of internal contents to leak from cells which can result in growth inhibition
and death (Galbraith & Miller, 1973; Shin et al., 2007; Wang & Johnson, 1992; Boyaval et al.
1995; Liu et al. 2008; Avis & Belanger, 2001; Altieri et al., 2007). The higher the fluidity in
the cell the greater the cell tendency to lysising or bursting because of the issue of unbalance
membrane fluidity of the cell (Desbois & Smith, 2010).
B. Inhibition of Enzymatic Activity and Disruption of Electron Transport Chains
Minor and major biochemical processes in the body depend upon enzymes for catalysis
to bring about homeostasis. However, these enzymes can be hampered by the physiological
conditions. Palmitic acid just like any other fatty acid might have the capability to penetrate
through the cell wall, which could cause irreversible deformation of the cell membranes (Pohl
et al., 2011; Liu et al. 2008; Avis & Belanger, 2001; Altieri et al., 2007). Since these are
protected by membrane proteins, the conformational forms of these proteins could be
disrupted depending on the levels require in bring such effect within the cell. When the
enzymes are inhibited it results in the disruption of the ATP synthesis due to decoupling
effect on the energy chain which affects the ATP synthase responsible for production and
regulation of ATPs in the cell (Desbois & Smith, 2010; Harold, 1972). Palmitic acid might
cause a direct binding to the electron carriers, insertion between carriers preventing
interaction, complete displacement of carriers from membranes and prevention of carriers
interaction by reducing fluidity of the membranes (Pohl et al., 2011; Liu et al. 2008; Avis &
Belanger, 2001; Altieri et al., 2007). However, further studies will be needed to actually
conclude its mechanism of action.
C. Impartment of Nutrient Uptake and Release
Every microorganism has well defined coordination systems that allow nutrient uptake
into the cellular level for the cell growth and bioenergetics purposes. However, if the cell
membranes and its associated proteins that is performing this role is disrupted then the
coordination becomes poor for the cell to do its selectivity in terms of nutrient uptake. It has
been proposed by (Desbois & Smith, 2010) that the presence of holes created by free fatty
acids causes leakages which cause impairment of active nutrient uptake by either indirectly
or direct on transport proteins. In most nutrient uptake the effect of this fatty acid is directly
influence by the inability for the transport protein to carry their function properly due to
conformational changes during cell membrane and its associated machinery disruption. On
the part of the indirect route used by free fatty acids to destabilized proper nutrient uptake into
vital parts of the cell could be attributed to the not favorable condition received by the cell
due to leaking, unregulated exchange of palmitic acid between inner and out membranes
machinery which results in the cells inability to produce ATP. The interesting biochemistry of
these effects is that in larger animals the cell organization is totaling different and that helps
prevent leakages in humans and associated mammalian species.

28

Melissa Johnson and Daniel A. Abugri

D. Release of Toxic Peroxidation and Autoxidations Products


During dietary intake of PA and metabolic activities within the cell certain toxic
peroxidation and autoxidations metabolites can result. These products when not cleared in a
fast manner could result in deleterious effect on bacteria and other related pathogens.
E. Palmitoylation and Cell Signaling
Fatty acid generally have the ability to acylate with protein, during posttranslational
modification makes them unique for up-regulating and down-regulating of certain cell
signaling in the cell (Resh, 2004; Smotrys and Linder, 2004).
Fatty acids are usually covalent bonded to proteins during posttranslational modification
(Drisdel et al. 2006).
This is common in eukaryotic cell than in prokaryotic cell (Drisdel et al. 2006). The
mechanisms employed by fatty acids to link up with proteins in the cellular levels are diverse
in nature and not only that, they involve several different processing events depending on the
fatty acid of interest. Palmitolylation, mysristolylation and prenylation are the most common
once observed in posttranslational modification (Resh, 2004; Smotrys & Linder, 2004).
The ability for palmitic acid to undergo posttranslational modification in a form of
palmitoylation is very important for cell signaling and binding to membrane proteins (Ross,
1995). For instance, palmitolylation aid in proper binding to particular proteins and
enhancement of membrane attachment (Ross, 1995). This allows for specificity in location of
G-proteins because of changes in receptors proteins in the cells (Ross, 1995). This may help
the body of the host to trafficked these proteins that might have been invaded by pathogens
for survival and replication (Ross, 1995).
F. Phospholipids Synthesis
Palmitic acids play an important role in the synthesis of phospholipids which are the key
class of lipids for cell membrane, brain and nerves coordination and integrity (Greseth &
Traktman, 2014). In some prokaryotes, PA is required for the phospholipids to be synthesis
which helps them to survive; for example, viral phospholipids biosynthesis for survival
(Greseth & Traktman, 2014).
G. PA Induced Proton Permeability of the Inner Mitochondrial Membrane
In 1950s, studies have testified the distribution of mitochondria energy coupling potential
by lipophilic extracts obtained from microsomes and other organic derived materials
(Wojtczak & Wieckowski, 1999). According to these authors, the uncoupling effects and
phosphorylation yield within the mitochondrial were observed to be controlled when an
isolated mitochondria using serum albumin with high binding ability for fatty acids was tested
(e.g. Palmitic acid) (Wojtczak, 1976; Wojtczak & Schnfeld, 1993).
Naturally, uncoupling compounds have been identified to include nonesterified fatty
acids forms in which palmitic acid could be one of them (Borst et al., 1962; Pressman &
Lardy, 1956; Wojtczak & Lehninger, 1961). Further evidence, on fatty acid been accumulated
in larger amounts inside or trapped by any isolated mitochondria has been reported to be
responsible for poor P/O ratios in aged rat liver mitochondria (Chefurka & Dumas, 1966).
These fatty acids turn to create swelling of the organelle (Mitochondria) resulting in

Occurrence, Biochemical, Antimicrobial and Health Effects of Palmitic Acid

29

improperly disruption of cell energy dispensation (Lehninger & Remmert, 1959; Wojtczak &
Lehninger, 1961; Lehninger, 1962).
The possible causes for these swelling in the mitochondrial depended on the chain length
and degree of unsaturation. This potential is similar to the features used to disrupt the energy
coupling processes in cellular (Pressman & Lardy, 1956).

Adapted from (Fatima, 2012 #103).


Figure 5. Fatty acid and triacylglycerol biosynthetic pathways.

30

Melissa Johnson and Daniel A. Abugri

There have been strong correlations between fatty acids and stimulation of mitochondrial
respiration and their potentials as protonophoric properties (Cunarro & Weiner, 1975).
Furthermore, the longer the chain of the fatty acid, the more the proton conductance will
occur in the phospholipid bilayer membranes (Gutknecht, 1988). This implies that multiple
studies are in agreement with the observation (Gutknecht, 1988). Palmitic acid may be using a
similar mechanism as any other fatty acids which are able to facilitate transmembrane flux of
some other cations, namely monovalent alkali metal cations ( Liu et al. 2008; Avis &
Belanger, 2001; Altieri et al., 2007; Zeng, Han, & Gross, 1998). However, research is needed
to draw a conclusive mechanism of PA under the membrane flux potential. Most of the
monovalent elements that palmitic can help in exchange between membranes are potassium,
sodium and lithium (Zborowski & Wojtczak, 1963). The mechanism is made possible by
(Pressman & Lardy, 1956; Zborowski & Wojtczak, 1963; Schonfeld, Schild & Kunz, 1989).
The capacity of palmitic acid in its undissociated form to perform flip-flop in the inner
mitochondrial membrane (Zborows; ki & Wojtczak, 1963) makes it potent for interfering
with the pathogens monovalent machinarys. The second possible way palmitic acid is
utilizing in inhibiting or killing pathogens could be due to its ability to exist in the anionic
form. This form is transferred by the adenine nucleotide translocator as well as other proteins.
It is important to note that the process mentioned above is possible based on size and the
structural nature of the fatty acid molecule in question. Many authors have found out that
fatty acid has highest potency to uncouple oxidative phosphorylation (Pressman & Lardy,
1956; Schnfeld, Schild, & Kunz, 1989), to induce mitochondrial swelling (Zborowski &
Wojtczak, 1963), and to promote energy-dependent accumulation of monovalent cations in
mitochondria (Wojtczak, 1974). This could possibly be the same as what has been observed
in palmitic acid in fungus inhibition and killing (Wojtczak, 1974; Choi et al. 2010).

1.5. BIOCHEMISTRY OF PALMITIC ACID


Metabolism of Palmitic Acid
The metabolism of palmitic acid may be broadly characterized as anabolic (i.e. synthesis)
or catabolic (i.e. degradation) (Figure 6). The primary route of palmitic acid metabolism is via
oxidative degradation or -oxidation within the mitochondria. During this process palmitic
acid is converted to acetyl-coA, which then enters the Citric Acid Cycle (or Krebs Cycle),
during which energy is generated (Figure 7). Depending upon the homeostatic needs, palmitic
acid may also be converted to other fatty acids such as stearic, oleic, palmitoleic and myristic
acids within the liver and intestinal mucosa. Further, palmitic acid may undergo omegaoxidation, a metabolic pathway often employed during episodes of increased hepatic fatty
acid influx and as a compensatory mechanism in the presence of a fatty acid oxidation
disorder (Hoek-van den Hil et al., 2013; Wanders, Komen, & Kemp, 2011).
Palmitic acid has been demonstrated to be metabolized utilizing different pathways than
other fatty acids. For example, PA was found to be metabolized in human skeletal muscle
cells in a mechanism quite different from that of oleic acid (Bakke et al., 2012). In addition,
cellular uptake, the incorporation into cellular phospholipids, desaturation and oxidation of
palmitic acid was found to differ from that of oleic, as well as stearic acids in hamster

Occurrence, Biochemical, Antimicrobial and Health Effects of Palmitic Acid

31

hepatocytes (Bruce & Salter, 1996). Others found palmitic acid to be metabolized at a much
lower rate than myristic acid in rat hepatocytes; myristic acid exhibited a significantly greater
rate of -oxidation and elongation (Rioux, Lemarchal, & Legrand, 2000). In the presence of
other fatty acids, the rates of gluconeogenesis, palmitic acid metabolism and the metabolism
of long-chain fatty acids is adapted accordingly in both humans and animals (Emken, 1994;
Mashek, Bertics, & Grummer, 2002). Palmitic acid also influences the metabolism of lipids,
lipoproteins, total and HDL cholesterol (Mensink et al., 2003; Ramamoorthy, Gupta, &
Khosla, 2000; Sanders et al., 2011; Snook et al., 1999). However, others found diets high in
palmitic acid to alter neither fasting nor postprandial levels of homocysteine or other
inflammatory biomarkers (e.g. TNF-, IL-1, IL-6 and IL-8, high-sensitivity C-reactive
protein and interferon-) (Voon et al., 2011).
Although researchers found palmitic acid supplementation to facilitate an increase in
plasma cholesterol concentrations in a metabolic-diet study in comparison to lauric acid
(Denke & Grundy, 1992), conflicting findings were presented elsewhere, with dietary
palmitic acid resulting in lower serum cholesterol concentrations than a lauric-myristic acid
combination (Sundram, Hayes, & Siru, 1994). In a study examining the effect of palmitic acid
intake (high vs low) on the endogenous synthesis of cholesterol and plasma lipoprotein
cholesterol levels, palmitic acid was unable to illicit a significant effect on lipoprotein
profiles, when recommended intakes of dietary linoleic acid (C18:2n6) were achieved
(Clandinin et al., 2000). Researchers have found increasing dietary palmitic acid to decrease
fatty acid oxidation and daily energy expenditure (Kien, Bunn, & Ugrasbul, 2005).

Figure 6. Summary of palmitic acid metabolism.

32

Melissa Johnson and Daniel A. Abugri

Figure 8. Simplified illustration outlining the catabolism of palmitic acid as indicated by: 1) -oxidation
of palmitic acid into acetyl CoA; 2) acetyl CoA entry into the Citric Acid Cycle; and 3) electron
transfer via the Electron Transport Chain.

Palmitic acid serves as the major metabolic fuel, particularly in the brain, which utilizes
fatty acids as a major fuel source during fatty acid oxidation and active transport into cerebral
microvessels (Williams et al., 1997). Up on transport across the palmitic is incorporated into
brain phospholipids, contributing to ~29% of phospholipids (mol/g) in rat brain (Rapoport,
2001). The uptake of palmitic acid is enhanced with the expression of the fatty acid transport
protein, which facilitates the transcellular transversion of fatty acids across the blood-brain

Occurrence, Biochemical, Antimicrobial and Health Effects of Palmitic Acid

33

barrier (Mitchell & Hatch, 2011). During metabolic conditions of starvation and diet-induced
obesity, palmitic acid in triglyceride form but not free fatty acid form, when administered
intravenously was able to inhibit the transport of leptin across the blood-brain barrier (Banks
et al., 2004). This suggests the role of increased triglyceride levels and subsequent
hypertriglyceridemia during starvation and conditions such as diabetes in mediating the
homeostasis of hormones such as leptin, which regulates fat storage and energy expenditure
and in certain cases inducing the resistance of certain hormones to enter the brain.

1.6. HEALTH IMPLICATIONS OF PALMITIC ACID


As with any dietary constituent, the evidence on the influence of palmitic acid on health
is variable. Rather than identifying the influence of palmitic acid on health as effects, it
may be more appropriate to associate the mechanisms, actions, and interactions of palmitic
acid as potential implications. As palmitic acid is one of the most abundant fatty acids,
functioning as potent uncoupling agents of cell communication it is not surprising that it
occupies a substantial role in metabolism, health and disease (Kremmyda et al., 2011;
Tvrzicka et al., 2011). Palmitic acid has been identified as a potential dietary agent for
increased cardiovascular disease (CVD) risk (Siri-Tarino et al., 2010).
Palmitic acid has been implicated as an important fatty acid in growth and development.
For example, the exposure of mice blastocyte cells to increasing concentrations of palmitic
acid resulted in increased apoptosis, decreased proliferation, suggesting the long-term effects
of palmitic acid exposure on embryonic metabolism and growth (Jungheim et al., 2011). In
addition, mice blastocyte exposure to palmitic acid resulted in fetal growth restriction, which
offspring were later able to overcome and reach weights that caught up with or surpassed
control mice. The ability of palmitic acid to induce endoplasmic reticulum stress and cellular
apoptosis has also been observed in hepatoma cells (Zhang et al., 2012), human granulosa
cells (Mu et al., 2001), rat cardiomyocytes (Dyntar et al., 2001) and rat testicular Leydig cells
(Lu et al., 2003). Palmitic acid is one of the major agents of free fatty acid-induced apoptosis
(i.e. lipoapoptosis) and toll-like receptor activation implicated in innate immune response
(Malhi & Gores, 2008).
Increases in dietary palmitic acid have been associated with decreases in fat oxidation
and daily energy expenditure in adults, which may increase the risk for obesity and insulin
resistance (Kien, Bunn, & Ugrasbul, 2005). Palmitic acid was positively associated with risk
of heart failure (Matsumoto et al., 2013). As a major participant in the de novo lipogenesis
pathway, palmitic acid and its successor palmitoleic acid have the potential to offer a
significant contribution to the risk of cardiovascular disease as influenced by the
endogenous fatty acid pool and lipid profile (Wu et al., 2011). In addition to modulating
tissue lipid profile, PA influences glucose metabolism via its actions on the hypothalamus
(Benoit et al., 2009), thus influencing insulin and leptin secretion, both key hormones in
appetite signaling, glucose metabolism, weight regulation, which may have implications on
risk for obesity, insulin resistance and diabetes mellitus. Palmitic acid, via the interruption of
-cell function, turnover and subsequent apoptosis, promotes the lipotoxicity and
glucotoxicity contributing to diabetes, the effects of which were ameliorated with palmitoleic
acid (Maedler et al., 2003; Maedler et al., 2001). A fatty acid profile characteristic of elevated

34

Melissa Johnson and Daniel A. Abugri

palmitoleic acid content have been observed in metabolic disorders such as anorexia nervosa,
obesity, metabolic syndrome (Kremmyda et al., 2011).
Recently it has been suggested that not all saturated fats increase the risk for
cardiovascular and other diseases, but that the triglyceride structure of the fatty acid is of
more importance when assessing disease risk (Fattore & Fanelli, 2013). Studies investigating
the influence of palm oil, which is comprised of ~50% palmitic acid, 40% oleic acid and 10%
linoleic acid, have yielded conflicting evidence in the relationship between palm oil and
cardiovascular disease risk. However, the replacement of trans fatty acids with palm oil more
favorably influenced biomarkers for disease (Fattore et al., 2014). The inclusion of palm oil
into the diets of moderately hyperlipidemic individuals adversely altered plasma lipid
profiles, more noticeable increases LDL-cholesterol concentrations (Vega-Lpez et al., 2006).
Insulin sensitivity was reduced, HDL- and total cholesterol significantly increased and fatty
acid oxidation was moderately increased in individuals consuming diets enriched in saturated
fatty acids (9% palmitic acid) compared to those consuming diets enriched in
monounsaturated fatty acids (9% oleic acid) (Lovejoy et al., 2002). Dietary monounsaturated
fatty acids have been proposed to provide protection from the risks associated with metabolic
syndrome and cardiovascular diseases (Gillingham, Harris-Janz, & Jones, 2011). Palmitoleic
acid (C16:1n7), the monounsaturated complement of palmitic acid (C16:0), is believed to
provide such protection.

Role of Palmitic acid in Inflammatory Pathways


The consumption of excessive energy intake prompts the conversion of excess
carbohydrates into fatty acids for storage and an enhanced adipocyte inflammatory response,
observed during the initiation and progression of obesity, insulin resistance and metabolic
syndrome (Kennedy et al., 2009). Palmitic acid, as a modulator of immune responses,
functions as an anti-inflammatory agent capable of binding to the active site of phospholipase
A2 (PLA2) and thus interfere with its enzyme kinetics and catalytic function inhibiting the
synthesis of the inflammatory eicosanoids resulting from the catalytic hydrolysis of
arachidonic acid (Aparna et al., 2012). However others found palmitic acid (when
administered to elicit a substantial hyperproliferation response) to induce the production of
proinflammatory cytokines in keratinocytes by inducing an upregulation of IL-6, TNF-, IL1 secretion and NF-activation and translocation (Zhou et al., 2013). In addition, palmitic
acid, isolated from Sargassum fusiforme, when bound to a novel pocket on the CD4+ cell
receptor was found to obstruct the entry of HIV into the cell and subsequent infection (Lee et
al., 2009; Lin et al., 2011; Paskaleva et al., 2014; Paskaleva et al., 2010).

CONCLUSION
This chapter emphasized palmitic acid, its physical, chemical and structural features as
pertaining to its functional characteristics, occurrence and percent distribution in plants, algae,
fungus, human and animal tissues. The antimicrobial mechanisms of action, metabolism,
general biochemistry and health implications of palmitic acid provide insight into the versatile

Occurrence, Biochemical, Antimicrobial and Health Effects of Palmitic Acid

35

nature of this fatty acid. Palmitic acid, one of the most abundant saturated fatty acids in nature
and within living systems, offers more than meets the eye. Palmitic acid is a master and
universal saturated fatty acid that has several benefits and effects when in high concentration.
Both de novo and salvage pathways ultilze palmitic acid for the biochemical synthesis of
short, medium, and long(er) chain fatty acids as well as other phospholipids, which are
required for cell mebrane structure and function, brain functionality and neuronal stability.
Palmitic acid is the only fatty acid posttranslation modification is reversible and because of
this unique feature cell signalling and trafficking is effect in the cell. Although, palmitic acid
has unique benefits, its biochemistry is still not explored into in both prokaryotes and
eukaryotes. Also because of the mechanisms exerted by palmitic acid for instance cell
signalling and trafficking, there is a possibility of it been used in theraupeutics and as
nutraceuticals for human microbial infection prevention. Although the evidence is conflicting
regarding the role of palmitic acid in health and its implications for health promotion and
disease prevention, there exists the potential for palmitic acid to serve as a functional and
bioactive endogenous and exogenous constituent of homeostasis.

REFERENCES
Abdulaziz, I., & Ai-Bahrany, I. (2002). Chemical Composition and Fatty Acid Analysis of
Saudi Hassawi Rice Oryza sativa L. Pak J Biol Sci, 5(2), 212-214.
Abugri, D. A., McElhenney, W. H., & Willian, K. R. (2012). Comparison of
Transesterification Methods for Fatty Acid Analysis in Higher Fungi: Application to
Mushrooms. Food Anal Methods, 5(5), 1159-1166.
Abugri, D. A., Tiimob, B. J., Apalangya, V. A., Pritchett, G., & McElhenney, W. H. (2013).
Bioactive and nutritive compounds in Sorghum bicolor (Guinea corn) red leaves and their
health implication. Food Chem, 138(1), 718-723.
Almazan, A. M., & Adeyeye, S. O. (1998). Fat and fatty acid concentrations in some green
vegetables. J Food Compost Anal, 11(4), 375-380.
Altieri, C., Cardillo, D., Bevilacqua, A., & Sinigaglia, M. (2007). Inhibition of Aspergillus
spp. and Penicillium spp. by fatty acids and their monoglycerides. J Food Prot, 70(5),
1206-1212.
Aparna, V., Dileep, K. V., Mandal, P. K., Karthe, P., Sadasivan, C., & Haridas, M. (2012).
AntiInflammatory Property of nHexadecanoic Acid: Structural Evidence and Kinetic
Assessment. Chem Biol Drug Des, 80(3), 434-439.
Armstrong, W. M. (1957). Surface active agents and cellular metabolism. I. The effect of
cationic detergents on the production of acid and of carbon dioxide by baker's yeast. Arch
Biochem Biophys, 71(1), 137-147.
Avis, T.J., & Belanger, R.R.(2001). Specificity and mode of action of the antifungal fatty acid
cis-9-heptadecenoic acid produced by Pseudozyma flocculosa. Applied and
Environmental Microbiology. 67, 956-960.
Bakke, S. S., Moro, C., Nikoli, N., Hessvik, N. P., Badin, P. M., Lauvhaug, L., & Kersten, S.
(2012). Palmitic acid follows a different metabolic pathway than oleic acid in human
skeletal muscle cells; lower lipolysis rate despite an increased level of adipose

36

Melissa Johnson and Daniel A. Abugri

triglyceride lipase. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of


Lipids, 1821(10), 1323-1333.
Banks, W. A., Coon, A. B., Robinson, S. M., Moinuddin, A., Shultz, J. M., Nakaoke, R., &
Morley, J. E. (2004). Triglycerides induce leptin resistance at the blood-brain barrier.
Diabetes, 53(5), 1253-1260.
Barros, L., Carvalho, A. M., & Ferreira, I. C. F. R. (2010). Leaves, flowers, immature fruits
and leafy flowered stems of Malva sylvestris: A comparative study of the nutraceutical
potential and composition. Food Chem Toxicol, 48(6), 1466-1472.
Benoit, S. C., Kemp, C. J., Elias, C. F., Abplanalp, W., Herman, J. P., Migrenne, S., & Yu, F.
(2009). Palmitic acid mediates hypothalamic insulin resistance by altering PKC-
subcellular localization in rodents. J Clin Invest, 119(9), 2577.
Bigogno, C., Khozin-Goldberg, I., Boussiba, S., Vonshak, A., & Cohen, Z. (2002). Lipid and
fatty acid composition of the green oleaginous alga Parietochloris incisa, the richest
plant source of arachidonic acid. Phytochemistry, 60(5), 497-503.
Blanc, M., Blaskovic, S., & Goot, F. (2013). Palmitoylation, pathogens and their host.
Biochem Soc Trans, 41(1).
Borst, P., Loos, J. A., Christ, E. J., & Slater, E. C. (1962). Uncoupling activity of long-chian
fatty acids. Biochimica et biophysica acta, 62(3), 509-518.
Boughton, B., & Wheatley, V. R. (1959). The fatty acid composition of the skin surface fat
(sebum) of normal human subjects. J Invest Dermatol, 33(2), 49-55.
Bruce, J., & Salter, A. (1996). Metabolic fate of oleic acid, palmitic acid and stearic acid in
cultured hamster hepatocytes. Biochem J, 316, 847-852.
Carmo Jr, A. C., de Souza, L. K. C., da Costa, C. E. F., Longo, E., Zamian, J. R., & da Rocha
Filho, G. N. (2009). Production of biodiesel by esterification of palmitic acid over
mesoporous aluminosilicate Al-MCM-41. Fuel, 88(3), 461-468.
Chamberlain, N. R., Mehrtens, B. G., Xiong, Z., Kapral, F. A., Boardman, J. L., & Rearick, J.
I. (1991). Correlation of carotenoid production, decreased membrane fluidity, and
resistance to oleic acid killing in Staphylococcus aureus 18Z. Infect Immun, 59(12),
4332-4337.
Chefurka, W., & Dumas, T. (1966). Oxidative phosphorylation in in vitro aged mitochondria.
II. Dinitrophenol-stimulated adenosine triphosphatase activity and fatty acid content of
mouse liver mitochondria. Biochem, 5(12), 3904-3911.
Choi, G.J., Jang, K.S., Choi, Y.H., Yu, J.H.& Kim, J-C.(2010). Antifungal activity of lower
alkyl fatty acids esters against powdery mildews. Plant Pathology Journal. 26(4), 360366.
Corvi, M.M., Alonso, A.M., and Caballero, M.C. (2012). Protein Palmitoylation and
Pathogenesis in Apicomplexan Parasites. Journal of Biomedicine and Biotechnology
Volume 2012 (2012), Article ID 483969, 9 pages
Clandinin, M. T., Cook, S. L., Konard, S. D., & French, M. A. (2000). The effect of palmitic
acid on lipoprotein cholesterol levels. Intl J Food Sci Nutr, 51(s1), s61-s71.
Denke, M. A., & Grundy, S. M. (1992). Comparison of effects of lauric acid and palmitic acid
on plasma lipids and lipoproteins. Am J Clin Nutr, 56(5), 895-898.
Desbois, A. P., & Smith, V. J. (2010). Antibacterial free fatty acids: activities, mechanisms of
action and biotechnological potential. Appl Microbiol Biotechnol, 85(6), 1629-1642.

Occurrence, Biochemical, Antimicrobial and Health Effects of Palmitic Acid

37

Dykstra, M., Cherukuri, A., Sohn, H. W., Tzeng, S.-J., & Pierce, S. K. (2003). Location is
everything: lipid rafts and immune cell signaling. Annu Rev Immunol, 21(1), 457-481.
doi: doi:10.1146/annurev.immunol.21.120601.141021
Dyntar, D., Eppenberger-Eberhardt, M., Maedler, K., Pruschy, M., Eppenberger, H. M.,
Spinas, G. A., & Donath, M. Y. (2001). Glucose and palmitic acid induce degeneration of
myofibrils and modulate apoptosis in rat adult cardiomyocytes. Diabetes, 50(9), 21052113.
Emken, E. A. (1994). Metabolism of dietary stearic acid relative to other fatty acids in human
subjects. Am J Clin Nutr, 60(6), 1023S-1028S.
Fattore, E., Bosetti, C., Brighenti, F., Agostoni, C., & Fattore, G. (2014). Palm oil and blood
lipidrelated markers of cardiovascular disease: a systematic review and meta-analysis of
dietary intervention trials. Am J Clin Nutr, ajcn. 081190.
Fattore, E., & Fanelli, R. (2013). Palm oil and palmitic acid: a review on cardiovascular
effects and carcinogenicity. Intl J Food Sci Nutr, 64(5), 648-659.
Fluhr, J. W., Kao, J., Jain, M., Ahn, S. K., Feingold, K. R., & Elias, P. M. (2001). Generation
of free fatty acids from phospholipids regulates stratum corneum acidification and
integrity. J Invest Dermatol, 117(1), 44-51.
Galbraith, H., & Miller, T. B. (1973). Physicochemical effects of long chain fatty acids on
bacterial cells and their protoplasts. J Appl Bacteriol, 36(4), 647-658. doi:
10.1111/j.1365-2672.1973.tb04150.x
Garaulet, M., Prez-Llamas, F., Prez-Ayala, M., Martnez, P., de Medina, F. S., Tebar, F. J.,
& Zamora, S. (2001). Site-specific differences in the fatty acid composition of abdominal
adipose tissue in an obese population from a Mediterranean area: relation with dietary
fatty acids, plasma lipid profile, serum insulin, and central obesity. Am J Clin Nutr, 74(5),
585-591.
Gillingham, L. G., Harris-Janz, S., & Jones, P. J. H. (2011). Dietary monounsaturated fatty
acids are protective against metabolic syndrome and cardiovascular disease risk factors.
Lipids, 46(3), 209-228.
Goldfine, H., & Bloch, K. (1961). On the origin of unsaturated fatty acids in Clostridia. J Biol
Chem, 236(10), 2596-2601.
Greenway, D. L. A., & Dyke, K. G. H. (1979). Mechanism of the inhibitory action of linoleic
acid on the growth of Staphylococcus aureus. J Gen Microbiol, 115(1), 233-245.
Greseth, M. D., & Traktman, P. (2014). De novo fatty acid biosynthesis contributes
significantly to establishment of a bioenergetically favorable environment for vaccinia
virus infection. PLoS Pathog, 10(3), e1004021. doi: 10.1371/journal.ppat.1004021
Grifths, R.G. Dancer, J., ONeill, E., and Harwood, J.L. (2003). Effect of culture conditions
on the lipid composition of Phytophthora infestans. New Phytologist, 158: 337344
Harold, F. M. (1972). Conservation and transformation of energy by bacterial membranes.
Bacteriol Rev, 36(2), 172.
Heleno, S. A., Barros, L., Sousa, M. J., Martins, A., & Ferreira, I. C. F. R. (2009). Study and
characterization of selected nutrients in wild mushrooms from Portugal by gas
chromatography and high performance liquid chromatography. Microchem J, 93(2), 195199.
Hoek-van den Hil, E. F., Keijer, J., Bunschoten, A., Vervoort, J. J. M., Stankova, B.,
Bekkenkamp, M., . . . van Schothorst, E. M. (2013). Quercetin Induces Hepatic Lipid

38

Melissa Johnson and Daniel A. Abugri

Omega-Oxidation and Lowers Serum Lipid Levels in Mice. PLoS ONE, 8(1), e51588.
doi: 10.1371/journal.pone.0051588
Hofmann, K., Henis, D. B., & Panos, C. (1957). Fatty acid interconversions in lactobacilli. J
Biol Chem, 228(1), 349-355.
Hofmann, K., Hsiao, C.-Y. Y., Henis, D. B., & Panos, C. (1955). The estimation of the fatty
acid composition of bacterial lipides. J Biol Chem, 217(1), 49-60.
Hofmann, K., Lucas, R. A., & Sax, S. M. (1952). The chemical nature of the fatty acids of
Lactobacillus arabinosus. J Biol Chem, 195(2), 473-485.
Hofmann, K., & Sax, S. M. (1953). The chemical nature of the fatty acids of Lactobacillus
casei. J Biol Chem, 205(1), 55-63.
Hofmann, K., & Tausig, F. (1955a). The chemical nature of the fatty acids of a group C
Streptococcus species. J Biol Chem, 213(1), 415-423.
Hofmann, K., & Tausig, F. (1955b). On the identity of phytomonic and lactobacillic acids. A
reinvestigation of the fatty acid spectrum of Agrobacterium (Phytomonas) tumefaciens. J
Biol Chem, 213(1), 425-432.
Houde, A., Kademi, A., & Leblanc, D. (2004). Lipases and their industrial applications. Appl
Biochem Biotechnol, 118(1-3), 155-170.
Innis, S. M., Dyer, R., & Nelson, C. M. (1994). Evidence that palmitic acid is absorbed assn-2
monoacylglycerol from human milk by breast-fed infants. Lipids, 29(8), 541-545.
Jaeger, K.-E., & Reetz, M. T. (1998). Microbial lipases form versatile tools for
biotechnology. Trends Biotechnol, 16(9), 396-403.
Johnson, M., Pace, R. D., Dawkins, N. L., & Willian, K. R. (2013). Diets containing
traditional and novel green leafy vegetables improve liver fatty acid profiles of
spontaneously hypertensive rats. Lipids Health Dis, 12(1), 168.
Jungheim, E. S., Louden, E. D., Chi, M. M.-Y., Frolova, A. I., Riley, J. K., & Moley, K. H.
(2011). Preimplantation exposure of mouse embryos to palmitic acid results in fetal
growth restriction followed by catch-up growth in the offspring. Biol Reprod, 85(4), 678683.
Kabara, J. J., Swieczkowski, D. M., Conley, A. J., & Truant, J. P. (1972). Fatty acids and
derivatives as antimicrobial agents. Antimicrob Agents Chemother, 2(1), 23-28.
Karmakar, K., Muslim, T., & Rahman, A. Fatty acid composition of some leafy vegetables of
Bangladesh Intl J Pharm Sci Res, 4(2), 694-697.
Keng, P. S., Basri, M., Zakaria, M. R. S., Rahman, M. B. A., Ariff, A. B., Rahman, R. N. Z.
A., & Salleh, A. B. (2009). Newly synthesized palm esters for cosmetics industry. Ind
Crops Prod, 29(1), 37-44.
Kennedy, A., Martinez, K., Chuang, C.-C., LaPoint, K., & McIntosh, M. (2009). Saturated
fatty acid-mediated inflammation and insulin resistance in adipose tissue: mechanisms of
action and implications. J Nutr, 139(1), 1-4. doi: 10.3945/jn.108.098269
Kien, C. L., Bunn, J. Y., & Ugrasbul, F. (2005). Increasing dietary palmitic acid decreases fat
oxidation and daily energy expenditure. Am J Clin Nutr, 82(2), 320-326.
Kremmyda, L.-S., Tvrzicka, E., Stankova, B., & Zak, A. (2011). Fatty acids as
biocompounds: their role in human metabolism, health and disease: a review. part 2: fatty
acid physiological roles and applications in human health and disease. Biomed Pap Med
Fac Univ Palacky Olomouc Czech Repub, 155(3).

Occurrence, Biochemical, Antimicrobial and Health Effects of Palmitic Acid

39

Lang, I., Hodac, L., Friedl, T., & Feussner, I. (2011). Fatty acid profiles and their distribution
patterns in microalgae: a comprehensive analysis of more than 2000 strains from the
SAG culture collection. BMC Plant Biol, 11(1), 124.
Lee, D. Y. W., Lin, X., Paskaleva, E. E., Liu, Y., Puttamadappa, S. S., Thornber, C., & Canki,
M. (2009). Palmitic acid is a novel CD4 fusion inhibitor that blocks HIV entry and
infection. AIDS Res Hum Retroviruses, 25(12), 1231-1241.
Lehninger, A. L., & Remmert, L. F. (1959). An endogenous uncoupling and swelling agent in
liver mitochondria and its enzymic formation. J Biol Chem, 234(9), 2459-2464.
Lehninger, A. L.(1962). Water Uptake and Extrusion by Mitochondria in Relation to
Oxidative Phosphorylation. Physiological Review. 42, 467-517.
Lin, X., Paskaleva, E. E., Chang, W., Shekhtman, A., & Canki, M. (2011). Inhibition of HIV1 infection in ex vivo cervical tissue model of human vagina by palmitic acid;
implications for a microbicide development. PLoS ONE, 6(9), e24803.
Liu, S., Ruan, W., Li, J., Xu, H., Wang, J., Gao, Y., & Wang, J. (2008). Biological control of
phytopathogenic fungi by fatty acids. Mycopathologia, 166(2), 93-102.
Lovejoy, J. C., Smith, S. R., Champagne, C. M., Most, M. M., Lefevre, M., DeLany, J. P., &
Bray, G. A. (2002). Effects of diets enriched in saturated (palmitic), monounsaturated
(oleic), or trans (elaidic) fatty acids on insulin sensitivity and substrate oxidation in
healthy adults. Diabetes care, 25(8), 1283-1288.
Lu, Z.-H., Mu, Y.-M., Wang, B.-A., Li, X.-L., Lu, J.-M., Li, J.-Y., . . . Nawata, H. (2003).
Saturated free fatty acids, palmitic acid and stearic acid, induce apoptosis by stimulation
of ceramide generation in rat testicular Leydig cell. Biochem Biophys Res Commun,
303(4), 1002-1007.
Maedler, K., Oberholzer, J., Bucher, P., Spinas, G. A., & Donath, M. Y. (2003).
Monounsaturated fatty acids prevent the deleterious effects of palmitate and high glucose
on human pancreatic -cell turnover and function. Diabetes, 52(3), 726-733. doi:
10.2337/diabetes.52.3.726
Maedler, K., Spinas, G., Dyntar, D., Moritz, W., Kaiser, N., & Donath, M. Y. (2001). Distinct
effects of saturated and monounsaturated fatty acids on -cell turnover and function.
Diabetes, 50(1), 69-76.
Malhi, H., & Gores, G. J. (2008). Molecular mechanisms of lipotoxicity in nonalcoholic fatty
liver disease. Paper presented at the Seminars in liver disease.
Mashek, D. G., Bertics, S. J., & Grummer, R. R. (2002). Metabolic fate of long-chain
unsaturated fatty acids and their effects on palmitic acid metabolism and gluconeogenesis
in bovine hepatocytes. J Dairy Sci, 85(9), 2283-2289.
Matsumoto, C., Hanson, N. Q., Tsai, M. Y., Glynn, R. J., Gaziano, J. M., & Djouss, L.
(2013). Plasma phospholipid saturated fatty acids and heart failure risk in the physicians'
health study. Clin Nutr, 32(5), 819-823.
Mensink, R. P., Zock, P. L., Kester, A. D. M., & Katan, M. B. (2003). Effects of dietary fatty
acids and carbohydrates on the ratio of serum total to HDL cholesterol and on serum
lipids and apolipoproteins: a meta-analysis of 60 controlled trials. Am J Clin Nutr, 77(5),
1146-1155.
Metzger, J. O., & Bornscheuer, U. (2006). Lipids as renewable resources: current state of
chemical and biotechnological conversion and diversification. Appl Microbiol
Biotechnol, 71(1), 13-22.

40

Melissa Johnson and Daniel A. Abugri

McGraw, L.J., Jager, A.K., & Van Staden, J. (2002). Isolation of antibacterial fatty acids from
Schotia brachypetala. Fitoterapia, 73, 431-433.
Mitchell, R. W., & Hatch, G. M. (2011). Fatty acid transport into the brain: Of fatty acid
fables and lipid tails. Prostaglandins Leukot Essent Fatty Acids, 85(5), 293-302.
Mu, Y.-M., Yanase, T., Nishi, Y., Tanaka, A., Saito, M., Jin, C.-H., & Nawata, H. (2001).
Saturated FFAs, palmitic acid and stearic acid, induce apoptosis in human granulosa
cells. Endocrinology, 142(8), 3590-3597. doi: doi:10.1210/endo.142.8.8293
O'Neil, M. J. (2013). The Merck index: an encyclopedia of chemicals, drugs, and biologicals:
RSC Publishing.
Paskaleva, E. E., Arra, M., Liu, Y., Guo, H., Swartz, G., Kennedy, J. S., & Canki, M. (2014).
Evaluation of Potential Genotoxicity of HIV Entry Inhibitors Derived from Natural
Sources. PLoS ONE, 9(3), e93108.
Paskaleva, E. E., Xue, J., Lee, D. Y. W., Shekhtman, A., & Canki, M. (2010). Palmitic acid
analogs exhibit nanomolar binding affinity for the HIV-1 CD4 receptor and nanomolar
inhibition of gp120-to-CD4 fusion. PLoS ONE, 5(8), e12168.
Pitto, M., Parenti, M., Guzzi, F., Magni, F., Palestini, P., Ravasi, D., & Masserini, M. (2002).
Palmitic is the main fatty acid carried by lipids of detergent-resistant membrane fractions
from neural and non-neural cells. Neurochem Res, 27(7-8), 729-734.
Pohl, C. H., Kock, J. L. F., & Thibane, V. S. (2011). Antifungal free fatty acids: a review.
Science against microbial pathogens: communicating current research and technological
advances. A Mendez-Vilas (Ed.), 60-71.
Pressman, B. C., & Lardy, H. A. (1956). Effect of surface active agents on the latent
ATPASE of mitochondira. Biochimica et biophysica acta, 21(3), 458-466.
Quintavalla, S., & Vicini, L. (2002). Antimicrobial food packaging in meat industry. Meat
Sci, 62(3), 373-380.
Ramamoorthy, L., Gupta, S. V., & Khosla, P. (2000). Effects of exchanging 4% en between
dietary stearic and palmitic acid on hamster plasma lipoprotein metabolism. Intl J Food
Sci Nutr, 51(1), S51-S59.
Rapoport, S. I. (2001). In vivo fatty acid incorporation into brain phosholipids in relation to
plasma availability, signal transduction and membrane remodeling. J Mol Neurosci, 16(23), 243-261; discussion 279-284.
Resh, M. D. (2004). Membrane targeting of lipid modified signal transduction proteins
Membrane Dynamics and Domains (pp. 217-232): Springer.
Resh, M. D. (2006). Trafficking and signaling by fatty-acylated and prenylated proteins. Nat
Chem Biol, 2(11), 584-590.
Rioux, V., Lemarchal, P., & Legrand, P. (2000). Myristic acid, unlike palmitic acid, is rapidly
metabolized in cultured rat hepatocytes. J Nutr Biochem, 11(4), 198-207.
Ross, E. M. (1995). Protein modification: palmitoylation in G-protein signaling pathways.
Curr Biol, 5(2), 107-109.
Rule, D. C. (1997). Direct transesterification of total fatty acids of adipose tissue, and of
freeze-dried muscle and liver with boron-trifluoride in methanol. Meat Sci, 46(1), 23-32.
Sabin, M. A., Crowne, E. C., Stewart, C. E., Hunt, L. P., Turner, S. J., Welsh, G. I., & Shield,
J. P. (2007). Depot-specific effects of fatty acids on lipid accumulation in childrens
adipocytes. Biochem Biophys Res Commun, 361(2), 356-361.

Occurrence, Biochemical, Antimicrobial and Health Effects of Palmitic Acid

41

Sanders, T. A. B., Filippou, A., Berry, S. E., Baumgartner, S., & Mensink, R. P. (2011).
Palmitic acid in the sn-2 position of triacylglycerols acutely influences postprandial lipid
metabolism. Am J Clin Nutr, 94(6), 1433-1441.
Scharff, T. G., & Beck, J. L. (1959). Effects of surface-active agents on carbohydrate
metabolism in yeast. Exp Biol Med, 100(2), 307-311.
Schmelzle, H., Wirth, S., Skopnik, H., Radke, M., Knol, J., Bckler, H. M., & Fusch, C.
(2003). Randomized Double-Blind Study of the Nutritional Efficacy and Bifidogenicity
of a New Infant Formula Containing Partially Hydrolyzed Protein, a High -Palmitic
Acid Level, and Nondigestible Oligosaccharides. J Pediatr Gastroenterol Nutr, 36(3),
343-351.
Schnfeld, P., Schild, L., & Kunz, W. (1989). Long-chain fatty acids act as protonophoric
uncouplers of oxidative phosphorylation in rat liver mitochondria. Biochimica et
Biophysica Acta (BBA)-Bioenergetics, 977(3), 266-272.
Seidel, V. & Taylor P.W. (2004). In vitro activity of extracts and constituents of Pelagonium
against rapily growing mycobacteria. International Journal of Antimicrobial Agents, 23,
613-619.
Shaw, M. K., & Ingraham, J. L. (1965). Fatty acid composition of Escherichia coli as a
possible controlling factor of the minimal growth temperature. J Bacteriol, 90(1), 141146.
Shin, S. Y., Bajpai, V. K., Kim, H. R., & Kang, S. C. (2007). Antibacterial activity of
eicosapentaenoic acid (EPA) against foodborne and food spoilage microorganisms. LWTJ Food Sci Technol, 40(9), 1515-1519.
Shirai, N., Suzuki, H., & Wada, S. (2005). Direct methylation from mouse plasma and from
liver and brain homogenates. Anal Biochem, 343(1), 48-53.
Sikkema, J., De Bont, J. A., & Poolman, B. (1995). Mechanisms of membrane toxicity of
hydrocarbons. Microbiol Rev, 59(2), 201-222.
Siri-Tarino, P. W., Sun, Q., Hu, F. B., & Krauss, R. M. (2010). Saturated fat, carbohydrate,
and cardiovascular disease. Am J Clin Nutr, 91(3), 502-509.
Smotrys, J. E., & Linder, M. E. (2004). Palmitoylation of intracellular signaling proteins:
regulation and function. Annu Rev Biochem, 73(1), 559-587.
Snook, J., Park, S., Williams, G., Tsai, Y., & Lee, N. (1999). Effect of synthetic triglycerides
of myristic, palmitic, and stearic acid on serum lipoprotein metabolism. Eur J Clin Nutr,
53(8), 597-605.
Sundram, K., Hayes, K. C., & Siru, O. H. (1994). Dietary palmitic acid results in lower serum
cholesterol than does a lauric-myristic acid combination in normolipemic humans. Am J
Clin Nutr, 59(4), 841-846.
Takigawa, H., Nakagawa, H., Kuzukawa, M., Mori, H., & Imokawa, G. (2005). Deficient
production of hexadecenoic acid in the skin is associated in part with the vulnerability of
atopic dermatitis patients to colonization by Staphylococcus aureus. Dermatology,
211(3), 240-248.
Temina, M., Rezankova, H., Rezanka, T., & Dembitsky, V. M. (2007). Diversity of the fatty
acids of the Nostoc species and their statistical analysis. Microbiol Res, 162(4), 308-321.
Tonon, T., Harvey, D., Larson, T. R., & Graham, I. A. (2002). Long chain polyunsaturated
fatty acid production and partitioning to triacylglycerols in four microalgae.
Phytochemistry, 61(1), 15-24.

42

Melissa Johnson and Daniel A. Abugri

Tvrzicka, E., Kremmyda, L.-S., Stankova, B., & Zak, A. (2011). Fatty acids as
biocompounds: their role in human metabolism, health and disease-a review. Part 1:
classification, dietary sources and biological functions. Biomed Pap Med Fac Univ
Palacky Olomouc Czech Repub, 155(2).
Vega-Lpez, S., Ausman, L. M., Jalbert, S. M., Erkkil, A. T., & Lichtenstein, A. H. (2006).
Palm and partially hydrogenated soybean oils adversely alter lipoprotein profiles
compared with soybean and canola oils in moderately hyperlipidemic subjects. Am J Clin
Nutr, 84(1), 54-62.
Viso, A.-C., & Marty, J.-C. (1993). Fatty acids from 28 marine microalgae. Phytochemistry,
34(6), 1521-1533.
Voon, P. T., Ng, T. K. W., Lee, V. K. M., & Nesaretnam, K. (2011). Diets high in palmitic
acid (16: 0), lauric and myristic acids (12: 0+ 14: 0), or oleic acid (18: 1) do not alter
postprandial or fasting plasma homocysteine and inflammatory markers in healthy
Malaysian adults. Am J Clin Nutr, 94(6), 1451-1457.
Wanders, R. J. A., Komen, J., & Kemp, S. (2011). Fatty acid omega-oxidation as a rescue
pathway for fatty acid oxidation disorders in humans. FEBS Journal, 278(2), 182-194.
doi: 10.1111/j.1742-4658.2010.07947.x
Wang, L.-L., & Johnson, E. A. (1992). Inhibition of Listeria monocytogenes by fatty acids
and monoglycerides. Appl Environ Microbiol, 58(2), 624-629.
Weitkamp, A. W., Smiljanic, A. M., & Rothman, S. (1947). The free fatty acids of human
hair fat. J Am Chem Soc, 69(8), 1936-1939.
Whetsell, M. S., B, R. E., & D, L. J. (2003). Human health effects of fatty acids in beef. Fact
sheet. In W. V. S. U. U. A. R. Service (Ed.), Extension Service West Virginia University
Willett, N. P., & Morse, G. E. (1966). Long-chain fatty acid inhibition of growth of
Streptococcus agalactiae in a chemically defined medium. J Bacteriol, 91(6), 2245-2250.
Williams, W. M., Chang, M. C. J., Hayakawa, T., Grange, E., & Rapoport, S. I. (1997). In
Vivo oncorporation from plasma of radiolabeled palmitate and arachidonate into rat brain
microvessels. Microvasc Res, 53(2), 163-166.
Wojtczak, L. (1974). Effect of fatty acids and acyl-CoA on the permeability of mitochondrial
membranes to monovalent cations. FEBS letters, 44(1), 25-30.
Wojtczak, L. (1976). Effect of long-chain fatty acids and acyl-CoA on mitochondrial
permeability, transport, and energy-coupling processes. J Bioenerg Biomembr, 8(6), 293311.
Wojtczak, L., & Lehninger, A. L. (1961). Formation and disappearance of an endogenous
uncoupling factor during swelling and contraction of mitochondria. Biochimica et
biophysica acta, 51(3), 442-456.
Wojtczak, L., & Schnfeld, P. (1993). Effect of fatty acids on energy coupling processes in
mitochondria. Biochimica et Biophysica Acta (BBA)-Bioenergetics, 1183(1), 41-57.
Wojtczak, L., & Wieckowski, M. R. (1999). The Mechanisms of Fatty Acid-Induced Proton
Permeability of the Inner Mitochondrial Membrane. Journal of Bioenergetics and
Biomembranes, 31(5), 447-455. doi: 10.1023/A:1005444322823
Wu, J. H. Y., Lemaitre, R. N., Imamura, F., King, I. B., Song, X., Spiegelman, D., &
Mozaffarian, D. (2011). Fatty acids in the de novo lipogenesis pathway and risk of
coronary heart disease: the Cardiovascular Health Study. Am J Clin Nutr, 94(2), 431-438.

Occurrence, Biochemical, Antimicrobial and Health Effects of Palmitic Acid

43

Yff, B.T.S., Lindsey, K.L., Taylor, M.B., Erasmus, D.G., & Jager, A.K.(2002). The
pharmacological screening of Pentanisia prunelloides and the isolation of the antibacterial
compound palmitic acid. Journal of Ethnopharmacology, 79, 101-107.
Zborowski, J., & Wojtczak, L. (1963). Induction of swelling of liver mitochondria by fatty
acids of various chain length. Biochimica et biophysica acta, 70, 596-598.
Zeng, Y., Han, X., & Gross, R. W. (1998). Phospholipid subclass specific alterations in the
passive ion permeability of membrane bilayers: separation of enthalpic and entropic
contributions to transbilayer ion flux. Biochem, 37(8), 2346-2355.
Zhang, Y., Xue, R., Zhang, Z., Yang, X., & Shi, H. (2012). Palmitic and linoleic acids induce
ER stress and apoptosis in hepatoma cells. Lipids Health Dis, 11(1), 1.
Zhou, B.-R., Zhang, J.-A., Zhang, Q., Permatasari, F., Xu, Y., Wu, D., & Luo, D. (2013).
Palmitic Acid Induces Production of Proinflammatory Cytokines Interleukin-6,
Interleukin-1, and Tumor Necrosis Factor- via a NF-B-Dependent Mechanism in HaCaT
Keratinocytes. Mediators Inflamm, 2013, 11. doi: 10.1155/2013/530429.

In: Palmitic Acid: Occurrence, Biochemistry and Health Effects ISBN: 978-1-63321-519-1
Editor: Lucas F. Porto
2014 Nova Science Publishers, Inc.

Chapter 3

PALMITIC ACID: EFFECT OF DIET


SUPPLEMENTATION AND OCCURRENCE IN ANIMAL
ORIGIN FOOD
P. G. Peiretti*
Institute of Sciences of Food Production,
Italian National Research Council, Grugliasco, Italy

ABSTRACT
In the last few decades, disagreement between opinions and findings concerning the
ability of palmitic acid (PA) and other saturated fatty acids (SFAs) to raise
cholesterolaemia has led to discussions on whether PA, which has been positively related
to high serum cholesterol levels, could increase the risk of cardiovascular diseases. This
study aims to review the PA content of meat, dairy products, fish, and other food of
animal origin in the human diet and discusses nutritional issues related to the occurrence
of this fatty acid (FA) in these foods due to different diet supplementation. Meat and
dairy products are considerable dietary sources of SFAs, such as PA. In most
industrialized countries, a high meat or dairy intake contributes to a higher than
recommended SFA intake. Palmitic and myristic acids are common FAs in meat and
dairy products, making up about 30-40% of total FA intake and are the main factors
responsible for raising cholesterol levels; indeed, strong evidence indicates that these two
SFAs increase serum cholesterol concentrations in humans. Stearic acid is partially
converted to oleic acid in vivo and has not been shown to elevate blood cholesterol, while
lauric acid is not as potent as PA at raising concentrations of total cholesterol and LDL
cholesterol in humans. The occurrence of PA in animal origin food is influenced by both
genetic and environmental factors, such as the composition of the animals diet, its
digestive system and its biosynthetic processes. The FA profile in food of animal origin
mainly reflects dietary lipid sources and has the potential to play a valuable role in human
nutrition by manipulating the composition of animal fat through diet. In order to explain
the variability in FA composition in food of animal origin, this review examines different

Corresponding author: P. G. Peiretti. Tel.: +39 11 6709230; fax: +39 11 6709297.


piergiorgio.peiretti@ispa.cnr.it.

E-mail address:

46

P. G. Peiretti
nutrition trials that have studied the effects of PA supplementation on the lipid profile of
animal origin food.

Keywords: Fatty acid, oilseed, meat, milk, cheese, fish, egg

INTRODUCTION
Palmitic acid (PA) or n-hexadecanoic acid is the most common saturated fatty acid (SFA)
found in plants, animals, and microorganisms (Gunstone et al., 2012). As the name suggests,
PA is characteristic of palm oil and its saponified form was discovered and isolated for the
first time by Frmy (1840). The major difference between palm oil and other vegetable oils is
the higher proportion of PA (Table 1).
The impact of excess SFA in the diet on cardiovascular diseases has been studied and
discussed both in animal and human studies (Crawford, 1968; Keys, 1970; Temme et al.,
1996). In the past, because of its high PA content, palm oil has been attacked as highly
saturated oil and accused of raising blood cholesterol and increasing the risk of
cardiovascular disease (Mukherjee and Mitra, 2009).
Several studies attack SFAs with regard to their hypercholesterolaemic and atherogenic
effects, which adversely affect cardiovascular risk (Kromhout et al., 1989; Menotti et al.,
1989; Verschuren et al., 1995). Lauric and myristic acid are the main cholesterol-raising
SFAs, whereas PA and stearic acid have much weaker cholesterol-raising potential (Sundram,
1994).
Table 1. Palmitic acid content of oils and fats of vegetable sources (expressed as
percentage mass-fraction of total fatty acids)
Palm oil
Cottonseed oil
Cocoa butter
Illipe fat
Olive oil
Oat bran oil
Avocado oil
Rice brain oil
Wheat germ oil
Corn oil
Tomato seed oil
Peanut oil
Soya bean oil
Pistachio nut oil
Grapeseed oil
Babassu fat
Poppyseed oil
Sesame oil

40.1-47.5
21.4-26.4
25.4
18.0-22.0
7.5-20.0
17.4
17.2
16.9
16.6
8.6-16.5
12.0-15.5
8.3-14.0
8.0-13.3
11.6
5.5-11.0
5.2-11.0
10.6
7.9-10.2

Palmitic Acid: Effect of Diet Supplementation and Occurrence


Palm kernel fat
Macadamia nut oil
Coconut oil
Walnut oil
Linseed oil
Almond oil
Pecan nut oil
Safflower oil
Linola oil
Cashew nut oil
Rapeseed oil
Sunflower oil
Hazelnut oil
Canola oil

47

6.5-10.0
8.9
8.2
3.9-7.2
4.0-7.0
6.5
6.4
4.8-6.2
6.0
4.0-6.0
1.5-6.0
5.4-5.9
5.2
4.0

Adapted from Beare-Rogers et al., 2001.

The negative effect attributed to lauric and myristic acid explains why foods rich in SFA
should be consumed in moderation and there is convincing evidence that PA increases the
risk of cardiovascular disease (World Health Organization, 2003). Palm oil raises plasma
cholesterol only when the diet contains excess dietary cholesterol, in which case the risk of
coronary heart disease may rise (Jones, 1989). Temme et al. (1996) reported that both PA and
lauric acid are hypercholesterolaemic compared with oleic acid.
Lauric acid raises total cholesterol concentrations more than PA, which is partly due to a
greater rise in HDL cholesterol. Enig (1993) reported that PA increased the level of blood
cholesterol more than other SFAs, including lauric acid and myristic acid, which are more
abundant in palm kernel oil and coconut oil than in palm oil. Clarke et al. (1997) concluded
that, compared to carbohydrates, PA raises blood cholesterol levels. However, some reviews
do not seem to support these conclusions (Edem, 2002; Ong and Goh, 2002; Sundram et al.,
2003; Oguntibeju et al., 2009; McNamara, 2010) and indicate that the effect of PA (found
mainly in palm oil) on blood cholesterol is relatively neutral when compared to other fats and
oils. We must therefore realize, although it may seem simplistic, that what matters is the
dietary context, rather than the individual nutrient. In a balanced diet, in fact, PA is usually
harmless (it is even synthesized by the body), but can become dangerous when consumed as
part of frequent caloric excesses or is consumed in particularly large quantities.
Hayes and Khosla (1992) suggested that PA may be neutral in normocholesterolaemic
subjects if the diet contains little cholesterol and linoleic acid intake is adequate. Fattore and
Fanelli (2013) reviewed the scientific literature on the evidence of the relationship between
palm oil and adverse effects on human health and concluded that there is no clear evidence of
a negative role of PA on health and much less of native palm oil, which is a complex
alimentary matrix, in which PA is only one of its components. However, more recent lipid
research on the topic seems to have reconsidered the negative role of dietary SFAs as a risk
factor for cardiovascular diseases. For instance, lauric acid and myristic acid have a greater
total cholesterol-raising effect than PA, whereas stearic acid has a neutral effect on the
concentration of total serum cholesterol, including no apparent impact on either LDL or HDL
(Daley et al., 2010).

48

P. G. Peiretti

Fattore and Fanelli (2013) showed that not only the type of fat, but also its triglyceride
structure, play a role in cholesterolaemia. PA is located at the Sn-1 position of the three
principal triglyceride species in palm oil (Small, 1991) and a high fraction of PA in palm oil
is bound at the Sn-1 or Sn-3 position of the glycerol molecule (Mu and Hy, 2004). This
location confers the non-hypercholesterolaemic property to the oil (Ng, 1997). This is in
contrast with the major triglyceride species in animal fats such as butter, which contains PA
in the Sn-2 position with resultant hypercholesterolaemic and atherogenic effects (Ng, 1997).
Evidence is now growing that the molecular structures of dietary triacylglycerols play an
important role in the development of atherosclerosis (Patsch, 1994), because triacylglycerols,
enriched with SFA at the Sn-2 position, exhibit different metabolic behaviour than
triacylglycerols with SFA at the Sn-1/3 position (Redgrave et al., 1988; Tuten et al., 1993;
Carnielli et al., 1995).
The enzymatic hydrolysis of dietary triglycerides by pancreatic and lipoprotein lipases
preferentially targets fatty acids (FAs) in the Sn-1/3 position rather than those esterified to the
Sn-2 position, which are substantially preserved in chylomicrons (Karupaiah and Sundram,
2007). These authors showed that the positioning of unsaturated versus SFAs in the Sn-2
position may explain the modulatory effects on atherogenicity and thrombogenicity.
Kritchevsky (2000) reported a higher degree of absorption of PA at the Sn-2 position in rabbit
models and this could be related to the increased atherogenicity of interesterified palm oil, in
comparison with the native one.
The predominant SFAs that occur naturally in animal fats and the main products of
cytosolic FA synthetase multienzyme complex in lipogenesis are stearic acid and PA, which
can be biosynthesized de novo by all known organisms, including fish (Sargent et al., 1989).
Some authors have determined enzyme activities in subcutaneous adipose tissue. StearoylCoA or 9 desaturase is the terminal step in the desaturation and conversion of PA, myristic
and stearic acid into the 9 monounsaturated FAs palmitoleic, myristoleic, and oleic acid,
respectively (De Smet et al., 2004). Kazala et al. (1999) suggested that FA elongation was
unable to keep pace with the de novo production of PA in animals that deposited greater
amounts of intramuscular fat. However, based on tissue incubations, it has been suggested
that 9 desaturation and not elongation is the rate-limiting step for the conversion of PA to
oleic acid in bovine subcutaneous adipose tissue (St. John et al., 1991).
This review examines the occurrence and variation of PA content in food of animal origin
in relation to different diets and different dietary regimens.

OCCURRENCE IN ANIMAL ORIGIN FOOD


PA and myristic acid are common FAs in dairy products and meat, contributing in total
about 3040% of FAs (Valsta et al., 2005). Approximately 60% of the SFAs in the US diet
are obtained from meat, poultry, fish and dairy products (Dupont et al., 1991). In a more
detailed analysis from the late 1980s of the FA consumption pattern of Americans, it was
shown that PA was the predominant SFA in the US diet at the time, contributing 5257% of
SFA intake (Jonnalagadda et al., 1995). Of the SFAs, short chain FAs and lauric and myristic
acid are obtained from dairy products, while the predominant sources of PA and stearic acid
are meat, poultry, fish and blended foods (Valsta et al., 2005).

Palmitic Acid: Effect of Diet Supplementation and Occurrence

49

FAT, MEAT AND MEAT PRODUCTS


In most industrialized countries, fat is an unpopular constituent of meat for consumers,
being considered unhealthy. This is because meat is seen to be a major source of fat in the
diet and especially of SFAs, which have been implicated in diseases associated with modern
life, especially in developed countries, such as various cancers and especially coronary heart
disease (Wood et al., 2003). High meat intake contributes to a higher than recommended total
and saturated fat and cholesterol intake and may replace sources of other important nutrients
in the diet (Valsta et al., 2005). Yet fat and FAs, whether in adipose tissue or muscle,
contribute significantly to various aspects of meat quality and are central to the nutritional
value of meat (Wood et al., 2008).
One important source of PA is meat, including poultry, beef and game meats (Table 2).
The amount of PA found in meat depends on its source, as well as on the method used to
prepare the food. Fatty cuts of red meat, as well as skin-on poultry, typically contain
relatively high levels of SFA and contain large amounts of PA.
Pork containing an increased proportion of unsaturated FAs and less PA would be better
for consumer health, provided that there is no concomitant increase in fat and cholesterol
content (Kouba and Mourot, 1999). These authors examined the influence of a diet high in
unsaturated fat on lipogenic enzyme activities, lipid content and FA composition of muscles
and adipose tissue in pigs. They concluded that diets with a high content of linoleic acid lead
to the production of pork enriched in this FA without any modification in intramuscular fat
content and PA percentage, thus conserving a totally acceptable level of cholesterol from a
human nutritional point of view.
Table 2. Palmitic acid content of meat and meat products (expressed as percentage
mass-fraction of total fatty acids)
Suet
Lard
Salami
Goose
Pancetta
Mortadella
Ham
Wurstel
Speck
Veal fillet (cooked)
Whole chicken with skin (raw)
Pork loin
Lamb
Rabbit thigh
Pork steak
Pork shoulder
Beef belly
Horse meat

24.02
21.07
5.73-7.55
7.41
5.67-5.99
5.70
3.93-4.93
4.03
3.71
2.89
2.19
2.06
0.58-1.99
1.95
1.90
1.63-1.77
1.66
1.65

50

P. G. Peiretti
Whole turkey with skin (raw)
Whole rabbit
Beef meat
Sheep meat
Goat meat
Ostrich meat
Deer meat

1.54
1.22
0.31-1.14
0.58
0.40
0.16
0.12

Adapted from Italian food composition tables - National Research Institute for Food and Nutrition
(INRAN).

A survey conducted by Enser et al. (1996) illustrated the differences in fat content and
FA profile of muscle between lamb, pork and beef; in particular, PA content, expressed as mg
per 100 g muscle, decreased in the order lamb>beef>pork, while PA, expressed as percentage
of total FAs, decreased in the order beef>pork>lamb.
According to Banskalieva et al. (2000), FA composition of fat depots in goats appears to
be in the range typical for ruminants, with average percentages of PA in goat muscles being
similar to those for other ruminant species. The PA concentration in kidney fat is similar in
goats and lambs, but lower for goat meat than in beef. Hilditch and Williams (1964) noted
that land animals tend to have a relatively constant amount of PA in fat depots.

MILK AND DAIRY PRODUCTS


Another dietary source of PA is dairy. Cow's milk naturally contains SFAs, so both the
milk itself and foods made from the milk typically contain PA.
Foods such as ice cream or butter can prove especially rich sources of FAs, as these foods
contain high levels of dairy fat (Table 3).
Table 3. Palmitic acid content of milk and dairy product (expressed as percentage
mass-fraction of total fatty acids)
Butter
Parmesan cheese
Fontina cheese
Cream
Gelato
Ricotta cheese (cow)
Ricotta cheese (sheep)
Cow's milk (condensed and sweetened)
Sheeps whole milk
Goats whole milk
Cow's whole milk
Yogurt
Semi-skimmed milk
Skimmed milk

20.86
8.04
7.31
5.72
4.10
3.49
2.85
1.97
1.58
1.34
0.92
0.92
0.45
0.05

Adapted from Italian food composition tables - National Research Institute for Food and Nutrition
(INRAN).

Palmitic Acid: Effect of Diet Supplementation and Occurrence

51

The FA composition of milk varies in relation to the influence of several factors related
to the animal and its environment (Palmquist et al., 1993; Perdrix et al., 1996). These include
numerous highly significant factors such as the nature of the food (Grummer, 1991; Coulon et
al., 1994), the type of ration, mode of administration, energy concentration and energy level
of the ration, the physical state of the food and the entire ration, quantity, quality and length
of the fiber, the type, shape and physical treatment of cereals, etc.

FISH AND OTHER FOOD OF ANIMAL ORIGIN


Gruger et al. (1965) studied the FA composition of oils from 21 species of marine fish,
freshwater fish and shellfish, and found that PA content varies widely among species from 9.5
to 33.4% of total FA. Steffen (1997) reported the FA composition of several freshwater fish
lipids and PA content ranged from 10.8 and 24.0% of total FA. zogul and zogul (2007)
found that PA ranged from 15.5 to 20.5% of total FAs and it was the primary SFA,
contributing 53-65% of the total SFA content of lipids in the flesh of eight commercially
important fish species from the Mediterranean, Aegean and Black Seas.
Milinsk et al. (2003) found a percentage of PA that ranged from 21.7 to 22.8% of total
FA in yolk lipids from eggs of White Lohman hens after 16 weeks of feeding with diets
enriched with different oils (canola, flaxseed, soybean or sunflower). Organic eggs are
reported to have higher (Hidalgo et al., 2008) or similar (Cherian et al., 2002) levels of SFAs
as compared with eggs from other production systems. Samman et al. (2009) reported that n-3
polyunsaturated fatty acid (PUFA) egg yolk contained lower percentages of PA compared
with conventional and organic eggs (22.7 vs 25.1 and 25.5% of total FA, respectively). Table
4 reports the PA content of the most common eggs.

EFFECT OF DIET SUPPLEMENTATION


Manipulation of the FA composition of animal tissues has been of great interest in recent
years in order to produce meat with desirable technological and nutritional qualities (Wood et
al., 2003).
Table 4. Palmitic acid content of eggs (expressed as percentage mass-fraction of
total fatty acids)
Hen egg (whole powdered)
Hen egg (yolk)
Duck egg
Goose egg
Turkey egg
Hen egg (whole)

9.96
5.98
3.00
2.98
2.72
1.90

Adapted from Italian food composition tables - National Research Institute for Food and Nutrition
(INRAN).

52

P. G. Peiretti

Scollan et al. (2001) examined the effects of different sources of dietary n-3 PUFA on the
FA composition of muscle and adipose tissue in beef cattle. These authors found that the
proportion of PA was decreased by the linseed diet in both neutral lipids and phospholipids of
muscle. However, fish oil significantly increased the proportion of PA in neutral lipids, while
Mandell et al. (1997) also found that fish meal feeding increased PA content, contrary to the
lack of effect observed by Mills et al. (1992) on feeding fish meal and Ashes et al. (1992)
who fed ruminally-protected fish oil.
Furthermore, there was a lower proportion of PA in phospholipids as a result of feeding
the linseed diet compared with the control diet. However, compared to the control diet, fish
oil did not alter the proportion of PA, but produced lower proportions of stearic and oleic acid
(Scollan et al., 2001).
Nuernberg et al. (2005) examined the effects of feeding system and breed on the content
of the beneficial n-3 PUFA and conjugated linoleic acids in beef muscle, finding that the
percentage of PA decreased significantly with the grass-based system, while there was no
influence of breed on the percentages of PA. Daley et al. (2010) report that grass-finished
cattle are typically lower in total fat as compared to grain-fed contemporaries. Interestingly,
there is no consistent difference in total SFA content between these two feeding regimens.
Those SFAs considered to be more detrimental to serum cholesterol levels, i.e., PA and
myristic acid, were higher in grain-fed beef as compared to grass-fed contemporaries in 60%
of the studies reviewed. Grass-finished meat contains elevated concentrations of stearic acid,
the only SFA with a net neutral impact on serum cholesterol. Thus, grass-finished beef tends
to produce a more favorable SFA composition, although little is known of how grass-finished
beef would ultimately impact serum cholesterol levels in hyper-cholesterolaemic patients as
compared to grain-fed beef (Daley et al., 2010).
Diet is known to affect the FA composition of pig adipose tissue, particularly perirenal
and back fat (Fontanillas et al., 1997; Larick et al., 1992) and to improve the FA profile of
carcass fat in pigs (Morgan et al., 1992; Van Oeckel and Boucqu, 1992). In particular,
dietary oils affected total FA content in pig longissimus muscle (Corino et al., 2002; Kouba
and Mourot, 1999; Van Oeckel et al., 1996). Teye et al. (2006) evaluated the effects of palm
oil, palm kernel oil and soyabean oil, in combination with high and low protein levels, on the
FA composition of the longissimus dorsi muscle in pigs; they found that palm kernel oil
increased the concentrations of PA and other SFAs, while it decreased linoleic acid levels
(Figure 1).
Wood et al. (2004) examined the effects of diet, breed and muscle on FA composition in
pigs and concluded that PA and myristic acid in neutral lipids were higher in Berkshire and
Tamworth than in Duroc and Large White, while the dietary effects on these FAs were small.
Dietary FA modification is considered a viable method of adding value to poultry
products for the health-conscious consumer (Hargis and Vanelswyk, 1993). The profile of
dietary FAs is of importance because it influences the quality of the fat deposited on the
broiler carcass (Figure 2). PA was the predominant SFA in the adipose tissue of birds (Kang
et al., 2001). These authors found that dietary supplementation of palm oil resulted in a
significant increase in PA in the liver and adipose tissue of broilers. According to Rodriguez
et al. (2002), palm oil or mixtures of palm oil, and FAs distilled from palm and calcic soap
are sources of vegetal oils with an FA profile that might replace animal fats without any kind
of negative impact on carcass quality. Consistent parallels between dietary fat and abdominal
fat in broilers have been described by Scaife et al. (1994). The FA pattern of abdominal fat

Palmitic Acid: Effect of Diet Supplementation and Occurrence

53

represents the FA pattern in the dietary fat sources very well and the key FAs which occur at
a somewhat constant rate in broiler adipose tissue are PA, stearic acid, and to some extent,
oleic acid (Zollitsch et al., 1997). Smink et al. (2008) found that the feeding of randomized
instead of native palm oil significantly raised the PA content of breast meat and abdominal fat
and lowered the ratio of unsaturated fatty acids to SFA.

Figure 1. Relationship between dietary palmitic acid percentage and muscle palmitic acid percentage in
pig nutritional trials: Peiretti et al. (2013; ), Teye et al. (2006; ), and Corino et al. (2002; ).

Figure 2. Relationship between dietary palmitic acid percentage and muscle palmitic acid percentage in
poultry nutritional trials: Zollitsch et al. (1997; ), Kang et al. (2001; ), and Smink et al. (2010; ).

Rabbits, like other monogastric animals, are able to directly incorporate dietary FAs into
adipose and intramuscular tissue lipids, thus making it possible to modify the FA profile of

54

P. G. Peiretti

rabbits through the strategic use of unsaturated dietary fat sources (Dalle Zotte, 2002). FA
levels vary a great deal on the basis of the nature of the rabbit diets. The influence of the FA
profile in the diet seems to be more pronounced on the FA composition of adipose tissue than
intramuscular fat (Xiccato, 1999). Peiretti (2012) showed that FA profile was clearly
influenced by diet composition and it was possible to linearly characterize the incorporation
of certain FAs, while PA failed to show a good correlation, both in muscle fat and in perirenal
fat, with its percentage in feed (Figures 3 and 4).

Figure 3. Regressions of palmitic acid percentage in rabbit muscle fat according to its contents in feed.

Lactation responses to dietary fat supplementation have been variable and have been
dependent on fat source, stage of lactation, and dry matter intake (Coppock and Wilks, 1991).
Several studies have examined the effects of PA supplementation on milk FA profile.
Grummer (1991) demonstrated that de novo FA synthesis decreased linearly as
supplementation of dietary fat increased, and that the changes in stearic acid and PA were
dependent on the ratio in the added fat. Steele and Moore (1968) reported reductions in yield
and concentration of short and medium-chain FAs (from butyric to myristic acids) and
dramatic increases in PA with increased dietary intake of PA; the concentration of PA in milk
increased from 38.7% of total FA in controls to 60.7% of total FA in cows supplemented with
PA. Noble et al. (1969) reported similar changes in milk FAs when diet was supplemented
with PA at 10%, they found that short- and medium-chain FAs decreased when compared
with a no-fat control, while milk PA increased from 36.4% of total FA in controls to 49.8% of
total FA in PA-treated cows. Banks et al. (1976) also observed decreases in short- and
medium-chain FAs in milk, with increases observed in concentrations of PA, palmitoleic, and
oleic acids. Using duodenal infusions of 500 g of PA, Enjalbert et al. (2000) reported that
concentrations of PA in milk increased 30% compared with controls. Mosley et al. (2007)
determined the optimum feeding level of a by-product rich in PA (86.6%) on dry matter
intake, milk yield, milk components, and milk FA profile in dairy cattle. They found that milk
FA concentrations were affected by the addition of this by-product. As the intake of PA
increased with the supplemented diets, milk PA concentrations increased. When 1.5 kg/d of
this by-product was consumed, milk PA concentration increased by 50% compared with the

Palmitic Acid: Effect of Diet Supplementation and Occurrence

55

control. This increase was countered by a general decrease in the weight percentage of many
other FAs, with the major difference being an increase in SFAs driven by the linear increase
in PA.

Figure 4. Regressions of palmitic acid percentage in rabbit perirenal fat according to its contents in
feed.

Chouinard et al. (1998) found that dietary Ca salts had no effect on the proportion of PA
in milk fat, but that dietary Ca salts of FA from palm oil typically increased PA (Atwal et al.,
1990; Klusmeyer and Clark, 1991; Elmeddah et al., 1994). Chouinard et al. (1998) reported
two possible explanations for the higher concentration of PA observed for dietary Ca salts of
FA from palm oil. First, palm oil contains a high proportion of PA (>40%) that may be
directly incorporated in milk fat. Second, exogenous PA stimulated synthesis and
incorporation of PA into triacylglycerols by dispersed mammary gland epithelial cells in
vitro, as found by Hansen and Knudsen (1987). Chouinard et al. (1998) reported that PA of
dietary origin might not have been sufficient to compensate for the depression in the de novo
synthesis that occurs, as observed by Grummer (1991), when a fat supplement containing
long-chain FA is fed.
Several researchers (Yu et al., 1977; Mugrditchian et al., 1981; Greene and Selivonchick,
1990; Ng et al., 2000) have reported that fish maintained a constant level of total SFA
regardless of the amount in their diet. Ng et al. (2001; 2004) have shown that palm oil has
some advantages compared to other vegetable oils when used in feeds for warm-water fish
species such as tilapia and catfish. Ng et al. (2007) found that FA composition of Atlantic
salmon fillet total lipid showed close correlations with dietary palm oil inclusion, such that
the concentrations of PA, oleic acid, linoleic acid, SFAs and monounsaturated FAs increased
linearly with increasing dietary oil supplementation.
Bell et al. (2002), studying the FA composition of muscle total lipid in Atlantic salmon
post-smolts fed diets containing increasing levels of crude palm oil, found that the
concentration of PA and other FAs increased linearly with increasing dietary palm oil (Figure
5).

56

P. G. Peiretti

Figure 5. Relationship between dietary palmitic acid percentage and muscle palmitic acid percentage in
Atlantic salmon (Bell et al., 2002; ) and in African catfish (Ng et al., 2003;).

Similarly, Ng et al. (2003) found that FA composition in the muscle lipids of African
catfish were strongly influenced by dietary treatments. In general, the FAs found in high
concentrations in the diet were also the most abundant in muscle and the converse was true of
the least abundant FAs. In particular, the concentration of PA was generally high in muscle
lipid irrespective of diet. PA was present in low concentrations in the sunflower oil and crude
palm kernel oil diets (7.2% and 7.7% of total FA, respectively) and at high concentrations in
the refined, bleached, deodorized palm olein and crude palm oil diets (44.3% and 49.5% of
total FA, respectively), but its concentration in muscle remained somewhat constant at a mean
of about 26%. Nevertheless, relatively higher concentrations of PA were still found in the
muscle of fish fed with diets having higher PA levels. PA was found in muscle lipids at a
relatively uniform concentration of 19.430.2% of total FA despite being fed with diets
containing varied levels of PA at concentrations of 7.249.5% of total FA. The conservation
of PA levels might be because this FA is the major FA in phosphatidylcholine, found in the
Sn-1 position.
The amounts of SFA and unsaturated FAs in egg yolk could be altered by dietary
manipulation (Milinsk et al., 2003). These authors found that there is a decrease in the
percentage of PA and stearic acid in egg yolk produced by hens fed diets enriched with
canola, flaxseed, soybean or sunflower oils in comparison with those of hens fed a control
diet. Kang et al. (2001) found that PA composition of egg yolk was not influenced by hen
diets containing different levels of palm oils and their PA percentage ranged from 21.8 to
24.0 % of total FA.

REFERENCES
Ashes, J.R., Siebert, B.D., Gulati, S.K., Cuthbertson, A.Z. and Scott, T.W. (1992)
Incorporation of n-3 fatty acids of fish oil into tissue and serum lipids of ruminants.
Lipids, 27, 629-631.

Palmitic Acid: Effect of Diet Supplementation and Occurrence

57

Atwal, A.S., Hidiroglou, M., Kramer, J.K.G. and Binns, M.R. (1990). Effects of feeding tocopherol and calcium salts of fatty acids on vitamin E and fatty acid composition of
cows milk. Journal of Dairy Science, 73, 2832-2841.
Banskalieva, V., Sahlu, T. and Goetsch, A.L. (2000). Fatty acid composition of goat muscles
and fat depots: a review. Small Ruminant Research, 37, 255-268.
Banks, W., Clapperton, J.L. and Ferrie, M.E. (1976). Effects of feeding fat to dairy cows
receiving a fat-deficient basal diet. II. Fatty acid composition of the milk fat. Journal of
Dairy Research, 43, 219-227.
Beare-Rogers, J., Dieffenbacher, A. and Holm, J.V. (2001). Lexicon of lipid nutrition.
IUPAC, Pure and Applied Chemistry, 73, 685-744.
Bell, J.G., Henderson, R.J., Tocher, D.R., McGhee, F., Dick, J.R., Porter, A., Smullen, R.P.
and Sargent, J.R. (2002). Substituting fish oil with crude palm oil in the diet of Atlantic
salmon (Salmo salar) affects muscle fatty acid composition and hepatic fatty acid
metabolism. Journal of Nutrition, 132, 222-230.
Carnielli, V.P., Luijendijk, I.H.T., van Beek, R.H.T., Boerma, G.J.M., Degenhart, H.J. and
Sauer, P.J.J. (1995). Effect of dietary triacylglycerol fatty acid positional distribution on
plasma lipid classes and their fatty acid composition in pre-term infants. American
Journal of Clinical Nutrition, 62, 776-781.
Cavani, C., Zucchi, P., Minelli, G., Tolomelli, B., Cabrini, L., and Bergami, R. (1996). Effect
of soybeans on growth performance and body fat composition in rabbits. In: Proceedings
of the 6th World Rabbit Congress, Toulouse, France, pp. 127-133.
Cherian, G., Holsonbake, T. B. and Goeger, M. P. (2002). Fatty acid composition and egg
components of speciality eggs. Poultry Science, 81, 30-33.
Chouinard, P.Y., Girard, V. and Brisson, G.J. (1998). Fatty acid profile and physical
properties of milk fat from cows fed calcium salts of fatty acids with varying
unsaturation. Journal of Dairy Science, 81, 471-481.
Coppock, C. E. and Wilks, D.L. (1991). Supplemental fat in high energy rations for lactating
cows: Effects on intake, digestion, milk yield and composition. Journal of Animal
Science, 69, 3826-3837.
Corino, C., Magni, S., Pagliarini, E., Rossi, R., Pastorelli, G. and Chiesa, L.M. (2002). Effects
of dietary fats on meat quality and sensory characteristics of heavy pig loins. Meat
Science, 60, 1-8.
Coulon, J.B., Agabriel, C., Brunscwig, G., Muller, C. and Bonaiti, B. (1994). Effects of
feeding practices on milk fat concentration for dairy cows. Journal of Dairy Science, 77,
2614-2620.
Crawford, M.A. (1968). Fatty-acid ratios in free-living and domestic animals: possible
implications for atheroma. The Lancet, 291, 1329-1333.
Daley, C.A., Abbott, A., Doyle, P.S., Nader, G.A. and Larson S. (2010). A review of fatty
acid profiles and antioxidant content in grass-fed and grain-fed beef. Nutrition Journal, 9,
10.
De Smet, S., Raes, K. and Demeyer, D. (2004). Meat fatty acid composition as affected by
fatness and genetic factors: a review. Animal Research, 53, 81-98.
Dupont, J., White, P.J., and Feldman, E.B. (1991). Saturated and hydrogenated fats in food in
relation to health. Journal of the American College of Nutrition, 10, 577-592.
Edem, D.O. (2002). Palm oil: biochemical, physiological, nutritional, hematological, and
toxicological aspects: a review. Plant Foods for Human Nutrition, 57, 319-341.

58

P. G. Peiretti

Elmeddah, Y., Doreau, M., Rouel, J. and Chilliard, Y. (1994). Effects of calcium salt
supplementation on dairy cow performances in early lactation. Influence of the nature of
concentrates. Annales De Zootechnie, 43, 341-353.
Enjalbert, F., Nicot, M.C., Bayourthe, C. and Moncoulon, R. 2000. Effects of duodenal
infusions of palmitic, stearic or oleic acids on milk composition and physical properties
of butter. Journal of Dairy Science, 83, 1428-1433.
Enser, M., Hallett, K., Hewett, B., Fursey, G.A.J. and Wood, J. D. (1996). Fatty acid content
and composition of English beef, lamb and pork at retail. Meat Science, 44, 443-458.
Fattore, E. and Fanelli, R. (2013). Palm oil and palmitic acid: a review on cardiovascular
effects and carcinogenicity. International Journal of Food Sciences and Nutrition, 64,
648-659.
Fontanillas, R., Barroeta, A., Baucells, M.D. and Codony, R. (1997). Effect of feeding highly
cis-monounsaturated, trans, or n-3 fats on lipid composition of muscle and adipose tissue
of pigs. Journal of Agricultural and Food Chemistry, 45, 3070-3075.
Frmy, E. (1842). Mmoire sur les produits de la saponification de lhuile de palme. Journal
de Pharmacie et de Chimie, XII, 757.
Greene, D.H.S. and Selivonchick, D.P. (1990). Effects of dietary vegetable, animal and
marine lipids on muscle lipid and hematology of rainbow trout (Oncorhynchus mykiss).
Aquaculture, 89, 165-182.
Gruger, E.H., Nelson, R.W. and Stansby, M.E. (1965). Fatty acid composition of oils from 21
species of marine fish, freshwater fish and shellfish. Journal of the American Oil
Chemistry Society, 41, 662-667.
Grummer, R.R. (1991). Effect of feed on the composition of milk fat. Journal of Dairy
Science, 74, 3244-3257.
Gunstone, F.D., Harwood, J.L. and Dijkstra, A.J. (2012). The Lipid Handbook with Cd-Rom,
3rd Edition.CRC Press, Boca Raton, Florida, p. 1472.
Hargis, P.S. and Vanelswyk, M.E. (1993). Manipulating the fatty acid composition of poultry
meat and eggs for the health conscious consumer. Worlds Poultry Science Journal, 49,
251-264.
Hayes, K.C. and Khosla, P. (1992). Dietary fatty acid thresholds and cholesterolemia. FASEB
Journal, 6, 2600-2607.
Hidalgo, A., Rossi, M., Clerici, F. and Ratti, S. (2008). A market study on the quality
characteristics of eggs from different housing systems. Food Chemistry, 106, 1031-1038.
Hilditch, T.P. and Williams, P.N. (1964). The Chemical Constitution of Natural Fats, 4th
Edition. Chapman and Hall, London, UK, p. 745.
Jonnalagadda, S.S., Egan, S.K., Heimbach, J.T., Harris, S.S. and Kris-Etherton, P.M. (1995).
Fatty acid consumption pattern of Americans: 19871988 USDA nationwide food
consumption survey. Nutrition Research, 15, 1767-1781.
Jones, J.M. (1989). Tropical Oils: Truth and Consequences. Cereal Foods World, 34, 866871.
Kang, K.R., Cherian, G. and Sim, J.S. (2001). Dietary palm oil alters the lipid stability of
polyunsaturated fatty acid-modified poultry products. Poultry Science, 80, 228-234.
Karupaiah, T. and Sundram, K. (2007). Effects of stereospecific positioning of fatty acids in
triacylglycerol structures in native and randomized fats: a review of their nutritional
implications. Nutrition & Metabolism, 4, 16.

Palmitic Acid: Effect of Diet Supplementation and Occurrence

59

Kazala, E.C., Lozeman, F.J., Mir, P.S., Laroche, A., Bailey, D.R.C. and Weselake, R.J.
(1999). Relationship of fatty acid composition to intramuscular fat content in beef from
crossbred Wagyu cattle. Journal of Animal Science, 77, 1717-1725.
Keys, A. (1970). Coronary heart disease in seven countries. Circulation 41, 186-195.
Klusmeyer, T. H., Lynch, G.L., Clark, J.H. and Nelson, D.R.. (1991). Effects of calcium salts
of fatty acids and protein source on ruminal fermentation and nutrient flow to duodenum
of cows. Journal of Dairy Science, 74, 2206-2219.
Kouba, M. and Mourot, J. (1999). Effect of feeding linoleum acid diet on lipogenic enzyme
activities and on the composition of the lipid fraction of the fat and lean tissues in the pig.
Meat Science, 52, 39-45.
Kritchevsky, D. (2000). Overview: dietary fat and atherosclerosis. Asia Pacific Journal of
Clinical Nutrition, 9, 141-145.
Kromhout, D., Keys, A., Aravanis, C., Buzina, R., Fidanza, F., Giampaoli, S., Jansen, A.,
Menotti, A., Nedeljkovic, S. and Pekkarinen M. (1989). Food consumption patterns in the
1960s in seven countries. American Society for Clinical Nutrition, 49, 889-894.
Larick, D. K., Turner, B.E., Shoenherr, W.D., Coffey, M.T. and Pilkington, D.H. (1992).
Volatile compound content and fatty acid composition of pork as influenced by linoleic
acid content of diet. Journal of Animal Science, 70, 1397-1403.
Lindsey, S., Benattar, J., Pronczuk, A. and Hayes, K.C. (1990). Dietary palmitic acid
enhances HDL-cholesterol and LDL receptor mRNA abundance in hamsters.
Experimental Biology and Medicine, 195, 261-269.
Mandell, I.B., Buchanan-Smith, J.G., Holub, B.J. and Campbell, C.P. (1997). Effects of fish
meal in beef cattle diets on growth performance, carcass characteristics and fatty acid
composition of longissimus muscle. Journal of Animal Science, 75, 910-919.
Milinsk, M.C., Murakami, A.E., Gomes, S.T.M., Matsushita, M. and de Souza, N.E. (2003).
Fatty acid profile of egg yolk lipids from hens fed diets rich in n-3 fatty acids. Food
Chemistry, 83, 287-292.
Mills, E.W., Comerford, J.W., Hollender, C., Harpster, H.W., House, B. and Henning, W.R.
(1992). Meat composition and palatability of Holstein and beef steers as influenced by
forage type and protein source. Journal of Animal Science, 70, 2446-2451.
McNamara, D.J. (2010). Palm oil and health: a case of manipulated perception and misuse of
science. Journal of the American College of Nutrition, 29, 240S-244S.
Menotti, A., Keys, A., Aravanis, C., Blackburn, H., Dontas, A., Fidanza, F., Karvonen, M.J.,
Kromhout, D., Nedeljkovic, D., Nissinen, A., Pekkanen, J., Punsar, S., Seccareccia, F.
and Toshima, H. (1989). Seven countries study. First 20-year mortality data in 12 cohorts
of six countries. Annals of Medicine, 21, 175-179.
Morgan, C.A., Noble, R.C., Cocchi, M. and McCartney, R. (1992). Manipulation of the fattyacid composition of pig meat lipids by dietary means. Journal of the Science of Food and
Agriculture, 58, 357-368.
Mosley, S.A., Mosley, E.E., Hatch, B., Szasz, J.I., Corato, A., Zacharias, N., Howes, D. and
McGuire, M.A. (2007). Effect of varying levels of fatty acids from palm oil on feed
intake and milk production in Holstein cows. Journal of Dairy Science, 90, 987-993.
Mu, H. and Hy, C.E. (2004). The digestion of dietary triacylglycerols. Progress in Lipid
Research, 43, 105-133.

60

P. G. Peiretti

Mugrditchian, D.S., Hardy, R.W. and Iwaoka, W.T. (1981). Linseed oil and animal fat as
alternative lipid sources in dry diets for chinook salmon (Oncorhynchus tshawytscha).
Aquaculture, 25, 161-172.
Mukherjee, S. and Mitra, A. (2009). Health Effects of Palm Oil. Journal of Human Ecology,
26, 197-203.
National Research Institute for Food and Nutrition (INRAN). Italian food composition tables.
http://www.inran.it/646/tabelle_di_composizione_degli_alimenti.html Accessed on 6
March 2014.
Ng, T.K.W. (1997). What food for the heart? World Health Forum, 18, 196-198.
Ng, W.K., Tee, M.C. and Boey, P.L. (2000). Evaluation of crude palm oil and refined palm
olein as dietary lipids in pelleted feeds for a tropical bagrid catfish Mystus nemurus
(Cuvier and Valenciennes). Aquaculture Research, 31, 337-347.
Ng, W.K., Lim, P.K. and Sidek, H. (2001). The influence of a dietary lipid source on growth,
muscle fatty acid composition and erythrocyte osmotic fragility of hybrid tilapia. Fish
Physiology and Biochemistry, 25, 301-310.
Ng, W.K., Lim, P.K. and Boey, P.L. (2003). Dietary lipid and palm oil source affects growth,
fatty acid composition and muscle -tocopherol concentration of African catfish, Clarias
gariepinus. Aquaculture, 215, 229-243.
Ng, W.K., Wang, Y., Ketchimenin, P. and Yuen, K.H. (2004). Replacement of dietary fish oil
with palm fatty acid distillate elevates tocopherol and tocotrienol concentrations and
increases oxidative stability in the muscle of African catfish, Clarias gariepinus.
Aquaculture, 233, 423-437.
Ng, W.K., Tocher, D R. and Bell, J.G. (2007). The use of palm oil in aquaculture feeds for
salmonid species. European Journal of Lipid Science and Technology, 109, 394-399.
Noble, R.C., Steele, W. and Moore, J.H. (1969). The effects of dietary palmitic and stearic
acids on milk fat composition in the cow. Journal of Dairy Research, 36, 375-381.
Nuernberg, K., Dannenberger, D., Nuernberg, G., Ender, K., Voigt, J., Scollan, N.D., Wood,
J.D., Nute, G.R. and Richardson, R.I. (2005). Effect of a grass-based and a concentrate
feeding system on meat quality characteristics and fatty acid composition of longissimus
muscle in different cattle breeds. Livestock Production Science, 94, 137-147.
Oguntibeju, O.O., Esterhuyse, A.J. and Truter, E.J. (2009). Red palm oil: nutritional,
physiological and therapeutic roles in improving human wellbeing and quality of life.
British Journal of Biomedical Science, 66, 216-222.
Ong, A.S. and Goh, S.H. (2002). Palm oil: a healthful and cost-effective dietary component.
Food and Nutrition Bulletin, 23, 11-22.
Palmquist, D.L., Beaulieu, A.D. and Barbano, D.M. (1993). Feed and animal factors
influencing milk fat composition. Journal of Dairy Science, 76, 1753-1771.
Patsch, J.R. (1994). Triglyceride-rich lipoproteins and atherosclerosis. Atherosclerosis, 110
Suppl., S23-S26.
Peiretti, P.G. (2012). Effects of dietary fatty acids on lipid traits in the muscle and perirenal
fat of growing rabbits fed mixed diets. Animals, 2, 55-67.
Peiretti, P.G., Mussa, P.P., Forneris, G., Gai, F. and Meineri, G. (2013). Performances and
apparent digestibility of growing pigs fed diets with different fat sources and
supplemented with organic red wine solids. Livestock Research for Rural Development,
24 (10), paper 179.

Palmitic Acid: Effect of Diet Supplementation and Occurrence

61

Perdrix, M.F., Sutter, F. and Wenk, C. (1996). Facteurs de variation de la composition en


acides gras de la matire grasse du lait de vache. Revue Suisse dAgriculture, 28, 71-76.
Redgrave, T.G., Kodali, D.R. and Small, D.M. (1988). The effect of triacyl-sn-glycerol
structure on the metabolism of chylomicrons and triacylglycerol-rich emulsions in the rat.
Journal of Biological Chemistry, 263, 5118-5123.
Rodrguez, M.A., Crespo, N.P., Corts, M., Creus, E. and Medel, P. (2002). Efecto del tipo de
grasa de la dieta en la alimentacin del broiler, con enfasis en los productos derivados del
aceite de palma. Selecciones Avcolas, 44, 693-702.
Samman, S., Kung, F.P., Carter, L.M., Foster, M.J., Ahmad, Z.I., Phuyal, J.L. and Petocz, P.
(2009). Fatty acid composition of certified organic, conventional and omega-3 eggs.
Food Chemistry, 116, 911-914.
Sargent, J.R., Henderson, R.J. and Tocher, D.R. (1989). The lipids. In: Halver, J.E. (Ed.),
Fish Nutrition, (pp.153-218). New York: Academic Press.
Scaife, J.R.. Moyo, J., Galbraith, M., Michie, M. and Campbell, V. (1994). Effect of different
dietary supplemental fats and oils on the tissue fatty acid composition and growth of
female broilers. British Poultry Science, 35, 107-118.
Scollan, N.D., Choi, N.-J., Kurt, E., Fisher, A.V., Mike Enser, M. and Wood J.D. (2001).
Manipulating the fatty acid composition of muscle and adipose tissue in beef cattle.
British Journal of Nutrition, 85, 115-124.
Small, D.M. (1991). The effect of glyceride structure on absorption and metabolism. Annual
Review of Nutrition, 11, 413-434.
Smink, W., Gerrits, W.J.J., Hovenier, R., Geelen, M.J.H., Lobee, H.W.J., Verstegen, M.W.A.
and Beynen, A.C. (2008). Fatty acid digestion and deposition in broiler chickens fed diets
containing either native or randomized palm oil. Poultry Science, 87, 506-513.
Smink, W., Gerrits, W.J.J., Hovenier, R., Geelen, M.J.H., Verstegen, M.W.A. and Beynen,
A.C. (2010). Effect of dietary fat sources on fatty acid deposition and lipid metabolism in
broiler chickens. Poultry Science, 89, 2432-2440.
Steele, W. and Moore, J.H. (1968). The effects of a series of saturated fatty acids in the diet
on milk-fat secretion in the cow. Journal of Dairy Research, 35, 361-369.
Steffens, W. (1997). Effects of variation feeds on nutritive in essential fatty acids in fish value
of freshwater fish for humans. Aquaculture, 151, 97-119.
St. John, L.C., Lunt, D.K. and Smith, S.B., (1991). Fatty acid elongation and desaturation
enzyme activities of bovine liver and subcutaneous adipose tissue microsomes. Journal of
Animal Science, 69 1064-1073.
Storry, J.E., Hall, A.J. and Johnson, V.W. (1968). The effect of increasing amounts of dietary
red palm oil on milk fat secretion in the cow. British Journal of Nutrition, 22, 609-614.
Sundram, K. (1994). Dietary palmitic acid results in lower serum cholesterol than does a
lauric-myristic acid combination in normolipidemic humans. American Journal of
Clinical Nutrition, 54, 841-846.
Sundram, K., Sambanthamurthi, R. and Tan, Y.A. (2003). Palm fruit chemistry and nutrition.
Asia Pacific Journal of Clinical Nutrition, 12, 355-362.
Temme, E., Mensink, R.P. and Hornstra, G. (1996). Comparison of the effects of diets
enriched in lauric, palmitic, or oleic acids on serum lipids and lipoproteins in healthy
women and men. American Journal of Clinical Nutrition, 63, 897-903.

62

P. G. Peiretti

Teye, G.A., Sheard, P.R., Whittington, F.M., Nute, G.R., Stewart, A. and Wood, J.D. (2006)
Influence of dietary oils and protein level on pork quality. 1. Effects on muscle fatty acid
composition, carcass, meat and eating quality. Meat Science, 73, 157-165.
Tholstrup, T., Marckmann, P., Jespersen, J., Vessby, B., Jart, A. and Sandstrom, B. (1994).
Effect on blood lipids, coagulation, and fibrinolysis of a fat high in myristic acid and a fat
high in palmitic acid. American Journal of Clinical Nutrition, 60, 919-925.
Tuten, T., Robinson, K.A. and Sgoutas, D.S. (1993). Discordant results for determinations of
triglycerides in pig sera. Clinical Chemistry, 39, 125-128.
Valsta, L.M., Tapanainen, H. and Mnnisto, S. (2005). Meat fats in nutrition. Meat Science,
70, 525-530.
Van Oeckel, M.J. and Boucque, C.V. (1992). Omega-3-fatty-acids in pig nutrition A
review. Landbouwtijdschrift Revue de lAgriculture, 45, 1177-1192.
Van Oeckel, M.J., Casteels, M., Warnants, N., Van Damme, L. and Boucqu, C.V. (1996).
Omega 3 fatty acid in pigs nutrition: implication for the intrinsic and sensory and quality
of meat. Meat Science, 44, 55-63.
Verschuren, W.M., Jacobs, D.R., Bloemberg, B.P., Kromhout, D., Menotti, A., Aravanis, C.,
Blackburn, H., Buzina, R., Dontas, A.S., Fidanza, F., Karvonen, M.J., Nedelijkovi, S.,
Nissinen, A. and Toshima, H. (1995). Serum total cholesterol and long-term coronary
heart disease mortality in different cultures. Twenty-five-year follow-up of the seven
countries study. Journal of American Medical Association, 274, 131-136.
Wood, J.D., Richardson, R.I., Nute, G.R., Fisher, A.V., Campo, M.M., Kasapidou, E.,
Sheard, P.R. and Enser, M. (2003). Effects of fatty acids on meat quality: a review. Meat
Science, 66, 21-32.
Wood, J. D., Nute, G. R., Richardson, R. I., Whittington, F. M., Southwood, O., Plastow, G.,
Mansbridge, R., da Costa, N. and Chang, K.C. (2004). Effects of breed, diet and muscle
on fat deposition and eating quality in pigs. Meat Science, 67, 651-667.
Wood, J.D., Enser, M., Fisher, A.V., Nute, G.R., Sheard, P.R., Richardson, R.I., Hughes, S.I.
and Whittington, F.M. (2008). Fat deposition, fatty acid composition and meat quality: A
review. Meat Science, 78, 343-358.
World Health Organization (2003). Diet, nutrition and the prevention of chronic diseases. In:
Technical Report Series 916 (pp. 1-104). Geneva, Switzerland: WHO.
Yu, T.C., Sinnhuber, R.O. and Putnam, G.B. (1977). Effect of dietary lipids on fatty acid
composition of body lipid in rainbow trout (Salmo gairdneri). Lipids, 12, 495-499.
Zollitsch, W., Knaus, W., Aichinger, F. and Lettner, F. (1997). Effects of different dietary fat
sources on performance and carcass characteristics of broilers. Animal Feed Science and
Technology, 66, 63-73.

In: Palmitic Acid: Occurrence, Biochemistry and Health Effects ISBN: 978-1-63321-519-1
Editor: Lucas F. Porto
2014 Nova Science Publishers, Inc.

Chapter 4

GENERAL ASPECTS OF PALMITIC ACID


Deusdlia Teixeira de Almeida, Mariana Melo Costa
and Sabrina Feitosa
Federal University of Bahia (UFBA), Brazil

ABSTRACT
Palmitic acid or hexadecanoic acids is the most abundant saturated fatty acid in
human nutrition and represents about 17.6g per day in the United Kington diet. It is the
first fatty acid produced during the lipogenesis. During this process, glucose is converted
to fatty acids, which then react with glycerol to produce triacylglycerols. Palmitic acid
mainly occurs as its ester in triglycerides, especially in palm oil (40-44 %) but also in lard
(20-30 %), dairy products (25-40 %) and cocoa butter (25-27 %). One of the main
applications of palmitic acid in the food industry has been the formulation of
interesterified fats, used as a replacement of trans fats. In breast milk, native lard,
enzyme-directed and randomly chemically interesterified plant fats, palmitic acid
is predominantly esterified to triacylglycerol, center or -position, in native palm oil and
cows milk, it is mainly at the external or -positions. A higher palmitic acid absorption
is obtained with formulas rich in palmitic acid esterified in triacylglycerol sn-2
position, than with those containing palmitic acid predominantly esterified in the sn-1,3
positions. These specific fatty acids distributions in triacylglycerol, determine the
physical properties of the fat, which affects its absorption, metabolism and distribution
into tissues. Many authors claim that a palmitic acid intake may promote increased risk of
hypercholesterolemia, liver disease, type 2 diabetes, insulin resistance and toxicity.
However, more recent investigations on the topic seem to have reconsidered the negative
role of the dietary saturated fatty acids as a risk factor for cardiovascular diseases and
show that not only the type of fat, but also that the triglyceride structure plays a role in
these diseases.

Keywords: Palmitic acid, palm oil, triacylglycerol, saturated fatty acid, dietetic fatty acid

64

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

LIST OF ABBREVIATIONS
3HB 3-Hydroxybutyrate;
AcAc Cetoacetate;
ACC Acetyl-Coenzyme-A Carboxylase;
ACP Acyl Carrier Protein;
ACSL Long-Chain Acyl-CoA Synthetase;
AMP Adenosine Monophosphate;
apo Apolipoproteins;
ARA Arachadonic Acid;
ATP Adenosine Triphosphate;
BC Biotin Carboxylase;
CACT Carnitine/Acylcarnitine Translocase;
CB Cocoa Butter;
CBE Cocoa Butter Equivalents;
CBR Cocoa Butter Replacers;
CBS Cocoa Butter Substitutes;
CoA Coenzyme-A;
CPT Carnitine Palmitoyltransferase;
CT Carboxyltransferase;
DHA Docosahexaenoic Acid;
DPA Docosapentaenoic Acid;
ELOVL Elongation of Very Long-Chain Fatty Acids;
EPA Eicosapentaenoic Acid;
ETC Electron Transport Chain;
FA Fatty Acids;
FAD Flavin Adenine Dinucleotide;
FADH Reduced Flavin Adenine Dinucleotide;
FAS Fatty Acid Synthase;
FABP Fatty Acid-Binding Protein;
FABPpm Plasma Membrane Fatty Acid-Binding Protein;
FAT/CD36 Fatty Acid Translocase/Cluster of Differentiation;
FAO Fatty Acid Oxidation;
FATP Fatty Acid Transport Protein;
GPx Glutathione Peroxidase;
GTP Guanosine-5'-Triphosphate;
HADC 3-Hydroxyacyl-CoA Dehydratase
HDL High Density Lipoprotein;
HMF Human Milk Fat;
HMG-CoA 3-Hydroxy-3-Methyl-Glutaryl Coenzyme A;
IL Interleukins;
JNK Jun Amino Terminal Kinase;
KAR 3-Ketoacyl-CoA Reductase;
LCAD Long-Chain Acyl Coenzyme A Dehydrogenase;
LCFA Long-Chain Fatty Acids;

General Aspects of Palmitic Acid


LDL Low Density Lipoprotein;
LRAT Lecithin Retinol Acyltransferase;
MAPK Mitogen-Activated Protein Kinase;
MCP Monocyte Chemoattractant Protein;
MG- Monoacylglycerols;
mRNA Messenger Ribonucleic Acid;
MUFA Monounsaturated Fatty Acid;
NAD Nicotinamide Adenine Dinucleotide;
NADP Nicotinamide Adenine Dinucleotide Phosphate;
NAFLD Nonalcoholic Fatty Liver Disease;
NASH Nonalcoholic Steatohepatitis;
NFkB Nuclear Factor Kappa B;
OAA Oxaloacetate;
OOL- Dioleoyl- Linoleoyl-Glycerol;
OOO Trioleoyl Glycerol;
OOS Dioleoyl Stearoyl Glycerol;
OPL 1-Oleoyl-2-Palmitoyl-3-Linoleoyl-sn-Glycerol;
OPO 1,3-Dioleoyl-2-Palmitoylglycerol;
PA Palmitic Acid;
Pi Inorganic Phosphate;
PKA Protein Kinase;
PKB Protein Kinase B;
PKC Protein Kinase C;
PLO Palmitoyl-Linoleoyl-Oleoyl-Glycerol;
PLP Dipalmitoyl Linoleoyl Glycerol;
POMF Palm Oil Mid-Fraction;
POO Palmitoyl Dioleoyl Glycerol;
POP Dipalmitoyl Oleoyl Glycerol;
POS Glycerol-1-Palmitate-2-Oleate-3-Stearate;
PPP Tripalmitoil Glycerol;
PUFA Polyunsaturated Fatty Acid;
RBD Bleached and Deodorized;
RDI Recommended Daily Intake;
RNA Ribonucleic Acid;
RNS Reactive Nitrogen Species;
ROS Reactive Oxygen Species;
RP Retinyl Palmitate;
RPB Retinol-Binding Protein;
SCD Stearoyl-CoA Desaturase;
SER Smooth Endoplasmic Reticulum;
SFA Saturated Fatty Acid;
SN Stereospecific Numbering;
SOD Superoxide Dismutase;
SOS Glycerol-1,3-Distearate-2-Oleate;
SPO 1-Stearoyl-2-Palmitoyl-3-Oleoyl Glycerol;
SREBP1c Sterol Regulatory Element-Binding Protein;

65

66

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa


SSS Tri-Saturated Fatty Acids;
SSU Di-Saturated Fatty Acids;
SUS Di-Saturated Fatty Acids;
SUU Diunsaturated;
TAG Triglyceride or Triacylglycerol;
TC Total Cholesterol;
TCA Tricarboxylic Acid Cycle;
TER trans-2,3-Enoyl-CoA Reductase;
TFA trans-Fatty Acid;
TLR Toll-Like Receptors;
TNF- Alpha Tumoral Necrosis Factor;
USU Diunsaturated;
UUU Triunsaturated Fatty Acids;
VLDL Very-Low-Density Lipoprotein.

INTRODUCTION
Fatty acids (FAs) have attracted the attention of the scientific community owing to their
striking fundamental properties which are interesting for science and technology. Fatty acids
are required not only for membrane synthesis, modifications of proteins and carbohydrates,
construction of various structural elements in cells and tissues, production of signaling
compounds, and energy storage, but also for solubilizing a variety of nonpolar and poorly
soluble cellular and extracellular constituents [1, 2].
All fats and oils are esters of glycerol and fatty acids. It is commonly referred to as
triglyceride or triacylglycerol (TAG) because of the glycerol molecule has three hydroxyl
groups where a fatty acid can be attached. All TAGs have the same glycerol units, and then
fatty acids contribute to the different properties. Fatty acids are often categorized into short
chain (up to 6 carbons), medium chain (8 to 12 carbons), or long chain (>12 carbons).
Although hydrocarbon chain length is an important determinant of function, fatty acids are
often classified based on whether or not the fatty acid carbon chain contains no double bonds
(saturated fatty acid SFA), one double bond (monounsaturated fatty acid MUFA), or more
than one double bond (polyunsaturated fatty acid PUFA), as well as the configuration of the
double bonds (cis or trans). In addition, PUFAs are often further classified based on the
position of the first double bond from the fatty acid methyl terminus, creating n-3 and n-6
fatty acids [3, 4].
It is the differences in chain length and saturation status that dictate their performance in
food and cooking, as well as their role in the body and impact on human health and disease
risk [5]. The major types of FAs in the circulation and in the tissues of mammals are the longchain and very-long-chain FAs with many degrees of saturation. These include palmitic acid
(C16:0), palmitoleic acid (C16:1), stearic acid (C18:0), oleic acid (C18:1n-9), linoleic acid
(C18:2n-6) and, particularly in smaller mammals, arachidonic acid (20:4n-6) and
docosahexaenoic acid (22:6n-3) [6].
The variety of fatty acid in common fats and oils is provided in Table 1.

Table 1. Fatty acid profiles (%)a of select animal and vegetable fats and oils

LIPID

SFA

8:0

10:0

12:0

14:0

16:0

18:0

MUFA

18:1

PUFA

18:2

18:3

Avocado oil
Beef tallow
Butterj
Canola oil
Coconut oil
Corn oil
Flaxseed oil
Grapeseed oil
Lard
Olive oil
Palm oil
Palm kernel oil
Rice bran
Salmon oil
Soybean oil

11.9
46.8
53.6
7.6
11.8
12.9
9.0
9.6
36.9
13.7
49.3
81.5
19.7
19.9
15.7

0.0
0.0
5.1
0.0
1.0
0.0
0.0
0.0
0.0
0.0
0.0
3.3
0.0

0.0

0.0
0.0
2.6
0.0
0.8
0.0
0.0
0.0
0.1
0.0
0.0
3.7
0.0

0.0

0.0
0.9
2.7
0.0
6.1
0.0
0.0
0.0
0.2
0.0
0.1
47.1
0.0

0.0

0.0
3.5
7.8
0.0
2.3
0.0
0.1
0.1
1.3
0.0
1.0
16.4
0.7
3.3
0.0

11.3
23.5
22.6
4.4
1.1
10.6
5.1
6.7
22.4
11.2
43.5
8.1
16.9
9.9
10.4

0.7
17.8
10.4
6.8
0.4
1.8
3.4
2.7
12.7
1.9
4.3
2.8
1.6
4.3
4.4

72.6
39.3
22.0
65.1
0.8
27.6
18.5
16.1
42.5
72.4
37.0
11.4
39.3
29.0
22.8

69.9
33.9
20.8
63.5
0.8
27.4
18.3
15.8
38.8
70.7
36.6
11.4
39.1
17.0
22.6

13.9
3.8
3.2
29.0
0.2
54.7
67.9
69.9
10.5
10.4
9.3
1.6
35.0
40.3
57.7

12.9
2.9
2.9
19.6
0.2
53.5
14.3
69.6
9.6
9.7
9.1
1.6
33.4
1.5
51.0

1.0
0.6
0.4
9.4
0.0
1.2
53.4
0.1
1.0
0.7
0.2
0.0
1.6
1.0
7.1

EPAe
DPAf
DHAg
0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.0
31.3
0.0

ARAh

TFAi

0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.0
0.7
0.0

0.0
0.0
0.0
0.4
0.0
0.3
0.1

0.0

0.5

Listed as percent of total fatty acid content, based on 13.6 g fatty acids/tablespoon. Cells without numbers did not have data in United
States Department of Agriculture Nutrient Database; bSFA saturated fatty acid; cMUFA monounsaturated fatty acid; dPUFA
polyunsaturated fatty acid; eEPA eicosapentaenoic acid; fDPA docosapentaenoic acid; gDHA docosahexaenoic acid; hARA
arachadonic acid; iTFA trans-fatty acid; jButter contains 16 % water and therefore the percentages are unable to be directly
compared with percentages of the other fats/oils [5].

68

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

PALMITIC ACID
The word 'palmitic' is French in origin, derived from the word palmitique which refers to
the pith of the palm tree. The palmitic acid (PA) (C16:0, hexadecanoic acid) is one of the
most common saturated fatty acids found in animals and plants with chemical formula
CH3(CH2)14COOH. It is a white solid that melts at 63-64 C, have a boiling point of 351352 C, a mass of 256.42 gmol and a density of 0.853 gcm3 at 62 C. PA is water-insoluble
(7.2 x 10-4 g/100 g) at 20 C and slightly soluble in ethanol and iced acetone, but it is highly
soluble in alcohol, heated acetone and chloroform with 4.76 0.02 pKa value [7]. It is
constituted of carbon atoms linked to each other through single bonds as shown in Figure 1.
The molecules are arranged as dimers through OHO hydrogen bonds. These dimers are
packed in bilayers with terminal methyl groups at both external faces and these layers are
parallel to the crystallographic (100) plane [8].
PA mainly occurs as its ester in TAG, especially in palm oil but also in lard, dairy
products, avocado oil, butter and beef tallow (Table 1). Palmitate is a term for the salts and
esters of palmitic acid. The palmitate anion is the observed form of palmitic acid at basic pH.
Aluminum salts of palmitic acid and naphthenic acid were combined during World War II to
produce napalm. The word "napalm" is derived from the words naphthenic acid and palmitic
acid [9].

Figure 1. Unit cell and molecular structure of palmitic acid [8].

Beyond PA presence in feed, it has utilities ranging from inks antioxidants application,
waterproofing in textile industry, candle manufacture together with paraffin and liquid
crystal, widely used in electronic industry [7].
Palmitic acid can participates in several chemical reactions as other acids of this same
class, which include:
- Neutralization reactions: salts of palmitic acid are formed through this reaction.
The palmitic acid reacts with a hard base, forming a salt of palmitic acid and water. Using

69

General Aspects of Palmitic Acid

sodium hydroxide as a base, for example, sodium palmitate is obtained as shown in Equation
1 (Figure 2) [7].

Figure 2. Equation 1. Neutralization reaction.

Those salts, especially lithium, sodium and potassium, have vast application as ink
constituent, lubricants and insulations that protect against water corrosive action [7].
- Esterification reaction: directly reaction of palmitic acid and a monohydric alcohol
obtaining an ester (Figure 3. Equation 2), or by a reaction between a salt of palmitic acid and
a haloalkane (Figure 3. Equation 3) [7].

(2)

(3)
Figure 3. Equation 2 and 3. Esterification reactions.

RETYNIL PALMITATE
The two most abundant retinoid forms that are present in the diet are retinol and retinyl
esters - a fatty acyl group is esterified to the hydroxyl terminus of retinol [10]. Dietary retinol
is taken up directly by mucosal cells. Nevertheless, dietary retinyl esters are cleaved in the
intestine by the pancreatic triglyceride lipase and intestinal brush border enzyme,
phospholipase B [11]. The free retinol taken up by the enterocyte is complexed with cellular
retinol-binding protein type 2 and the complex serves as a substrate for reesterification of the
retinol by the enzyme lecithin retinol acyltransferase (LRAT). The resulted retynyl ester are
incorporated with other neutral lipid esters (i.e., triacylglycerols and cholesteryl esters) into
chylomicrons and absorbed via the lymphatics [11, 12]. In the vascular compartment, much of
the chylomicron triacylglycerol is hydrolyzed by lipoprotein lipase in extrahepatic tissues,
resulting in the production of a chylomicron remnant that contains most of the newly
absorbed retinyl esters [13]. Under conditions of adequate vitamin A nutrition, the liver is the
main site of vitamin A storage, with more than 95 % of the total neutral retinoid being present

70

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

as retinyl esters, predominately retinyl palmitate and stearate [12, 14, 15]. According to Ihara
et al. [12] the explanation for the use of those fatty acids would be that the retinyl esters
together cholesterol present in its particle would be release into the liver cells via LDL
receptor wherein saturated fatty acid regulate it pathway largely mRNA level. Once the cells
has met it requirements for retinol, saturated fatty acids inhibit receptor activity, so that the
receptor is no longer able to internalize retynil esters. In the response to cellular requirements
the liver release retinol in forms of a retinol-binding protein (RPB4), target cells, a cell
surface receptor for retinol RBP4 remove retinol from RPB4.
Esterification techniques using palmitic acid have yielded more stable esters in the form
of retinyl palmitate which has been used successfully as a supplement as well as a way to
fortify numerous foods, including vegetable oil, rice, monosodium glutamate, cereal flours
and sugar [review 16, 17]. This application is due to its high stability in relation to vitamin A
and its low cost [16]. The oil matrix protects against the oxidation of vitamin A during
storage, improves stability of the retinol and facilitates the vitamins absorption by the body
[18].
The advantages of oil fortified with retinyl palmitate have historically been utilized by
food aid programs, where a daily intake of 16 g of oil provided approximately 50 % of the
recommended daily intake (RDI) of an adult male [19]. Surman et al. [20] reports that retinyl
palmitate it also can be associated with toxicities at high doses. The precise human dose
required to ensure efficacy without toxicity remains a point of controversy. Also, retinyl
palmitate (RP) is widely used in pharmaceutical and cosmetics products to improve the skin
elasticity [21, 22].

OCCURRENCE OF PALMITIC ACID


Palm Oil
Elaeis guineensis palm tree fruits are yellow or orange ovoid little coconuts, with
variable-size and seeds that produce two types of oil: the palm oil from fruit mesocarp and
palm kernel oil from seed [23, 24]. The palm oil has become the major edible oil in world,
markets accounting for 57 % worldwide vegetable oils exports and approximately 62 million
tons of palm oil is projected to 2015 against 45.5 million tons in 2010 [25]. The major palm
oil producers are Malaysia, Indonesia and Nigeria [26, 27].
Palm oil and palm kernel oil differentiate to physico-chemical properties as their intended
applications [28]. Palm kernel oil is minority and produces 50 % of oil with dark coloration
and high contents of lauric and myristic FAs. It is used in manufacturing of confectionery
products, ice cream, soaps, detergents, among others [23; 29; 30].
Palm oil contains 6-10 % of tri-saturated fatty acids (SSS), especially tripalmitoil
glycerol (PPP). Saturated fraction corresponds to 1-5 % of lauric and myristic acid and 17-23
% to palmitic acid (C16:0) in sn-2 position, while the myristic (C14:0) and the stearic (C18:0)
in 1 and 3 position [23]. The unsaturated fraction represents 44-50 % of di-unsaturated fatty
acids (SSU or SUS): dipalmitoyl oleoyl glycerol (POP) and dipalmitoyl linoleoyl glycerol
(PLP); 38-42 % diunsaturated (SUU or USU): POO and PLO, palmitoyl dioleoyl glycerol and

General Aspects of Palmitic Acid

71

palmitoyl-linoleoyl-oleoyl-glycerol, respectively; and 5-8 % of triunsaturated fatty acids


(UUU), especially trioleoyl glycerol (OOO) and dioleoyl- linoleoyl-glycerol (OOL) [23, 30].
Palm oil physico-chemical properties allow it to be the most widely fractioned oil
(Table 2). Fractioning involves physical or chemical refine applying high temperatures,
desodorisation and deacidification of oil under vacuum, in both cases. Physical
deacidification accurs at 250-270 C under vacuum up to 3-5 Torr, whereas chemical uses
220-240 C. The high temperatures and vacuum are necessary to remove undesirable
compounds as traces of metals, free fatty acids, oxidation and decomposition products.
Nevertheless, those procedures also remove some tocopherols and tocotrienols, and all
carotenoids presented in the oil [30, 31, 32, 33, 34].
Two types of oil with different physical and chemical properties are obtained from the
fractioning process: the olein (65-70 %), liquid phase, melting point at 18-20 C; and stearin
(30-35 %), solid, melting point at 48-50 C. Refining industry obtain the others fractions from
those two, applying in the manufacture of several products [23; 34; 35].
Malaysia Palm Oil Research Institute patented the physical refine method that involves
degumming and bleaching, followed by clarification and desodorisation by molecular
distillation using lower temperatures (< 170 C) and pressure (100 mTorr), obtaining an oil
comparable to the RBD (bleached and deodorized) with less than 0.1 % of acidity and
impurities, but with vitamin E retention and 80 % of carotenoids, called carotino [28, 29, 33,
34]. It is also observed the use of crude palm oil in Bahia (Brazil) and Africa, being part of
many culinary dishes [36]. The crude palm oil is reddish because it contains a high amount of
-caroten (500-1000 mgkg1) [37].
Palm oil liquid phase, olein (65-70 %), is liquid at room temperature and can present
TGAs precipitation by higher melting point in case of cold storage. It is extensively used as
cooking oil and blended with other oils. The stearin is a co-product of olein that is used in
vegetable fats manufacturing as margarines, pastas and bakery products [31]. Palm olein and
super olein have higher contents of linoleic and oleic FAs and lower content of palmitic acid
than the stearin fraction [23]. Whereas the middle fraction present properties between olein
and stearin, with 60 % of palmitic acid and 40 % of oleic acid, being used as cocoa butter
substitute [23; 35]. Palm oil composition shows that most triglycerides are esterified in sn-2
position with unsaturated fatty acids (> 58.25 % of oleic acid and > 18.41 % of linoleic acid)
and high proportion of palmitic acid [38].
Edem [23] points out some characteristics that made palm oil an important food industry
ingredient. These characteristic include: (a) a high solid glyceride content giving required
consistency without hydrogenation, (b) resistance to oxidation and therefore, long shelf life
(c) high melting point triglycerides together with relatively low solids content at 10 C, which
is helpful in formulation of products with a wide plastic range, that are suitable for hot
climates and some industrial applications, (d) a competitive price, (e) use in limited quantities
in margarine specifying a high polyunsaturated fatty acid (PUFA) level because of its linoleic
acid content (1011 %), (f) relatively slow melting properties because of the wide plastic
range and (g) slow crystallization properties capable of leading to structural hardness and a
tendency for recrystallization.

72

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa


Table 2. Fatty acid composition of palm oil and its fractions

Fatty acid
12:0
14:0
16:0
16:1
18:0
18:1
18:2
18:3
20:0
Saturates
Monounsaturates
Polyunsaturates
[23].

Red palm oil


00.2
0.81.3
43.146.3
Trace0.3
4.05.5
36.740.8
9.411.9
0.10.4
0.10.4
50.2
39.2
10.5

Palm olein
0.10.2
0.91.0
39.540.8
Trace0.2
3.94.4
42.743.9
10.611.4
00.4
0.10.3
45.8
42.5
11.6

Super olein
0.4
1.4
31.5

3.2
49.2
13.7
0.3
0.4
36.6
49.2
14.0

Super stearin
0.10.2
1.01.3
46.568.9
Trace0.2
4.45.5
19.938.4
4.19.3
0.10.2
0.10.3
52.176.2
19.938.6
4.29.5

Butter
Butter or buttermilk is an important edible fat in northern Europe, North America and
Brazil, being about the third product of worlds milk production. It is a yellow-to-white solid
and an emulsion of fat globules, water and inorganic salts produced by churning the cream
from cows milk. Butter has high energy (~715 Kcal/100 g), cholesterol (~215 mg/100 g) and
a major content of FAs, respectively: palmitic (25-32 %, C16:0), oleic (22-29 %, C18:1),
myristic (C14:0) and stearic (C18:0) about same proportions (8-13 %), linoleic (C18:2) and
lauric (C12:0) with less than 4.5 %. In despite of high SFAs contents, MUFAs are higher than
PUFAs in buttermilks [39, 40]. Verardo et al. [40] determined the fatty acid composition of
different samples of butter and the samples manufactured by a traditional method showed
higher levels of MUFAs and PUFAs compared with industrial samples. Palmitic acid
presented the higher fatty acids contents, 29.8 to 31.1 % and 30.3 to 33.6 % from traditional
and industrial process, respectively.

Cocoa Butter
The generic name of cocoa is Theobroma belonging to the family of Sterculiaceae, also
called Food of God. It contains about 3050 beans, covered with pulp. About 500 years
ago, cocoa beans were originated from Latin America and within a few years it was spread to
Europe [41, 42].
Cocoa butter (CB) is a highly valued ingredient primarily used in the confectionery
industry due to its specific physical and chemical properties. CB is solid at room temperature
(below 25 C), and liquid at body temperature (~37 C) [43]. Furthermore, the predominant
presence of symmetrical TAG, about 90 % of the TAG species in CB, is mainly responsible
for the functionality of this fat [44]. The major FAs of cocoa butter are palmitic acid (C16)
2533.7 %, stearic acid (C18:0) 33.740.2 %, oleic acid (C18:1) 26.335 % and linoleic acid

General Aspects of Palmitic Acid

73

(C18:2) 1.73 % which contribute about 98 % of the total fatty acid [45]. Regarding the
palmitic acid composition of natural cocoa butter produced from various countries ranged
from 24.1 to 27.9 % [43]. CB fat contains significantly higher amount of saturated acid
leadings to triglycerides of glycerol-1,3-dipalmitate-2-oleate (POP), glycerol-1-palmitate-2oleate-3-stearate (POS) and glycerol-1,3-distearate-2-oleate (SOS). Among these three TAGs,
POS is the major leading triacylglycerol component present in cocoa butter with range 42.5
46.4 % yield followed by SOS (27.833.0 %) and POP (18.922.6 %). Therefore, palmitic
acid occupies mostly the sn-1 position in cocoa butter [45].

Avocado Oil
Avocado (Persea americana Mill) is an important tropical fruit and a good source of
lipophilic phytochemicals such as monounsaturated fatty acids, carotenoids, vitamin E and
sterols [46]. It has several cultivars that present great variation on time of fruit production and
oil content in the pulp. Studies have indicated that the avocado oil is similar to olive oil and
can be used in cosmetics and also for human consumption [47, 48]. New Zealand, Mexico,
Chile United States and South America are among the main avocado oil producers. Avocado
oil has the advantage that can be obtained from the fruit by means of a cold extraction
methods, which is an easy and low technology that allow maintain in the oil significant
amounts of the bioactive phytochemicals present in the fruit [47].
In comparison to other vegetable sources, avocado oil is characterized by its contents of
palmitic (C16:0), linoleic (C18:2), palmitoleic (16:1) and alpha-linolenic (18:3) FAs that are
13.5, 12.6, 3.26 and 1.0 % of total fatty acids, respectively. Stearic (18:0), tridecanoic (13:0),
tetradecanoic (14:0), cis-10-heptadecenoic (17:1) and cis-13-16-eicosenoic (20:2) FAs are
present in trace amounts [49]. Ozdemir and Topuz [50] showed in Fuerte and Hass avocado
varieties present a reduction of palmitc acid according to fruit ripening, with variations of
22.4-12 % to Fuerte variety and 23.3-16.8 % to Hass variety. Yanty et al. [51] studying three
Malaysia avocados varieties, found oleic acid as the major fatty acid (43.6551.22 %)
followed by palmitic (26.4130.37 %) and linoleic (12.7517.45 %) FAs. Oils of avocado
fruits are generally found to have extremely low amounts of stearic acid (0.271.56 %).

Beef Tallow
Beef tallow is one residual material from slaughterhouses which main destination is the
soap industry, however because of its high melting point (45 C) and low level of
polyunsaturated fatty acids (< 3 %) [52, 53] beef tallow is considered as a less valuable fat
not suitable for direct human consumption [54]. From the nutritional point of view, vegetable
oils are preferred over animal fats, because contain a high proportion of saturated fatty acids
and low proportion of polyunsaturated fatty acids. Regarding fatty acids composition, tallow
has about 29 % palmitic acid, 25-37 % stearic acid and 23-31 % oleic acid. Thus, saturated
fatty acid content is responsible for over 50 % of total fatty acids in the beef tallow [52, 53].
The higher stearic and palmitic acid content of beef tallow are accounted for the unique
properties of high melting point and high viscosity [55]. In relation to TAG, beef tallow and
other bovine adipose tissues have nearly 50% of the fatty acids in the sn-2 position, which are

74

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

oleic acid (unsaturated fatty acid), palmitic and stearic acids. Nevertheless, oleic acids are the
most fatty acids in the sn-1,3 positions [52, 56].

Lard
Animal fats such as lard and tallow have long been recognized as raw material for food
and industrial applications. Lard has exceptional properties compared to other vegetable oils
such as firmness and special flavor values. Nevertheless, lard has lost its significance to
numerous substitutes such as hydrogenated cottonseed and soybean oil due to its negative
nutritional values such as low digestibility, high calories and high content of saturated fatty
acids. Nonetheless, lard remains a major ally in the meat product industry due to its positive
contributions in flavor and texture [57, 58].
Lard contains about 28.4 % palmitic acid (C16:0), 21 6 % stearic acid (C18:0), 31.4 %
oleic acid (C18: 1) 11.1 % linoleic acid (C18: 2). It is unique among animal depot fats,
because it has a strong predominance of saturated fatty acids in the sn-2 position [52]. In lard,
C16:0 is located exclusively at the sn-2 position, with an unsaturated fatty acid at sn-3 but the
fatty acid occupying the sn-1 position is highly variable, as in SPO, OPL and OPO TAGs
(species dominants in lard) [56].

Milk
Bovine milk and dairy products have long traditions in human nutrition. Its fat fractions
are widely used in a variety of food products such as liquid milk, cream, butter, cheese and
ice cream due to many favorable physical, chemical and nutritional properties of milk fat
[59].
Milk contains, in average, about 33 g total lipid/L, being 95-98 % of triacylglycerols
composed of fatty acids of different length and saturation, less than 0.5 % of cholesterol,
about 1 % of phospholipids and less than 0.5 % of free fatty acids. SFA represent more than
half of total milk fat, about 19 g/L especially lauric (C12:0), myristic (C14:0) and palmitic
(C16:0). Furthermore it is rich in oleic acid (about 25 %), however, a relatively poor source of
polyunsaturated fatty acids as linoleic (C18:2) and alpha linolenic (C18:3), with contents in
the order of 3 % and 2 %, respectively [59; 60; 61].
In relation to TAG molecules, the most probable sn-position of the main fatty acids in
milk fat are: sn-position 1 C16:0, C18:0 and C18:1 with 44.1 %, 54.0 % and 37.3%
respectively; sn-position 2 C12:0 (62.9 %), C14:0 (65.6 %) and C16:0 (45.4 %); and snposition 3 98.1 % of C4:0, 93.0 % of C6:0, 34.5 % of C8:0 and 41.5 % of C18:1. Therefore,
the most probable sn-position of palmitic acid in TAG molecules of milk are in sn-1 and sn-2
[61].

Human Milk
The human mammary gland has evolved with unusual pathways for acylation of fatty
acids into triglycerides for secretion in milk, and major portion of milk fat is comprised by

General Aspects of Palmitic Acid

75

these molecules that represent 98 % of total fat [61, 62]. The average fat content of human
milk is about 3.83.9 g/100 ml, but it varies widely [61]. The predominant fraction are
saturated fatty acids, followed by a relatively high proportion of monounsaturated fatty acids
such as oleic acid (18:1n 9) [59]. Palmitic acid is the qualitatively and quantitatively major
SFA in human milk and constitutes approximately one fourth of this with a concentration
highly conserved, regardless of ethnic origin or the nutritional status of the woman [62]. PA
comprises 17 % to 25 % of the total FAs [63] and it is an important source of energy thereby
contributing 1012 % of breast-fed infants' dietary energy intake [64].
The stereospecific numbering (sn) designates the location of fatty acids within the
triglyceride molecule. If the glycerol is drawn with the first and third hydroxyl groups to the
right and the second to the left, the first carbon is termed sn-1; the second, sn-2, and the third
sn-3 [61]. Most of the 16:0 in human milk is located in the sn-2 position of the triacylglycerol
molecules (70 % to 75 %) [63], in contrast to cows milk and vegetable oils which have 40 %
and 5 % to 20 %, respectively, of the 16:0 in the sn-2 position [62]. The major unsaturated
fatty acid in human milk is oleic acid (18:1n-9) and this is mostly esterified at the triglyceride
sn-1,3 (outer) positions, with the result that triglycerides with the structure 18:1n-916:0
18:1n-9 (1,3-dioleoyl-2-palmitoylglycerol, OPO) are a major triglyceride species in human
milk and represent an estimated 11.8 % of the total triglyceride species [65]. Since lipolytic
enzymes will cleave the FA in sn-1 and sn-3-positions, human milk palmitic acid will appear
primarily in the remaining monoglyceride which has a higher polar than free palmitic acid
[61]. Hence, most of the 16:0 is absorbed as the sn-2 and this structure is preserved through
and beyond the intestinal wall.

SPECIAL OILS
Pequi Tree
Pequi tree (Caryocar brasiliensis Camb.) is a member of the Central and South American
family Caryocaraceae [66, 67]. It stands out by high occurrence in Brazilian Cerrado and
extensive period of fruit production, which can be collected from September to February in
Cerrado of Goias (Brazil) [68]. It is a Brazilian oleaginous fruit, rich in A, E, C and B2
vitamins in both edible parts: pulp and kernel [69, 70].
The oil is extracted by rendering the mesocarp and kernel of pequi fruit and the oil is
generally used to cook rice with the objective of adding specic avors and a light-yellow
color to the nal product [71]. The fruit is rich in -carotene and selenium. It is even used to
produce fermented liquor. Pequi has a considerable economic importance in some parts of
Brazil and has a substantial ecological impact on the country. In relation to fatty acids
composition, kernel and pulp present high content of palmitic acid (35.17 % and 43.76 %)
and oleic (43.59 % and 55.87 %), respectively. The TAG composition of pequi oil is also
relatively simple with trioleoyl glycerol (OOO, 56 gkg1), palmitoyl dioleoyl glycerol (POO,
466 gkg1) and dipalmitoyl oleoyl glycerol (POP, 452 gkg1) comprising 974 gkg1 of the
total. Dioleoyl stearoyl glycerol (OOS) was found in small amounts (5.2 gkg1). The
relatively simple composition of pequi oil may be of interest for selected applications.

76

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

It appears that is has some promise as a less expensive chocolate substitute upon fractionation
[23, 38].

Pili Nut Oil


The pili (Canarium ovatum) is a tropical tree, native to the Philippines, with about 600
species in the Burseraceae family. The pili fruit is a drupe with 4 to 7 cm long and 2.3 to 3.8
cm in diameter and weighs 15.7 to 45.7 g which consists of a pulp (68 % by weight), a shell
(25 % by weight) and a seed (7 % by weight) [72]. The fibrous pulp is edible and is usually
consumed as an appetizer or dessert. Already the pili pulp oil is comparable to coconut oil
and can be used for cooking. It can also be used as fuel for lighting, and for the manufacture
of soaps, perfumes and other cosmetic products. Nevertheless, the kernel or known as pili nut
is the most important part of the pili fruit, with a taste comparable to that of walnut and
almond, has been used in chocolates, baked goods and ice cream. It contains about 7075 %
oil which can be used for domestic and industrial purposes [73].
In the pili nut oil the fatty acids saturates (palmitic and stearic FAs) account for 33.3 and
10.9 %, respectively and the oleic acid represents 44.7 % of pili nut oil. Furthermore, this oil
is very low in polyunsaturated fatty acids (18:2 and 18:3), with the combination of linoleic
and linolenic less than 11 % [72].
With regard to triacylglycerol (TAG) composition in the roasted pili nut oil, 54.3 % is of
1-palmitoyl-2-oleyl-3-oleyl-sn-glycerol (POO), 13 % of 1,3-dipalmitoyl-2-oleoyl-glycerol
(POP), 8.2 % of triolein (OOO) and 6.1 % of 1-palmitoyl-2-oleyl-3-stearoyl glycerol (POS),
whereas unrosted pili nut oil has about 61.1 % of 1-palmitoyl-2-oleyl-3-oleyl-sn-glycerol
(POO), 12.5 % of 1,3-dipalmitoyl-2-oleoyl-glycerol (POP), 7.7 % of Triolein (OOO) and 6.2
% of 1-palmitoyl-2-oleyl-3-stearoyl glycerol (POS) respectively. Thus, the pili nut oil not
contain the trisaturated TAGs [74].

Occurrence of Palmitic Acid Interesterified Foods


Oils and fats industry has been looking for alternatives to replace the called hydrogenated
fats which are rich in trans fatty acids, because of the human health implications [75, 76, 77].
One of the methods that have been applied in this way is the interesterification of fats. In this
reaction, the fatty acids remain unchanged occurring a redistribution of them in triglyceride
molecule, resulting in a modification of triglyceride composition. The final characteristic is
completely determined by initial fatty acids composition of raw material. Intesterification can
be carried out chemically or enzimatically. A chemical, such as sodium methoxide, is used as
a catalyst in chemical interesterification which produces complete positional randomization of
the acyl group in the TAG. On the other hand, enzymatic interesterification uses
microbiological lipases as catalyst [78; 79].
Commonly used interesteried fats, which provide suitable functionality for the food
industry, include fats that are rich in the long-chain SFA, palmitic acid (16:0) and stearic acid
(18:0) [77]. Chemical and enzymatic interesterification has been specially used in the
formulation of margarines and shortenings to provide products with no TFA but that still
maintain physical properties, taste, and stability [80].

General Aspects of Palmitic Acid

77

Shortenings and Margarines


For preparation of various food products industries take into account the polymorphism
of fats, defined as the ability of a chemical compound to form different crystalline or liquid
crystalline structures and three forms of , ' and are typical polymorphic forms of fat [81].
Polymorph ', the meta-stable form, is used in margarine and shortening because of its
optimal crystal morphology and fat crystal networks which give rise to optimal rheological
and texture properties [82]. Because of the high content of palmitic acid, palm oil is '
tending, and hence, palm oil plays the role of a ' in margarines and shortenings. It was
related that margarines at 15 C with palmitic acid below 11 % are in form, while those with
17 % and above in ' crystal form [80]. This phenomenon leads to the production of smooth,
continuous and homogenous products.

Cocoa Butter Alternatives


Food industries are looking for alternative fats to cocoa butter (CB) from natural matrices
that are denoted as cocoa butter replacers (CBRs), cocoa butter equivalents (CBEs) and cocoa
butter substitutes (CBSs) fat [41; 83]. CBRs are dened as non-lauric fats that could replace
cocoa butter either partially or completely in the chocolate or other food products. On the
other hand, a cocoa butter equivalent (CBE) is a type of fat that has a very similar chemical
composition, but its triglycerides derive from other source than cocoa beans, such as palm
kernel oil, palm oil, mango seed fat, kokum butter, sal fat, shea butter, illip butter, soya oil,
rape seed oil, cotton oil, ground nut oil and coconut oil [43].
Replacing the cocoa butter either partially or wholly with other natural fats has been
investigated due to the technological and economic advantages. CBE contains approximately
40 % 1-palmito, 2-olein, 3-sterin glycerol (POS), 27 % of 1,3distearin-monooleate glycerol
(SOS) and 21 % of 1,3 dipalmitin-2-monooleato glycerol (POP) and minor amounts of other
triglycerides. A suitable raw material for the production of CBE in terms of cost, availability
and composition is palm oil mid-fraction (POMF), which is obtained by double fractionation
of palm oil. POMF contains approximately 73 % POP, 13 % POS, 2 % SOS and 12 % of
other triglycerides [84].

Palmitic Acid in Infant Formulas


Developing infant milk fat similar to human milk fat (HMF) is of great interest and a
challenge to food processors. Infant formulas have been designed to provide infants with the
required for optimal growth and development [62; 85; 86]. The quantity of fat in its formulas
provides an infant with 4050 % of its total daily energy intake [87]. Complex mixtures of
oils that contain modest levels of long-chain saturated fatty acid can be used in developing
formulas with fatty acid profiles closer to that of human milk [86; 87] but not the
triacylglycerol structure [62; 88; 89]. Simple mixtures of unsaturated vegetable oils (e.g., corn
or soy oil) and fats that contain a predominance of lauric acid or shorter saturated fatty acids
(e.g., coconut oil and palm kernel) are common fat blends in some formulas [90]. Several
types of enzymatic reactions appear in the literature to synthesize structured TAGs rich in PA

78

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

at position 2 and in other fatty acids at positions 1 and 3 (oleic acid, caprylic acid, etc.). These
fat compositions can be obtained by subjecting fatty mixtures comprising glycerides
consisting substantially of more saturated 2-palmitoyl glycerides to a rearrangement catalyst,
such as a lipase, which is regiospecific in activity in the 1- and 3-positions of the glycerides.
Under the influence of the catalyst, unsaturated fatty acid residues may be introduced into the
1- and 3-positions of the 2-palmitoyl glycerides by exchange with unsaturated free fatty acids
or their alkyl esters.
Unfortunately, such formulas have resulted in poor absorption of fats and minerals,
particularly when studied in infants during the first few weeks of life [62, 91, 92]. This is
because PA is present in the sn-2 position (human breast milk, lard native, enzyme-directed
and randomly chemically interesterified fats plant) compared with the sn-1 and sn-3 positions
(bovine milk, randomly chemically interesterified lard or crude palm oil). It was found the
relative absorption of palmitic acid and full fat was linearly related to the proportion of
palmitic acid in the sn-2 position of the TAG in human infants. Pancreatic lipase selectively
hydrolyses the fatty acids at the sn-1 and sn-3 positions, yielding free fatty acids and
monoacylglycerols (MGs). The TAG sn-2 position is absorbed more efficiently than free
palmitic acid and it is conserved through the digestion, absorption and chylomicron TAG
synthesis [63, 91].
Nonetheless, after digestion, the free PA solidify in the intestine because of their high
melting temperature, creating insoluble and indigestible complexes with dietary minerals (eg,
calcium), and causing hard stools [87]. Quinlan et al. [93] were able to relate stool hardness to
stool composition. They concluded that differences in the triacylglycerol palmitate content of
formula and breast milk resulted in more calcium soap formation in formula- fed infants and
thus in harder stools. HMF containing palmitic acid at the sn-2 position yields 2-MG during
digestion which does not lead to the formation of calcium soaps. Consequently, both calcium
and 2-MG become bioavailable for the infant [94]. Yaron et al. [63] studying two infant
formulas demonstrates that -palmitate may affect the intestinal microbiota composition
during the first weeks of life by increasing Lactobacillus and Bifidobacteria abundance in the
stool, and thus may provide beneficial effects for the health and well-being of formula-fed
infants. They concluded that the effects of the infant formulas of the gut microflora are due to
lipid estructure. It is therefore important to synthesize TAGs with a composition and
distribution of fatty acids similar to those of human milk. In particular there is considerable
interest in the synthesis of 1,3-diolein-2-palmitin (OPO), which is the most abundant TAG in
human milk.

BIOCHEMISTRY OF PALMITIC ACID


Lipogenesis
Fats used by or stored in animal tissues come from two sources: enzymatic synthesis and
diet. When a cell or organism has more than enough metabolic fuel available to meet its
energetic needs, the excess is generally converted to FAs and stored as lipids such as TAGs
[95]. Insulin stimulates fatty acid synthesis in adipose tissue, liver and lactating mammary
glands along with formation and storage of TAG in adipose tissue and liver [96, 97].

General Aspects of Palmitic Acid

79

In glycolysis, glucose (C6H12O6) is converted to two molecules of pyruvate [97]. The


pyruvate dehydrogenase complex contributes to transforming pyruvate into acetyl-CoA by a
process called pyruvate decarboxylation. When energy needs in a cell are not high, citrate, the
condensation product of oxaloacetate and acetyl-CoA in the glicolysis, builds up in the
mitochondrial matrix.
Nonetheless, mitochondria do not contain an acetyl CoA transporter, therefore a shuttle
system, called the citrate shuttle, is required to move the C2 units across the membrane citrate
transport out of the mitochondria provides a mechanism to stimulate fatty acid synthesis in
the cytosol where acetil Co-A is cleaved back to oxaloacetate and acetyl-CoA by the ATPcitrate lyase (Figure 4) [98, 99].
The oxaloacetate (OAA) is converted to malate by cytosolic malate dehydrogenase The
malate is then either transported to the mitochondrial matrix where it is reduced by
mitochondrial malate dehydrogenase or oxidized and decarboxylated by malic enzyme in the
cytosol to form pyruvate, NADPH and CO2 [100]. The production of cytosolic NADPH by
malic enzyme provides additional reducing equivalents for fatty acid synthesis and
supplements the NADPH generated by the pentose phosphate pathway. The pyruvate formed
by malic enzyme is transported back into the mitochondrial matrix where it is carboxylated by
pyruvate carboxylase to form oxaloacetate. Note that while malic enzyme generates NADPH
for fatty acid synthesis, the pyruvate carboxylase reaction consumes an ATP in the matrix to
generate OAA. The liver cells can still run the glycolytic pathway as the NADH/NAD+ ratio
is low in the cytoplasm while NADPH/NADP+ ratio is high [98].
The acetyl-CoA can be converted to malonyl-CoA via the action of acetyl-CoA
carboxylase (ACC; biotin-dependent enzyme) [101]. Two isoforms have been identified,
ACC1 and ACC2, with ACC1 being principally expressed in lipogenic tissues such as
adipose tissue and liver, while ACC2 is predominantly expressed in oxidative tissues like
heart and skeletal muscle [102].
This reaction, which proceeds in two half-reactions, a biotin carboxylase (BC) reaction
and a carboxyltransferase (CT) reaction is the first committed step in fatty acid biosynthesis
and is the rate limiting reaction for the pathway [103].
The ACC is controlled by three global signals glucagon, epinephrine and insulin that
correspond to the overall energy status of the organism. Insulin stimulates FA synthesis by
activating the ACC, whereas glucagon and epinephrine have the reverse effect. The levels of
citrate, palmitoyl CoA and adenosine monophosphate (AMP) within a cell also exert control.
Citrate, a signal that building blocks and energy are abundant, activates the carboxylase.
Palmitoyl CoA and AMP, in contrast, lead to the inhibition of the carboxylase [102, 103,
104].
The remaining series of reactions of fatty acid synthesis in eukaryotes is catalyzed by
fatty acid synthase (FAS) is a homodimeric and multifunctional complex of 250 kDa and
contains seven different enzymatic activities plus a domain that covalently binds a molecule
of 4-phosphopantetheine, of the acyl carrier protein (ACP) [105, 106]. PA, the most abundant
acid, is synthesized de novo from acetyl-CoA as a primer, malonyl-CoA as a carbon donor,
and NADPH as a reducing equivalent according to the following reaction [104, 105, 107].

80

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

Figure 4. Origin of cytoplasmic acetyl-CoA: carnitine shuttle.

Figure 5. Acetyl-coenzyme-A carboxylase (ACC) has critical roles in fatty acid metabolism. The ACCcatalyzed biotin carboxylase (BC) and carboxyltransferase (CT) reactions [103].

Basically, this reaction consists of elongating the acetyl group by C2 units derived from
malonyl-CoA, so that each step takes place by condensation, reduction, dehydration and
further reduction [99] (Figura 6):
1. Transfer of the acetyl group of acetyl-CoA to ACP-catalyzed by acetyl-CoA-ACP
transacylase;

General Aspects of Palmitic Acid

81

2. Next, this two-carbon fragment is transferred to a temporary holding site, the thiol
group of a cysteine residue on the enzyme;
3. The now-vacant ACP accepts a three-carbon malonate unit from malonyl CoA
catalyzed by malonyl CoA-ACP transacylase;
4. The acetyl group on the cysteine residue condenses with the malonyl group on ACP
as the CO2 originally added by acetyl CoA carboxylase is released. The result is a
four-carbon unit attached to the ACP domain. The loss of free energy from the
decarboxylation drives the reaction catalyzed by 3-Ketoacyl-ACP synthase;
5. Reduction of the Beta-keto group to a Beta-hydroxyl group with NADPH catalyzed
by Beta-keto-ACP reductase;
6. Dehydration between the alpha and Beta by Beta-hydroxyacyl-ACP dehydrase. A
molecule of water is removed to introduce a double bond between carbons 2 and 3
(the - and -carbons);
7. Reduction of the trans double bond by NADPH catalyzed by enoyl-ACP reductase).

Figure 6. Reaction sequence for biosynthesis of fatty acids de novo by the animal FAS.

The result of these seven steps is production of a four-carbon compound (butyryl) whose
three terminal carbons are fully saturated, and which remains attached to the ACP. These
seven steps are repeated, beginning with the transfer of the butyryl chain from the ACP to the
Cys residue, the attachment of a molecule of malonate to the ACP (3), and the condensation
of the two molecules liberating CO2 (4). The carbonyl group at the -carbon is then reduced
(5), dehydrated (6), and reduced (7), generating hexanoyl-ACP. This cycle of reactions is
repeated five more times, each time incorporating a two-carbon unit (derived from malonyl
CoA) into the growing fatty acid chain at the carboxyl end. When the fatty acid reaches a
length of 16 carbons, the synthetic process is terminated with palmitoyl-S-ACP. All the
carbons in PA have passed through malonyl CoA except the two donated by the original
acetyl CoA, which are found at the methyl-group () end of the fatty acid [98, 99, 105, 106].

82

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

Palmitic acid can be further elongated by the addition of two-carbon units in the smooth
endoplasmic reticulum (SER). Elongation involves the addition of two-carbon units to a fatty
acyl-CoA, employing malonyl-CoA as the donor and NADPH as the reducing agent. In
mammals, the initial and rate-controlling condensation reaction is catalysed by the elongase
enzymes referred to as elongation of very long-chain fatty acids (ELOVLs) [108]. To date,
seven ELOVL proteins (ELOVL1-7) have been identified, with ELOVL1, ELOVL3,
ELOVL6 and ELOVL7 preferring saturated and monounsaturated fatty acids as substrate; and
ELOVL2, ELOVL4 and ELOVL5 being selective for PUFAs [109, 110]. The process of
elongation requires four separate enzymatic reactions: condensation between the fatty acylCoA and malonyl-CoA to yield 3-ketoacyl-CoA; reduction of 3-ketoacyl-CoA to generate 3hydroxyacyl-CoA; dehydration of 3-hydroxyacyl-CoA to produce trans-2-enoyl-CoA, and;
reduction of trans-2-enoyl-CoA to form the two-carbon elongated acyl-CoA (Figure 7). [108,
109, 111].

Figure 7. Enzymatic steps in long-chain fatty acid elongation. Enzymatic steps of microsomal fatty acyl
chain elongation. ELOVL, elongation of very-long-chain fatty acids; KAR, 3-ketoacyl-CoA reductase;
HADC, 3-hydroxyacyl-CoA dehydratase; TER, trans-2,3-enoyl-CoA reductase [108].

General Aspects of Palmitic Acid

83

Fatty acids are also formed in endoplasmic reticulum and the reaction is catalyzed by
enzymatic systems, generically designated as Acyl-CoA desaturases [112]. Acyl-coenzymeA
(CoA) desaturases introduce a double bond at a specific position on the acyl chain of longchain fatty acids, thereby influencing several of the key biological properties of the fatty acid
itself and of more complex lipids containing this acyl chain. Mammalian cells express 9, 6
and 5-desaturase activities [108]. The desaturation process involves an oxidoreductase chain
(including cytochrome b5) that O2 works as last oxidant of Acyl-CoA and NADPH (or
NADH). The PA product of FAS and its metabolite produced by Stearoyl-Coa Desaturase
(SCD-1), C16:1n-7, can both be further elongated by ELOVL6 to yield stearic acid (C18:0) and
vaccenic acid (C18:1, n7), respectively [108; 109].

Lipolisis
In the fasted state, most tissues, except the brain and red blood cells, rely heavily on the
direct utilization of FA to generate energy. The prime pathway for the degradation of fatty
acids is mitochondrial fatty acid -oxidation (FAO), a key metabolic pathway for energy
homoeostasis in organs [113, 114]. Long-chain fatty acids (LCFA) (C16-18) in tissues exist
as components of TAG or phospholipids. Adipose tissue TAG storages are the primary source
of fatty acids used for FAO during fasting conditions [115].
TAGs are first hydrolyzed by the action of endothelium-bound lipoprotein lipase release
free FAs, which are transported to tissues via the bloodstream. The uptake of FAs seems to be
largely mediated by membrane proteins, although passive uptake probably also occurs. This
implies that several transport steps are necessary before fatty acids are oxidized [115, 116,
117].
The solubility of LCFA in aqueous solutions is extremely low, so the fatty acids must
cross the cell membrane via a protein-mediated mechanism. Membrane-associated fatty acidbinding proteins (fatty acid transporters) are small (15kDa) cytosolic proteins that enhance
the uptake of long chain and very long chain fatty acids into cells. Through their control of
fatty acid transport, metabolism and storage, FABPs are proposed to be central regulators of
lipid metabolism, inflammation and energy homeostasis. In humans, FATPs comprise a
family of six highly homologous proteins, FATP1FATP 6, which are found in all FAs
utilizing tissues of the body [118, 119]. Besides FATPs, FAT/CD36 (fatty acid translocase
/Cluster of Differentiation 36) has been shown to function as a plasma membrane LCFA
transporter in various tissues, including skeletal muscle, heart, liver, adipose tissue and the
small intestine [120, 121]. Other mechanism is fatty acid uptake plasma membrane fatty acidbinding protein (FABPpm) which is associated with the plasma membrane in many tissues
including liver, adipose tissue, cardiac muscle and vascular [122, 123, 124, 125].
After transport across the plasma membrane, FAs must be esterified to coenzyme A, on
the outer mitochodrial membrane by long chain acyl-CoA synthetase activity (ACSL; C12 to
C20) before they can undergo oxidative degradation. This reaction is coupled with two ATP
hydrolysis to AMP and 2Pi. The mitochondrial membrane is not permeable to long chain
acyl-CoA (i.e., C16-C18), therefore requires the initial conversion of acyl-CoA to an ester
acylcarnitine, followed by transport of the acylcarnitine across the inner mitochondrial
membrane into the mitochondrial matrix and subsequent delivery of acyl-CoA [126]. This
process is referred to as carnitine shuttle and requires the concerted action of 3 proteins 6:

84

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

carnitine palmitoyltransferase I (CPT I), carnitine/acylcarnitine translocase (CACT), and


carnitine palmitoyltransferase II (CPT II). [116, 127, 128, 129]. The FAO pathway as
described above is displayed in Figure 8:
1. CPT I converts acyl-CoA compounds to their acylcarnitine metabolites at the outer
mitochondrial membrane and further transported across the inner mitochondrial
membrane by CACT.
2. The enzyme CPT2 is responsible for the conversion of the acylcarnitines back to the
corresponding acyl-CoAs, the true substrates of the FAO pathway.

Figure 8. The mitochondrial carnitine shuttle. Abbreviations: CPT1, carnitine-palmitoyl transferase 1;


CPT2, carnitine-palmitoyl-transferase 2; carnitine/acylcarnitine translocase (CAC or CACT) [127].

The complete oxidation PA is achieved in the following three steps (Figure 9): betaoxidation of fatty acid chain yielding acyl-CoA; the oxidation of acetyl CoA to CO2 and
production FADH2 and NADH2 in citric acid cycle; the transfer of electron from reduced
electrons carries FADH2 and NADH2 to mitochondrial respiratory chain resulting into ATPs
[130]. This process involves a variety of enzymes: long-chain acyl coenzyme dehydrogenase
(LCAD), enoyl-CoA hydratase, hydroxyacyl-CoA dehydrogenase and ketoacyl-CoA thiolase
[117, 131];
1. In the first step, LCAD, catalyzes oxidation of the fatty acid moiety of acyl-CoA to
produce a double bond is introduced into a carboxylic acid between the and
carbons, FAD is the electron acceptor, and electrons from the reaction ultimately
enter the respiratory chain and are carried to O2 with the concomitant synthesis of
two ATP molecules per electron pair;
2. In the second step of the fatty acid oxidation cycle, water is added to the double bond
of the trans-2-enoyl-CoA to form the L stereoisomer of -hydroxyacyl-CoA. This
reaction, catalyzed by enoyl-CoA hydratase;
3. In the third step, the L--hydroxyacyl-CoA is dehydrogenated to form -ketoacylCoA by the action of -hydroxyacyl-CoA dehydrogenase; NAD+ is the electron
acceptor. The NADH formed in this reaction donates its electrons to NADH

General Aspects of Palmitic Acid

85

dehydrogenase an electron carrier of the respiratory chain. Three ATP molecules are
generated from ADP per pair of electrons passing from NADH to O2 via the
respiratory chain;
4. Finally hydroxy-acyl-CoA is dehydrogenated to 3-keto-acyl-CoA. Then, thiolytic
cleavage of the 3-keto-acyl-CoA produces a two-carbon chain-shortened acyl-CoA
plus acetyl-CoA. Each cycle yields an acyl-CoA shortened by two carbon atoms, an
acetyl-CoA, and one nicotinamide adenine dinucleotide (NADH) and one flavin
adenine dinucleotide (FADH2) as electron carriers (or reducing equivalents).
The PA undergoes seven passes through this oxidative sequence, in each pass losing two
carbons as acetyl-CoA. At the end of seven cycles the last two carbons of palmitate
(originally C-15 and C-16) are left as acetyl-CoA. Generally, the total ATP yield due to the
complete oxidation of palmitic acid in the following equation [99, 130]:
Palmitic acid + 8 Coenzyme A + 7 FAD+ + 7NAD+ + ATP 8 CH3CO-SCoA (AcetylCoA) + 7FADH2 + 7 (NADH + H+) + AMP + PPi.
If one acetyl CoA involved in TCA cycle gives = 10 ATPs; ATPs due 8 acetyl-CoA = 8
x 10 = 80; ATPs due to 7 FADH2 = 1.5 x 7= 10.5; ATPS due 7 (NADH + H+) = 2.5 x 7.5 =
17.5. The total of number ATPs produced 108. During the initiation of the - oxidation
pathway a 2 ATPs converts into a 2 AMP and 2 Pi for the activation of fatty acid. So, net
ATPs produced by palmitic acid are 106. These calculations assume that mitochondrial
oxidative phosphorlation produces 1.5 ATPs/FADH2 oxidized and 2.5 ATP/NADH2 oxidized.
The Guanosine-5'-triphosphate (GTP) produced directly in the acid citric cycle yields ATP in
the reaction catalyzed by nucleoside diphosphate kinase [99, 130].

Figure 9. The -oxidation of saturated fatty acids involves a cycle of four enzyme-catalyzed reactions.
Each cycle produces single molecules of FADH2, NADH, and acetyl-CoA and yields a fatty acid
shortened by two carbons.

86

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

Furthermore, the produced acetyl-CoA during PA -oxidation goes to TCA or to ketone


bodies production. The oxaloacetate availability to make acetyl-CoA get in the citric acid
will determinate which metabolic pathway acetyl-CoA will follow [132]. In some situations,
as fasting or catecholaminergic stress, oxaloacetate molecules will be intended to glucose
synthesis by gluconeogenesis. With this, oxaloacetate availability content will be decreased,
and consequently, less acetyl-CoA will get into TCA, thereby having Ketogenesis [133]. In
this process, ketone bodies are formed. These are small water-soluble molecules, easily
transportable form of acetyl units that can be oxidized to carbon dioxide and water to yield
energy. Ketone bodies include acetoacetate (AcAc), 3-hydroxybutyrate (3HB) and acetone,
which are an important source of energy for peripheral tissues, supply up to 50% of basal
energy requirements these tissues and up to 70% for the brain which cannot derive energy
from other sources when blood glucose levels are low [134, 135].

HEALTH EFFECTS OF PALMITIC ACID


Effects of Palmitic Acid in Serum Lipids and Lipoproteins
Dietary and endogenous fats are carried to the target organs by different lipoproteins, i.e.
chylomicrons, LDL and HDL. These particles contain a core of TAG liposoluble vitamins
and cholesteryl esters, surrounded by a phospholipid and free cholesterol layer. These also
contain, by the way, specific proteins, called apolipoproteins (apo) which act as enzyme
cofactors or receptor ligands. Exogenous fat is transported in chylomicrons from the intestinal
epithelium to the peripheral cells, reaching the bloodstream via the lymphatic system [11].
After entry in the blood stream the chylomicrons are hydrolyzed by the endothelial-bound
lipoprotein lipase with apo C-I as a co-factor, allowing the delivery of free FAs to muscle and
adipose tissue. The chylomicron remnants are rapidly taken up into the liver via especial
receptor. ApoE is the moiety required for rapid hepatic removal. Its activity is inhibited by C
apolipoproteins, especially apoC-I. The liver utilizes the exogenous fat and can release
surplus lipids via VLDL into the blood. The VLDL is another substrate for lipoprotein lipase.
The remaining VLDL remnants can either be taken up into the liver or are hydrolyzed to
LDL. These last delivers cholesterol to all body cells via its receptor [136]. Moreover other
type of lipoprotein denominated as highdensity protein (HDL) is an important scavenger of
surplus cholesterol transporting it from cell membranes to the liver, where it is degraded or
converted into biliary salts, an then eliminated by the entero-hepatic cycle [137].
Keys et al. [138] and Hegsted et al. [139] showed from mathematical equations that
serum cholesterol concentration of a person would be predicted by the diet fat consumption.
Furthermore, SFAs were two times more effective in raising cholesterol, and PUFA reducing
them. In this context, SFA can not correspond with more than 1/3 of fat diet intake.
Nevertheless, there are great reservations about those equations, due to they were made with
middle-aged men who consumed high fats contents and, therefore, theyre not necessarily
valid to other populations in different diet conditions. In addition, in 1960, when the
biochemical and dietary analyses were done, trans and n-3 PUFA could not be identified and
hence only information on saturated monounsaturated and polyunsaturated fats were report.

General Aspects of Palmitic Acid

87

Keys et al. [138]: TC = 2.76 SFA 1.356 PUFA 1 1.56D C1/2


Where TC is the total cholesterol, SFA is the proportion of SFA (% of the total energy
amount), UFA is the proportion of unsaturated FA (% of the total energy amount) and C is
the dietary cholesterol (in mg per 1000 kcal/day).
Hegsted et al. [139]: TC = 2.166 SFA 1.356 PUFA 1 1.56 C
Where C is the dietary cholesterol (in mg/day).
Much effort has gone into proving that SFA is the major dietary factor for
hypercholesterolemia, although the mechanism by which this would occur still remains
unclear. Some hypotheses have been cited to explain its effect. On the one hand, they might
be responsible for a decrease in the LDL receptor expression on the hepatocyte surface, and
on the other hand, their incorporation into the cell membrane phospholipids might impair its
fluidity, thus disturbing the receptor action. A diet rich in SFAs may inhibit, to some extent,
the hepatic esterification of cholesterol, thus raising the concentration of unesterified
cholesterol. In response to raised unesterified cholesterol, the hepatic concentration of LDL
receptor mRNA is decreased, thus reducing LDL receptor activity, resulting in raised plasma
LDL cholesterol levels [140].
Nonetheless, the debate over what constitutes the "ideal" fat is controversial, since it uses
the term saturated fat without distinguishing individual SFA. In this sense, even a cursory
analysis of some of the so-called saturated fats (e.g., palm oil, lard, tallow, butter, coconut oil)
reveals that they have distinct profiles and empirically exert different metabolic effects.
Accordingly, research in recent years has shifted toward elucidating the effects of specific
dietary fatty acids in TAG, as opposed to specific classes of fats, on plasma lipids and
lipoprotein metabolism [141].
Scientific studies point out an association between saturated fat and blood lipids increase
due to the presence of three principal FAs: myristic, lauric and palmitic. Since PA is the
principal saturated acid in the diet, it is the chief saturated component used to develop the diet
equations [138; 139], has meant that the cholesterol-raising property of SFA has generally
been attributed to their palmitic acid content. Researches that assess your action in many body
functions had been based on human and animals studies, especially, with palm oil and its
fractions as principal source of PA [review 142].
Whereas most studies have shown a cholesterol-raising effect of this FA [140, 143, 144,
145], some others have demonstrated relative neutrality [141]. According to Grundy and
Denke [140], three nutritional factors would be responsible for raising serum LDL levels;
these are SFA, cholesterol itself and excess caloric intake. These authors point out the major
cholesterol-raising SFA in the diet would be the PA. In the same way, Sun et al. [146]
reviewed 25 trials of at least 2 weeks that compared the effects of palm oil consumption with
natural highly unsaturated and partially hydrogenated vegetable oils and animal fat. These
authors concluded that palm oil consumption results in higher LDL cholesterol levels than
other natural unsaturated vegetable oils and it may be preferable to trans-fat rich oils based on
its effect on HDL cholesterol and suggested that more studies are needed to evaluate the
effects of palm oil consumption on incidence of coronary heart diseases.
Contrary to the predictions of the Keys et al. [138] and Hegsted et al. [139] equations, the
review about the cholesterolaemic effects of palm oil by Wai [review 142], indicates that the

88

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

substitution of palm oil or its liquid fractions (palm olein, super olein) for habitual fats in the
diet does not result in an elevation of total serum cholesterol. In another review, Edem [23]
concluded that, in animal experiment and human studies, palm oil administration with
approximately 50 % of saturated fatty acid, does not behave as saturated oil, reducing the
blood levels of total cholesterol, LDL cholesterol. A review by Sundram [29] also concluded
that high levels of palmitic acid in the diet do not significantly affect serum total and LDL
cholesterol levels.
Khosla and Hayes [141] conclusions about cholesterolaemic effects of the saturated fatty
acid of palm oil suggest that not all SFAs are cholesterol-raising. According to authors, when
fatty acids contents are similar, the palmitic acid appears to have no impact on the plasma
cholesterol in normocholesterolaemic subjects. Above 400 mg of dietary cholesterol intakeed
per day, PA might be cholesterol increasing, even more than myristic acid and quite neutral
underneath this value. Nevertheless, if cholesterol consumption exceeds the critical value or
when hypercholesterolaemic subjects are studied, the PA appears to increase the plasma
cholesterol. Furthermore, authors linked the different PA actions to the differences in LDLreceptor status. It seems that more studies are needed to explain these inconclusive results.
Free FAs absorption rate depends on the type of fatty acid and intestine emulsifier
environment. An important explanation of why palm oil does not "follow the Keys [138] and
Hegsted [139] model is due to unsaturated fatty acids are in sn-2 position (> 58.25 % of oleic
acid and > 18.41 % of linoleic acid) and a high proportion of PA is in sn-1 and 3 positions
(17-23 %) [23]. Therefore, as already mentioned in this text, fatty acids in sn-2 position are
preferentially absorbed at bowel wall and, thereby, more bioavailable than the fatty acids in
sn-1 and 3 positions [87]. On the assumption that all SFA localized at sn-1 and 3 positions in
palm oil are preferentially absorbed, whilst saturated are faecal excreted as salts, therefore
only 8 % of SFA localized at sn-2 position would be absorbed as consequence there is a less
caloric intake and a lower serum TGA content.
Another explanation to palm oil hypocholesterolemic action would be the presence of
tocotrienols. They are admittedly considered hypocholesterolemic once they regulate
cholesterol synthesis through 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA)
reductase inactivation enzyme that primarily synthesizes the cholesterol [147].
In a review of Hayes and Khosla [148], the authors concluded that cholesterolemic
effects of PA are large extent determined by the concomitant level of linoleic FAs. It is why
C18:2 regulate numerous lipogenic genes involved in fatty acid synthesis. PUFA maximally
inhibit hepatic gene transcription when they provide 20 % of the dietary calories.
Nonetheless, as little as 5% of calories as PUFA will inhibit gene expression 50 %. In this
way, once 18:2 intake is above threshold (57 %) the detrimental effects of the SFA on
LDL are no longer observed in part because LDL receptors are maximally up-regulated and
sterol regulatory element-binding protein (SREBP1c; that group of proteins uptake
cholesterol and FAs biosynthesis) is inhibited, resulting in decreased FAs synthesis and
decreased VLDL secretion. As a consequence, PA appears neutral above the threshold
requirement for linoleic FAs (Figure 10). Simply stated it implies that a certain level of 18:2
is required by an individual to prevent certain SFA-rich fats from raising the serum LDL
level. If you drop below your 18:2 threshold, LDL will increase, with the increase being most
severe during consumption of 12:0 1 14:0-rich fats. Also note that if 18:2 intake is high
enough (610 % en depending on the LP setpoint), SFA no longer have a significant
cholesterol-raising effect. Simply stated it implies that a certain level of 18:2 is required by an

General Aspects of Palmitic Acid

89

individual to prevent certain SFA-rich fats from raising the serum LDL level. If you drop
below your 18:2 threshold, LDL will increase, with the increase being most severe during
consumption of 12:0 1 14:0-rich fats. Also note that if 18:2 intake is high enough (610 %
depending on the LP setpoint), SFA no longer have a significant cholesterol-raising effect
[148].

Figure 10. The scheme depicts the 18:2/SFA ratio hypothesis, which can be applied to data in Simply
stated it implies that a certain level of 18:2 is required by an individual to prevent certain SFA-rich fats
from raising the serum LDL level. If you drop below your 18:2 threshold, LDL will increase, with the
increase being most severe during consumption of 12:0 1 14:0-rich fats. Also note that if 18:2 intake is
high enough (610 % depending on the lipids setpoint), SFA no longer have a significant cholesterolraising effect [148].

Palmitic Acid and Fatty Liver Disease


The liver is a central organ in metabolism that serves multiple functions such as protein
synthesis, glycogen storage, hormone production, and detoxification [149]. Fatty liver is the
earliest and the most common form of both nonalcoholic steatohepatitis (NASH) and
nonalcoholic fatty liver disease (NAFLD) [150, 151]. Hepatocellular injury, inflammation
and fibrosis are hallmarks of NASH, which are observed in only a fraction of subjects with
NAFLD, although the exact mechanisms leading from NAFLD to NASH are still largely
unknown [152]. Several lines of evidence indicate the importance of both quantitative and
qualitative (e.g. saturated vs unsaturated) changes in dietary FAs as relevant mechanisms for
the development of NAFLD both in rodent models and in humans [150, 153, 154, 155].
NAFLD develops when consumption of energy exceeds the combustion of calories and
the unburnt energy is conserved in the form of TAG in liver [150]. Donnellys et al. [156]
research with obese patients showed that TAG accounted for in liver, 59.0 % of TAG arose
from nonesterified fatty acid; 26.1 % from de novo lipogenesis and 14.9 % from the diet.
Patients under those conditions develop with a high production of reactive
oxygen/nitrogen species (ROS/RNS) that promote hyper-stimulation of Ito cells, increasing
collagen production in hepatic parenchyma with a consequent fibrosis and cirrhosis that can
lead to hepatocellular carcinoma [157, 158]. Oxidative stress refers to an imbalanced cellular
state in which the production of ROS/RNS are increased to an extent that overrides the
normal operating free radical clearing mechanisms; such as glutathione peroxidase (GPx),
superoxide dismutase (SOD) and catalase [159]. This process plays a significant role in the

90

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

development of hepatic steatosis to NASH. The increase in FA oxidation in the steatotic state
could potentially induce an increased electron flux through the electron transport chain
(ETC), which may lead a major production of reactive oxygen/nitrogen specie (ROS/RNS).
These reactive lipid derivatives have the potential to amplify intracellular damage by
mediating the diffusion of ROS/RNS into the extracellular space thus causing tissue damage
[160].
Enzymatic and no enzymatic systems are also capable to avoid hepatic damage, inducing
an inflammatory process initiation. Damage and lipid peroxidation products induce an
inflammatory response with up-regulation of pro-inflammatory cytokines including alpha
tumoral necrosis factor (TNF-), interleukins 6 and 1 (IL-6 and IL-1). These cytokines are of
major importance for directing polymorphonuclear and mononuclear leukocytes into inflamed
tissues [158]. The greater cytokines pro-inflammatory production, especially TNF- and IL-6
and IL-1, can contribute to peripheral and hepatic of insulin, which induce to an infiltration in
the hepatic parenchyma, in a vicious cycle that promotes more tissue injury. This mechanism
is described as the two hit model with the first hit being steatosis and insulin resistance,
and the second hit needed to initiate NASH requiring other factor(s) that promote lipid
peroxidation, inflammatory cascade, oxidative stress, tissue injury and inflammatory process
[161]. Moreover a multiple parallel hits model has been suggested to promote progression
of steatosis to NASH because of failure of the antilipotoxic protection systems of the liver
and multiple hits from the gut and/or adipose tissue [162].
Palmitic acid roles in NAFLD installation and development have been discussed. PA
overloading is known to induce apoptotic cell death and a large number of molecular
mechanisms have been implicated in this action: nitric oxide (NO) synthesis, suppression of
antiapoptotic factors such as Bcl-2 [163, 164] reactive oxygen species generation,
endoplasmic reticulum stress [165], nuclear factor-kB activation [166].
Apoptosis can be triggered by mitochondrial damage, which is followed by the release of
cytochrome c and the caspase cascade [167]. The BAX protein activates mitochondrial
release of cytochrome c [168], while Bcl-2 is a mitochondrial protein inhibits the apoptotic
process and promotes cell survival [169]. Ji et al. [163] research showed that occur a decrease
in mitochondrial Bcl-2 and a markedly increase in mitochondrial level of Bax in the HepG2
cells treated with PA from 200 to 400 mM concentrations. The authors suggest that this
mechanism can contribute to NASH and NAFLD installation, and especially may play an
important role in the transition from steatosis to steatohepatitis in human. Other studies [170]
on the effect of FAs-induced steatosis on cellular apoptosis have demonstrated that palmitic
and oleic FA mixtures-induced steatosis is associated with apoptosis in hepatocyte cell
cultures. Joshi-Barves et al. [171] studies also showed that exposure to excess palmitic acid
induces apoptosis and IL-8 production in hepatocytes in a relation of dose-dependent and
time dependent manner, via activation of c-Jun amino terminal kinase (JNK/AP-1), and
nuclear factor kappa B (NF-B) transcription factors for IL-8 expression.

Palmitic Acid and Diabetes


Diabetes is a group of metabolic diseases characterized by hyperglycemia resulting from
defects in insulin secretion, insulin action, or both [172, 173]. The vast majority of cases of

General Aspects of Palmitic Acid

91

diabetes fall into two broad etiopathogenetic categories. In one category, type 1 diabetes, the
cause is an absolute deficiency of insulin secretion. Individuals at increased risk of
developing this type of diabetes can often be identified by serological evidence of an
autoimmune pathologic process occurring in the pancreatic islets and by genetic markers. In
the other, much more prevalent category, type 2 diabetes, is associated with a combination of
pancreatic -cell dysfunction and insulin resistance [174]. The chronic hyperglycemia
produces glucotoxicity characterized by -cell function gradual deterioration and insulin
resistance aggravation. It is similar to the paradoxically deleterious effects of chronic
hyperglycemia, if lipotoxicity is produced, once the free FAs which are essential fuels in
the normal state, become toxic when they are chronically present in excessive levels
[164, 175].
Under diabetic conditions, oxidative stress and endoplasmic reticulum stress are induced
in various tissues [173, 176, 177, 178, 179]. Moreover, the -cells have very low levels of
antioxidative enzymes, becoming them more susceptible to the stress [172]. ROS can function
as signaling molecules to activate a number of cellular stress-sensitive pathways that cause
cellular damage, and are ultimately responsible for the late complications of diabetes.
Evidence suggests that common stress-activated signaling pathways such as nuclear factor
nuclear factor-B (NF-B) [180, 181, 182], p38 mitogen-activated protein kinase (MAPK)
[183], protein kinase C (PKC) [184], toll-like receptors (TLRs) [185, 186], and c-Jun Nterminal kinase (JNK) [187; 188] underlie the development of these diabetic complications.
Nuclear factor kappa-B (NFkB), a redox-sensitive transcription factor regulating a battery
of inflammatory genes, has been indicated to play a role in the development of numerous
pathological states [189]. Activation of NFkB induces gene programs leading to transcription
of factors that promote inflammation, such as leukocyte adhesion molecules, cytokines and
chemokines [181].
Mitogen-activated protein kinases (MAPKs) are a family of serine/threonine kinases and
consist, among others, Jun N-terminal protein kinase (JNK), p38s MAP kinase, cyclic AMP
dependent protein kinase (PKA), protein kinase B (PKB) and protein kinase C (PKC) [190].
It is involved in the regulation of a wide range of cellular responses, including cell
proliferation, differentiation, and survival [191]. Its also well established that p38 and JNK
play important roles in mediating apoptosis caused by various stimuli [190]. There are three
isozymes of JNK: JNK1, JNK2 and JNK3, and that only JNK1 has been shown to be
implicated in type 2 diabetes [192] probably to reduce insulin gene expression [187]. The
TLR family is known to consist of 10 members (TLR1-TLR10) that are pattern recognition
receptors which initiate innate immune responses upon recognition of a wide range of
pathogen-associated molecular patterns [185, 193].
In vitro studies have shown that -cells are vulnerable to palmitate, in the presence of
high glucose concentration [194, 195]. Many authors report that the palmitate induces -cell
dysfunction in vivo by activating inflammatory processes within islets, for example:
activation of nuclear factor-kB, resulted in increased expression of several proinflammatory
cytokines (TNF-, IL-1, IL-6, MCP1) in rat liver as well as an increase in circulating MCP1
levels. The rise in plasma MCP1 is particularly interesting because MCP1 is well established
to regulate macrophage recruitment to sites of inflammation [196].
Additionally, palmitate increased the expression and secretion of inflammatory cytokines
(e.g. IL-6 and TNF-) and impaired insulin sensitivity via an NFkB/PKC pathway in muscle
cells [197, 198]. Too increased intracellular concentration of PA producing diacylglycerol

92

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

have been shown to activate and cause cellular redistribution of protein kinase C isoforms
[199], which result in the induction of inflammatory pathways via NF-B activation [200].
Jiang [201] studies with PA showed to induce endothelial progenitor cells apoptosis via p38
and JNK mitogen activated protein kinases MAPK pathways in a time-and-dose-dependent
manner. Eguchi et al. [186], through a combination of in vivo and in vitro studies, reported
that SFA palmitate induces -cell dysfunction. According to authors this cell responds to
palmitate via the TLR4/MyD88 pathway and produce chemokines that recruit M1-type
proinflammatory monocytes/macrophages to the islets. Depletion of M1-type cells protected
mice from palmitate-induced -cell dysfunction.
After insulin binds to insulin receptor on cell surface, insulin receptor and its substrates
are phosphorylated, which leads to activation of various insulin signaling pathways [202].
Reynoso et al. [203] evaluated several aspects of the insulin resistance induced by palmitic
acid in rats and found that after treatment with 0.09 g/kg of palmitic acid there is a delay in
the curve of tolerance to glucose. The authors concluded that occur an increase in the
phosphorylations in serine of the insulin receptor after the treatment with palmitate,
suggesting that PKC has a role as negative regulator of the insulin receptors activation in the
insulin resistance induced by palmitic acid.

CONCLUSION
Palmitic acid is the most abundant saturated fatty acid in human nutrition. It is a major
component of palm oil, but can also be found in beef tallow, lard, cocoa butter, human, cows
milk and interesterification food. PA is the first fatty acid produced during fatty acid synthesis
and the precursor to longer fatty acid. Through this bioprocesses, glucose is converted to fatty
acids, which then react with glycerol to produce triacylglycerols. Furthermore, palmitic acid
can participates in several chemical reactions as other acids of this same class, being attached
to the alcohol form of vitamin A which has been used successfully as a supplement due to its
high stability in relation to vitamin A and low cost. Several studies have documented that
palmitic acid position in TGC molecule has a great importance in fatty acids action in several
human metabolic bioprocesses. In addition, a higher palmitic acid absorption is obtained with
formulas rich in palmitic acid esterified in triacylglycerols sn-2 position, than those
predominantly esterified in sn-1,3 positions. Some authors suggest that palm oil does not
behave as saturated oil, reducing the blood levels of total cholesterol, LDL cholesterol due to
TGC sn position. In the other hand, if cholesterol consumption exceeds the critical value or
when hypercholesterolaemic subjects are studied, the palmitic acid appears to increase those
cholesterol levels. There are still other researches that observed adverse healthy effects by the
use of palmitic acid. Moreover, also several studies have documented roles in NAFLD
installation and insulin resistance higher levels development in diets rich in palmitic acid.
Nonetheless, those studies have still much divergent results, being necessary more researches
to clarify the real participation of PA in these processes.

General Aspects of Palmitic Acid

93

REFERENCES
[1]
[2]
[3]
[4]

[5]
[6]
[7]

[8]

[9]
[10]
[11]
[12]

[13]
[14]

[15]

[16]

[17]

German, JB; Dillard, CJ. Saturated fats: what dietary intake? 13. Am J Clin Nutr, 2004;
80: 5509.
Van Meer, G; Voelker, DR; Feigenson, GW. Membrane lipids: where they are and how
they behave. Nat Rev Mol Cell Biol, 2008; 9: 11224.
FAO/WHO Expert Consultation. Fats and Fatty Acids in Human Nutrition. Food Nutr
Pap, 2010; 91: 1166.
Ratnayake, WM; Galli, C. Fat and fatty acid terminology, methods of analysis and
fat digestion and metabolism: A background review paper. Ann Nutr Metab, 2009; 55
(1-3): 843.
Vannice, G; Rasmussen, H. Position of the Academy of Nutrition and Dietetics:
Dietary Fatty Acids for Healthy Adults. J Acad Nutr Dietetic, 2014; 2014, 13653.
Kruger, MC; Coetzee, M; Haag, M; Weiler, H. Long-chain polyunsaturated fatty acids:
selected mechanisms of action on bone. Prog Lipid Res, 2010; 49: 43849.
Sales, ALC. Sntese, caracterizao e anlise trmica dos sais de ltio, sdio e potssio
do cido Palmtico e de seu ster etlico [dissertation]. So Carlos, (SP): Instituto de
Qumica de So Carlos, Universidade de So Paulo; 2006.
Sousa, FF; Saraiva, GD; Freire, PTC; Lima Jr, JA; Alcantara Jr, P; Melo, FEA; et al.
Pressure-induced phase transitions in palmitic acid: C form. J Raman Spectrosc, 2012;
43: 14652.
Fieser, LF; Harris, GC; Hershberg, EB; Morgana, M; Novello, FC; Putnam, ST.
Napalm. Anal Chem (Industrial and engineering chemistry), 1946; 38(8): 76873.
OByrne, SM; Blaner, WS. Retinol and retinyl esters: biochemistry and physiology. J
Lipid Res, 2013; 54: 17343.
Harrison, EH. Mechanisms of digestion and absorption of dietary vitamin A. Annu Rev
Nutr, 2005; 25: 87103.
Ihara, H; Tani, A; Kakinoki, T; Shino, Y; Morita, Y; Matsumoto, N; et al. Metabolism
of retinyl esters in humans after ingestion of retinol. J Anal Bio-sci, 2012; 35(2): 146
50.
Hussain, MM. A proposed model for the assembly of chylomicrons. Atheroscler, 2000;
148(1): 115.
Blaner, WS; OByrne, SM; Wongsiriroj, N; Kluwe, J; DAmbrosio, DM; Jiang, H; et
al. Hepatic stellate cell lipid droplets: a specialized lipid droplet for retinoid storage.
Biochim Biophys Acta, 2009; 1791: 46773.
Harrison, EH; Blaner, WS; Goodman, DS; Ross, AC. Subcellular localization of
retinoids, retinoid-binding proteins, and acyl-CoA:retinol acyltransferase in rat liver. J
Lipid Res, 1987; 28: 97381.
Souganidis, E; Laillou, A; Leyvraz, M; Moench-Pfanner, R. A Comparison of Retinyl
Palmitate and Red Palm Oil -Carotene as Strategies to Address Vitamin A Deficiency.
Nutrients, 2013; 5: 325771.
United States Department of Agriculture. USDA Nutrient Database for Standard
Reference, Release 17 [online]. Nutrient Data Laboratory Homepage. Agric Res Serv,
2004 [cited 2014 Apr 02]. Available from: URL: http://www.nal.usda.gov
/fnic/foofcomp.

94

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

[18] Dary, O; Mora, JO. Food fortification to reduce Vitamin A deficiency: International
Vitamin A Consultative Group Recommendations. J Nutr, 2002; 132: 2927S33S.
[19] Bagriansky, J; Ranum, P. Vitamin A Fortification of PL480 Vegetable Oil. Washington,
DC, USA: SUSTAIN; 1998.
[20] Surman, SL; Jones, BG; Rudraraju, R; Sealy, RE; Hurwitza, JL. Intranasal
Administration of Retinyl Palmitate with a Respiratory Virus Vaccine Corrects
Impaired Mucosal IgA Response in the Vitamin A-Deficient Host. Clin Vaccine
Immunol, 2014; 21(4): 598601.
[21] Godic, A; Poljak, B; Adamic, M; Dahmane, R. The Role of Antioxidants in Skin
Cancer Prevention and Treatment. Oxidative Med Cell Longevity, 2014; 2014: 16.
[22] Rawlings, AV; Stephens, TJ; Herndon, JH; Miller, M; Liu, Y; Lombard, K. The effect
of a vitamin A palmitate and antioxidant-containing oil-based moisturizer on
photodamaged skin of several body sites. J Cosmet Dermatol, 2013; 12(1): 2535.
[23] Edem, DO. Palm oil: Biochemical, physiological, nutritional, hematological, and
toxicological aspects: A review. Plant Foods Hum Nutr, 2002; 57: 31941.
[24] Mohammad, R; Sarmidi, MR; Hesham, A; El Enshasy, H; Hamid, MA. Oil Palm: The
Rich Mine for Pharma, Food, Feed and Fuel Industries. Am Eurasian J Agric Environ
Sci, 2009; 5(6): 76776.
[25] Adnan, H. Global demand for palm oil growing rapidly [online]. The Star On Line,
2011 [cited 2014 Mar 10]. Available from: URL: http://www.thestar.com.my/story.
aspx?sec=busines...&file=%2f2011%2f3%2f10%2fbusiness%2f8224650.
[26] Berger, KG. Palm oil. Chan Jr., HT (Ed.). New York: Handbook of Tropical Foods,
Marcel Dekker Inc.; 1983. P. 43368.
[27] Murphy, DJ. Future prospects for oil palm in the 21st century: Biological and related
challenges. Eur J Lipids Sci Technol, 2007; 109: 296306.
[28] Ong, ASH; Goh, SH. Palm oil: A healthful and cost-effective dietary component. Food
Nutr Bull, 2002; 23: 1122.
[29] Sundram, K; Sambanthamurthi, R; Tan, Y-A. Palm fruit chemistry and nutrition. Asia
Pac J Clin Nutr, 2003; 12(3): 35562.
[30] Gibon, V; De Greyt, W; Kellens, M. Palm oil refining. Eur J Lipid Sci Technol, 2007;
109(4): 31535.
[31] Malaysian Palm Oil Council, MPOC. Fact Sheets. Malaysian Palm Oil [online].
Malaysia: Malaysian Palm Oil Council and Malaysian Palm Oil Board, 66 p. 2014
[2014 Mar 10]. Available from: URL: www.mpoc.org.my/envo_pub_190607_01.asp.
[32] Nagendran, B; Unnithan, UR; Choo, YM; Sundram, K. Characteristics of red palm oil,
a carotene- and vitamin E- rich refined oil for food uses. Food Nutr Bull, 2000; 21(2):
18994.
[33] Rossi, M; Gianazza, M; Alamprese, C; Stanga, F. The effect of bleaching and physical
refining on color and minor components of palm oil. J Am Oil Chem Soc, 2001; 78(10):
10515.
[34] Scrimshaw, NS. Nutritional potential of red palm oil for combating vitamin A
deficiency. Food Nutr Bull, 2000; 21(2): 195201.
[35] Sambanthamurthi, R; Sundram, K; Tan, Y. Chemistry and a biochemistry of palm oil.
Prog Lipid Res, 2000; 39(6): 50758.

General Aspects of Palmitic Acid

95

[36] Almeida, DT; Nunes, IL; Conde, PL; Rosa, RPS; Rogrio, WF; Machado, ER. A
quality assessment of crude palm oil marketed in Bahia, Brazil. Grasas y Aceites, 2013;
64(4): 38494.
[37] Palm Oil Refiners Association of Malaysia, PORAM. Standard Specifications for
Crude Palm Oil [online]. 2014 [2014 Feb 17]. Available from: URL:
http://www.poram.org.my/database/contract/specifications.htm.
[38] Gee, PT. Analytical characteristics of crude and refined palm oil and fractions. Eur J
Lipid Sci Technol, 2007; 109: 3739.
[39] Hettinga, D. Butter. In: Shahidi, F. Baileys Industrial Oil and Fat Products. 6th ed. Six
Volume Set. John Wiley & Sons, Inc.; 2005.
[40] Verardo, V; Gmez-Caravaca, AM; Gori, A; Losie, G; Cabonia, MF. Bioactive lipids
in the butter production chain from Parmigiano Reggiano cheese area. J Sci Food
Agric, 2013; 93: 362533.
[41] Lipp, M; Anklam, E. Review of cocoa butter and alternative fats for use in Chocolate.
Part A. Compositional data. Food Chem, 1998; 62: 7397.
[42] Minin, VPR; Cecchi, HM; Minin, LA. Determinao de Substitutos da Manteiga de
Cacau em Coberturas de Chocolate Atravs da Anlise de Triacilgliceris. Cinc
Tecnol Alim, 1999; 19: 27781.
[43] Jahurul, MHA; Zaidul, ISM; Norulaini, NAN; Sahena, F; Jinap, S; Azmir, J; et al.
Cocoa butter fats and possibilities of substitution in food products concerning cocoa
varieties, alternative sources, extraction methods, composition, and characteristics. J
Food Eng, 2013; 117: 46776.
[44] Lipp, M; Simoneau, C; Ulberth, F; Anklam, E; Crews, C; Brereton, P; et al.
Composition of Genuine Cocoa Butter and Cocoa Butter Equivalents. J Food
Composition Anal, 2001; 14: 399408.
[45] Asep, EK; Jinap, S; Tan, TJ; Russly, AR; Harcharan, S; Nazimah, SAH. The effects of
particle size, fermentation and roasting of cocoa nibs on supercritical uid extraction of
cocoa butter. J Food Eng, 2008; 85: 4508.
[46] Frega, N; Bocci, F; Lercker, G; Bortolomeazzi, R. Lipid composition of some avocados
cultivars. Int J Food Sci, 1990; 3: 197204.
[47] Berasategi, I; Barriuso, B; Ansorena, D; Astiasarn, I. Stability of avocado oil during
heating: Comparative study to olive oil. Food Chem, 2012; 132(1): 43946.
[48] Swisher, HE. Avocado oil: from food use to skin care. J Am Oil Chem Soc, 1988; 65:
17046.
[49] Villa-Rodrguez, JA; Molina-Corral, FJ; Ayala-Zavala, JF; Olivas, GI; GonzlezAguilar, GA. Effect of maturity stage on the content of fatty acids and antioxidant
activity of Hass avocado. Food Res Int, 2011; 44: 12317.
[50] Ozdemir, F; Topuz, A. Changes in dry matter, oil content and fatty acids composition
of avocado during harvesting time and post-harvesting ripening period. Food Chem,
2004; 86: 7983.
[51] Yanty, NAM; Marikkar, JMN; Long, K. Effect of Varietal Differences on Composition
and Thermal Characteristics of Avocado Oil. J Am Oil Chem Soc, 2011; 88: 1997
2003.
[52] Kallio, H; Yli-Jokipii, K; Kurvinen, J; Sjvall, O; Tahvonen, R. Regioisomerism of
Triacylglycerols in Lard, Tallow, Yolk, Chicken Skin, Palm Oil, Palm Olein, Palm

96

[53]

[54]
[55]
[56]

[57]

[58]
[59]
[60]
[61]

[62]

[63]

[64]

[65]

[66]

[67]

[68]

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa


Stearin, and a Transesterified Blend of Palm Stearin and Coconut Oil Analyzed by
Tandem Mass Spectrometry. J Agric Food Chem, 2001; 49, 33639.
Marikkar, JMN; Ghazali, HM; Che Man, YB; Lai, OM. The use of cooling and heating
thermograms for monitoring of tallow, lard and chicken fat adulterations in canola oil.
Food Res Int, 2002; 35: 100714.
Ali, Y; Hanna, MA; Cuppett, SL. Fuel Properties of Tallow and Soybean Oil Esters. J
Am Oil Chem Soc, 1995; 72(12): 155764.
Ma, F; Hanna, MA. Biodiesel production: a review. Bioresour Technol, 1999; 70: 1-15.
Karupaiah, T; Sundram, K. Effects of stereospecific positioning of fatty acids in
triacylglycerol structures in native and randomized fats: a review of their nutritional
implications. Nutr Metab, 2007; 4(16): 117.
Cheong, LZ; Zhang, H; Xu, Y; Xu, X. Physical characterization of lard partial
acylglycerols and their effects on melting and crystallization properties of blends with
rapeseed oil. J Agric Food Chem, 2009; 57: 50207.
Yanty, NAM; Marikkar, JMN; Che Man, YB; Long, K. Composition and thermal
analysis of lard stearin and lard olein. J Oil Sci, 2011; 60: 3338.
Haug, A; Hostmark, AT; Harstad, OM. Bovine milk in human nutrition a review. Lip
Health Dis, 2007; 6(25): 116.
Kennelly, JJ. The fatty acid composition of milk fat as influenced by feeding oilseeds.
Anim Feed Sci Technol, 1996; 60: 13752.
Kontkanen, H; Rokka, S; Kemppinen, A; Miettinen, H; Hellstrm, J; Kruus, K; et al.
Enzymatic and physical modification of milk fat: A review. Int Dairy J, 2011; 21: 3
13.
Straarup, EM; Lauritzen, L; Faerk, J; Hoy, C-E; Michaelsen, KF. The Stereospecific
Triacylglycerol Structures and Fatty Acid Profiles of Human Milk and Infant Formulas.
J Pediatr Gastroenterol Nutr, 2006; 42(3): 2939.
Yaron, S; Shachar, D; Abramas, L; Riskin, A; Bader, D; Litmanovitz, I; et al. Effect of
High -Palmitate Content in Infant Formula on the Intestinal Microbiota of Term
Infants. J Pediatr Gastroenterol Nutr, 2013; 56(4): 37681.
Innis, SM; Nelson, CM. Dietary triacyglycerols rich in sn-2 palmitate alter postprandial lipoprotein and unesterified fatty acids interm infants. Prostaglandins Leukot
Essent FattyAcids, 2013; 89: 14551.
Robles, A; Jimnez, MJ; Esteban, L; Gonzlez, PA; Martn, L; Rodrguez, A; et al.
Enzymatic production of human milk fat substitutes containing palmitic and
docosahexaenoic acids at sn-2 position and oleic acid at sn-1,3 positions. Food Sci
Technol, 2011; 44: 198692.
Facioli, NL; Gonalves, LAG. Modificao por via enzimtica da composio
triglicerdica do leo de pequi (Caryocar brasiliense Camb). Qumica Nova, 1998;
21(1): 169.
Segall, SD; Artz, WE; Raslan, DS; Ferraz, VP; Takahashi, JA. Triacylglycerol analysis
of pequi (Caryocar brasiliensis Camb.) oil by electrospray and tandem mass
spectrometry. J Sci Food Agric, 2006; 86: 44552.
Vera, R; Souza, ERB; Fernandes, EP; Naves, RV; Soares Jnior, MS; Caliari, M; et al.
Caracterizao fsica e qumica de frutos do pequizeiro (Caryocar brasiliense Camb.)
oriundos de duas regies no Estado de Gois, Brasil. Pesq Agropec Trop, 2007; 37(2):
939.

General Aspects of Palmitic Acid

97

[69] Arvalo-Pinedo, A; Maciel, VBV; Carvalho, KM; Coelho, AFS; Giraldo-Zuiga, AD;
Arvalo, ZDS; et al. Processamento e estudo da estabilidade de pasta de pequi
(Caryocar brasiliense). Cinc Tecnol Alim, 2010; 30(3): 6648.
[70] Mariano-da-Silva, S; Brait, JDA; Faria, FP; Silva, SM; Oliveira, SL; Braga, PF; et al.
Chemical characteristics of pequi fruits (Caryocar brasiliense Camb.) native of three
municipalities in the State of Gois Brazil. Cinc Tecnol Alim, 2009; 29(4): 7717.
[71] Arajo, FD. A review of Caryocar brasiliense (Caryocaceae): an economically
valuable species of the central Brazilian cerrados. Econ Bot, 1995; 49: 408.
[72] Kakuda, Y; Jahaniaval, F; Marcone, MF; Ontevirgen, LM; Montevirgen, Q.; Umali, U.
Characterization of pili nut (Canarium ovatum) oil: Fatty acid and triacylglycerol
composition and physicochemical properties. J Am Oil Chem Soc, 2000; 77(9): 9917.
[73] Gallegos, RKB; Suministrado, DC; Amongo, RMC; Madlangbayan, MS. Some
Physical and Mechanical Properties of Pili (Canarium ovatum Engl. cv. Katutubo) Nut
as a Function of Nut Moisture Content. Philipp Agric Scientist, 2013; 96(1): 6674.
[74] Zarinah, Z; Maaruf, AG; Nazaruddin, R; Wong, WWW; Xuebing, X. Extraction and
determination of physico-chemical characteristics of Pili nut oil. Int Food Res J, 2014;
21(1): 297301.
[75] Rodrigues, JN; Gioielli, LA. Chemical interesterification of milkfat and milkfat-corn
oil blends. Int Food Res, 2003; 36: 14959.
[76] Idris, NA; Dian LHM. Interesterified palm products as alternatives to hydrogenation.
Asia Pac J Clin Nutr, 2005; 14: 396401.
[77] Berry, SEE. Triacylglycerol structure and interesterification of palmitic and stearic
acid-rich fats: an overview and implications for cardiovascular disease. Nutr Res Rev,
2009; 22: 317.
[78] Ahmadi, L; Wright, AJ; Marangoni, AG. Structural and mechanical behavior of
tristearin/triolein-rich mixtures and the modification achieved by interesterification.
Food Biophys, 2009; 4: 6476.
[79] Costalez-Rodriguez, R; Gibon, V; Verhe, R; De Greyt, W. Chemical and enzymatic
interesterication of a blend of palm stearin: soybean oil for low trans-Margarine
formulation. J Am Oil Chem Soc, 2009; 86: 68197.
[80] Aini, IN; Miskandar, MS. Utilization of palm oil and palm oil products in shortenings
and margarines. Eur J Lipid Sci Technol, 2007; 104: 42232.
[81] Sato, K; Ueno, S. Crystallization, transformation and microstructures of polymorphic
fats in colloidal dispersion states. Curr Opin Colloid Interface Sci, 2011; 16: 38490.
[82] Sato, K. Crystallization behaviour of fats and lipids a review. Chem Eng Sci, 2001;
56(7): 225565.
[83] Bloomer, S; Adlercreutz, P; Mattiasson, B. Triglyceride interesterification by lipases. I.
Cocoa butter equivalents from Fraction Palm Olein (FPO). J Am Oil Chem Soc, 1990;
67: 51924.
[84] Undurraga, D; Markovits; Erazo, S. Cocoa butter equivalent through
enzymicinteresterification of palmoilmidfraction. Process Biochem, 2001; 36(10): 933
9.
[85] Maduco, CO; Akoh, CC; Park, YW. Enzymatic interesterification of tripalmitin with
vegetable oil blends for formulation of caprine milk infant formula analogs [J]. J Dairy
Sci, 2007; 90: 594601.

98

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

[86] Yadong, Z; Guicheng, H. Advance on Production of Human Milk Fat Substitutes


(HMFS). J Northeast Agric Univ (English Edition), 2011; 18(2): 926.
[87] Alles, MS; Scholtens, PAMJ; Bindels, JG. Current trends in the composition of infant
milk formulas. Curr Paediatr, 2004; 14: 5163.
[88] Ilyasoglu, H; Gultekin-Ozguven, M; Ozcelik, B. Production of human milk fat
substitute with medium-chain fatty acids by lipasecatalyzed acidolysis: Optimization
by response surface methodology. Food Sci Technol, 2011; 44: 9991004.
[89] Jensen, C; Buist, NRM; Wilson, T. Absorption of individual fatty acids from Long
chain or medium chain triglycerides in very small infants. Am J Clin Nutr, 1986; 43:
74551.
[90] Leite, MEQ; Lasekan, J; Baggs, G; Ribeiro, T; Menezes-Filho, J; Pontes, M; et al.
Calcium and fat metabolic balance, and gastrointestinal tolerance in term infants fed
milk-based formulas with and without palm olein and palm kernel oils: a randomized
blinded crossover study. BMC Pediatr, 2013; 13(215): 19.
[91] Lien, EL. The role of fatty acid positional distribution infants. J Pediatr, 1994; 125:
S628.
[92] Lopez, A; Castellote, AI; Campoy, C; Rivero, M; Tormo, R; Infante, D; et al. The
influence of dietary palmitic acid triacylglyceride position on the fatty acid, calcium
and magnesium contents of at term newborn faeces. Early Hum Dev, 2001; 65: S83
94.
[93] Quinlan, PT; Lockton, S; Irwin, J; Lucas, AL. The relationship between stool hardness
and stool composition in breastand formula-fed infants. J Pediatr Gastroenterol Nutr,
1995; 20(1): 8190.
[94] Turan, D; Yeilubuk, N; Akoh, CC. Enrichment of sn-2 position of hazelnut oil with
palmitic acid: Optimization by response surface methodology. Food Sci Technol, 2013;
50: 76672.
[95] Fatima, A; Lisa, S; Shahida, H; Hubert, K; Nousheen, Z. De novo lipogenesis in health
and disease. Metab, 2014. DOI: 10.1016/j.metabol.2014.04.003.
[96] Haag, M; Dippenaar, NG. Dietary fats, fatty acids and insulin resistance: short review
of a multifaceted connection. Med Sci Monit, 2005; 11(12): RA359-67.
[97] Turner, N; Cooney, GJ; Kraegen, EW; Bruce, CR. Fatty acid metabolism, energy
expenditure and insulin resistance in muscle. J Endocrinol, 2014; 220: T6179.
[98] Berg, JM; Tymoczko, JL; Stryer, L. Biochemistry. 5th edition. New York: W H
Freeman; 2002.
[99] Harvey, R; Ferrier, D. Biochemistry. 5th edition. Philadelphia, Pa, USA: Wolters
Kluwer/Lippincott Williams & Wilkins; 2011.
[100] Minrik, P; Tomkov, N; Kollrov, M; Antalk, M. Malate Dehydrogenases
Structure and Function. Gen Physiol Biophys, 2002; 21: 25765.
[101] Saggerson, D. Malonyl-CoA, a key signaling molecule in mammalian cells. Annu Rev
Nutr, 2008; 28: 25372.
[102] Kim, KH. Regulation of mammalian acetyl-coenzyme A carboxylase. Annu Rev Nutr,
1997; 17: 7799.
[103] Tong, L; Harwood Jr, HJ. Acetyl-Coenzyme A Carboxylases: Versatile Targets for
Drug Discovery. J Cell Biochem, 2006; 99: 147688.
[104] Wakil, SJ; Stoops, JK; Joshi, VC. Fatty acid synthesis and its regulation. Ann Rev
Biochem, 1983; 52: 53779.

General Aspects of Palmitic Acid

99

[105] Smith, S. The animal fatty acid synthase: one gene, one polypeptide, seven enzymes.
FASEB J, 1994; 8: 124859.
[106] Wakil, SJ. Fatty Acid Synthase, A Proficient Multifunctional Enzyme. Biochem, 1989;
28(11): 452330.
[107] Smith, S; Witkowski, A; Joshi, AK. Structural and functional organization of the
animal fatty acid synthase. Prog Lipid Res, 2003; 42: 289317.
[108] Guillou, H; Zadravec, D; Martin, PGP; Jacobsson, A. The key roles of elongases and
desaturases in mammalian fatty acid metabolism: Insights from transgenic mice. Prog
Lipid Res, 2010; 49: 18699.
[109] Jakobsson, A; Westerberg, R; Jacobsson, A. Fatty acid elongases in mammals: Their
regulation and roles in metabolism. Prog Lipid Res, 2006; 45: 23749.
[110] Moon, YA; Horton, JD. Identification of two mammalian reductases involved in the
two-carbon fatty acyl elongation cascade. J Biol Chem, 2003; 278: 733543.
[111] Leonard, AE; Pereira, SL; Sprecher, H; Huang, Y-S. Elongation of long-chain fatty
acids. Prog Lipid Res, 2004; 43: 3654.
[112] Hodson, L; Fielding, BA. Stearoyl-CoA desaturase: rogue or innocent bystander? Prog
Lipid Res, 2013; 52: 1542.
[113] Bartlett, K; Eaton, S. Mitochondrial beta-oxidation. Eur J Biochem, 2004; 271: 4629.
[114] Ghisla, S. -Oxidation of fatty acids. Eur J Biochem, 2004; 271: 45961.
[115] Kompare, M; Rizzo, WB. Mitochondrial Fatty-Acid Oxidation Disorders. Semin
Pediatric Neurol, 2008; 15(3): 1409.
[116] Eaton, S. Control of mitochondrial beta-oxidation flux. Prog Lipid Res, 2002; 41: 197
239.
[117] Houten, SM; Wanders, RJA. A general introduction to the biochemistry of
mitochondrial fatty acid -oxidation. J Inherit Metab Dis, 2010; 33: 46977.
[118] Kazantzis, M; Stahl, A. Fatty acid transport proteins, implications in physiology and
disease. Biochim Biophys Acta - Mol Cell Biol Lipids, 2012; 1821(5): 8527.
[119] Thumser, AE; Moore, JB; Plant, NJ. Fatty acid binding proteins: tissue-specific
functions in health and disease. Curr Opin Clin Nutr Metab Care, 2014; 17: 1249.
[120] Meunier-Durmort, C; Poirier, H; Niot, I; Forest, C; Besnard, P. Up-regulation of the
expression of the gene for liver fatty acid-binding protein by long-chain fatty acids.
Biochem J, 1996; 31: 4837.
[121] Smith, BK; Jain, SS; Rimbaud, S; Dam, A; Quadrilatero, J; Ventura-Clapier, R; et al.
FAT/CD36 is located on the outer mitochondrial membrane, upstream of long-chain
acyl-CoA synthetase, and regulates palmitate oxidation. Biochem J, 2011; 437: 12534.
[122] Clarke, DC; Miskovic, D; Han, X-X; Calles-Escandon, J; Glatz, JF; Luiken, JJ; et al.
Overexpression of membrane associated fatty acid binding protein (FABPpm) in vivo
increases fatty acid sarcolemmal transport and metabolism. Physiol Genomics, 2004;
17 (1): 317.
[123] Luiken, JJ; Turcotte, LP; Bonen, A. Protein-mediated palmitate uptake and expression
of fatty acid transport proteins in heart giant vesicles. J Lipid Res, 1999; 40: 100716.
[124] Memon, RA; Fuller, J; Moser, AH; Smith, PJ; Grunfeld, C; Feingold, KR. Regulation
of putative fatty acid transporters and Acyl-CoA synthetase in liver and adipose tissue
in ob/ob mice. Diabetes, 1999; 48(1): 1217.

100

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

[125] Schwenk, RW; Holloway, GP; Luiken, JJFP; Bonen, A; Glatz, JFC. Fatty acid
transport across the cell membrane: Regulation by fatty acid transporters.
Prostaglandins Leukot Essent Fatty Acids, 2010; 82(46): 14954.
[126] Reuter, SE; Evans, AM; Carnitine and Acylcarnitines. Clin pharmacokinet, 2012;
51(9), 94167.
[127] Indiveri, C; Iacobazzi, V; Tonazzi, A; Giangregorio, N; Infantino, V; Convertini, P; et
al. The mitochondrial carnitine/acylcarnitine carrier: Function, structure and
physiopathology. Mol Aspects Med, 2011; 32: 22333.
[128] Ramsay, RR; Gandour, RD; Van Der Leij, FR. Molecular enzymology of carnitine
transfer and transport. Biochim Biophys Acta, 2001; 1546: 2143.
[129] Schreurs, M; Kuipers, F; Van Der Leij, FR. Regulatory enzymes of mitochondrial oxidation as targets for treatment of the metabolic syndrome. Obes Rev, 2010; 11(5):
3808.
[130] Reddy, BV; Prasad, BR; Sinha, SN; Ahmed, N. New mathematical derivations for
calculation of ATP yield due to the complete oxidation of different types of fatty acids.
Indian J Biochem Biophys, 2014; 51(1), 527.
[131] Eaton, S; Bartlett, K; Pourfarzam, S. Mammalian mitochondrial -oxidation. Biochem
J, 1996; 320: 34557.
[132] Fukao, T; Lopaschuk, GD; Mitchell, GA. Pathways and control of ketone body
metabolism: on the fringe of lipid biochemistry. Prostaglandins Leukot Essent Fatty
Acids, 2004; 70: 24351.
[133] Laffel, L. Ketone bodies: a review of physiology, pathophysiology and application of
monitoring to diabetes. Diabetes Metab Res Rev, 1999; 15(6): 41226.
[134] Mitchell, GA; Kassovska-Bratinova, S; Boukaftane, Y, Robert, MF; Wang, SP;
Ashmarina, L; Lambert, M; et al. Medical aspects of ketone body metabolism. Clin
Invest Med, 1995; 18: 193216.
[135] White, H; Venkatesh, B. Clinical review: Ketones and brain injury. Crit Care, 2011;
15(219): 110.
[136] Cooper, AD. Hepatic uptake of chylomicron remnants. J Lipid Res, 1997; 38: 217392.
[137] Academy of Nutrition and Dietetics (the Academy). Position of the Academy of
Nutrition and Dietetics: Dietary Fatty Acids for Healthy Adults. J Acad Nutr Diet,
2014; 114: 13653.
[138] Keys, A; Anderson, JT; Grande, F. Serum cholesterol response to changes in the diet:
IV. Particular saturated fatty acids in the diet. Metab, 1965; 14: 77686.
[139] Hegsted, DM; McGandy, RB; Myers, ML; Stare, FJ. Quantitative effects of dietary fat
on serum cholesterol in man. Am J Clin Nutr, 1965; 17: 28195.
[140] Grundy, SM; Denke, MA. Dietary influences on serum lipids and lipoproteins. J Lipid
Res, 1990; 31: 114972.
[141] Khosla, P; Hayes, KC. Cholesterolaemic effects of the saturated fatty acids of palm oil.
Food Nutr Bull, 1994; 15(2): 72.
[142] Wai, TNK. A critical review of the cholesterolaemic effects of palm oil. Food Nutr
Bull, 1994.
[143] Bonanome, A; Grundy, SM. Effect of dietary stearic acid on plasma cholesterol and
lipoprotein levels. N Engl J Med, 1988; 318: 12448.
[144] Grundy, SM; Vega, GL. Plasma cholesterol responsiveness to saturated fatty acids. Am
J Clin Nutr, 1988; 47: 8224.

General Aspects of Palmitic Acid

101

[145] Mattson, EH; Grundy, SM. Comparison of effects of dietary saturated,


monounsaturated, and polyunsaturated fatty acids on plasma lipids and lipoproteins in
man. J Lipid Res, 1985; 26: 194-202.
[146] Sun, Y; Neelakantan, N; Wu, Y; van Dam, RM. Poster Abstract Presentations Session
Title: Nutrition 1 Abstract P065: Effects of Palm Oil Consumption on Blood Lipids: A
Meta-Analysis of Clinical Trials. Circulation, 2014; 129: AP065.
[147] Qureshi, AA; Burgerl, WC; Peterson, DM; Elson, CE. The Structure of an Inhibitor of
Cholesterol Biosynthesis Isolated from Barley. J Biol Chem, 1986; 261(23): 1054450.
[148] Hayes, KC; Khosla, P. The complex interplay of palm oil fatty acids on blood lipids.
Eur J Sci Technol, 2007; 109: 45364.
[149] Park, E-J; Lee, AY; Park, S; Kim, J-H; Cho, M-H. Multiple pathways are involved in
palmitic acid-induced toxicity. Food Chem Toxicol, 2014; 67: 2634.
[150] Birkenfeld, AL; Shulman, GI. Nonalcoholic Fatty Liver Disease, Hepatic Insulin
Resistance, and Type 2 Diabetes. Hepatol, 2014; 59(2): 71323.
[151] Matteoni, C; Younossi, ZM; McCullough, A. Nonalcoholic fatty liver disease: a
spectrum of clinical pathological severity. Gastroenterol, 1999; 116: 14139.
[152] Marra, F; Gastaldelli, A; Baroni, GS; Tell, G; Tiribelli, C. Molecular basis and
mechanisms of progression of non-alcoholic steatohepatitis. Trends Mol Med, 2008;
14(2): 7281.
[153] Hernandez, R; Martinez-Lara, E; Canuelo, A; del Moral, ML; Blanco, S; Siles, E; et al.
Steatosis recovery after treatment with a balanced sunflower or olive oil-based diet:
involvement of perisinusoidal stellate cells. World J Gastroenterol, 2005; 11: 74805.
[154] Musso, G; Gambino, R; De Michieli, F; Cassader, M; Rizzetto, M; Durazzo, M; et al.
Dietary habits and their relations to insulin resistance and postprandial lipemia in
nonalcoholic steatohepatitis. Hepatol, 2003; 37: 90916.
[155] Nehra, V; Angulo, P; Buchman, AL; Lindor, KD. Nutritional and metabolic
considerations in the etiology of non-alcoholic steatohepatitis. Dig Dis Sci, 2001; 46:
234752.
[156] Donnelly, KL; Smith, CI; Schwarzenberg, SJ; Jessurun, J; Boldt, MD; Parks, EJ.
Sources of fatty acids stored in liver and secreted via lipoproteins in patients with
nonalcoholic fatty liver disease. J Clin Invest, 2005; 115(5): 134351.
[157] Albano, E; Mottaran, E; Occhino, G; Reale, E; Vidali, M. Review article: role of
oxidative stress in the progression of non-alcoholic steatosis. Aliment Pharmacol Ther,
2005; 22(Suppl s2): 7173.
[158] Koek, GH; Liedorp, PR; Bast, A. The role of oxidative stress in non-alcoholic
steatohepatitis. Clin Chim Acta, 2011, 412(1516): 1297305.
[159] Toyokuni, S. Reactive oxygen species-induced molecular damage and its application in
pathology. Pathol Int, 1999; 49: 91102.
[160] Tariq, Z; Green, CJ; Hodson, L. Are oxidative stress mechanisms the common
denominator in the progression from hepatic steatosis towards non-alcoholic
steatohepatitis (NASH)? [online]. Liver Int, 2014 [cited 2014 Apr 02]. DOI:
10.1111/liv.12523.
Available
from:
URL:
http://onlinelibrary.wiley.com
/doi/10.1111/liv.12523/full.
[161] Day, CP; James, OF. Steatohepatitis: a tale of two hits? Gastroenterol, 1998;
114: 8425.

102

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

[162] Tilg, H; Moschen, AR. Evolution of inflammation in nonalcoholic fatty liver disease:
the multiple parallel hits hypothesis. Hepatol, 2010; 52: 183646.
[163] Ji, J; Zhang, L; Wang, P; Mu, Y-M; Zhu, X-Y; Wu, Y-Y; et al. Saturated free fatty
acid, palmitic acid, induces apoptosis in fetal hepatocytes in culture. Exp Toxicol
Pathol, 2005; 56: 36976.
[164] Unger, R; Orci, L. Lipoapoptosis: its mechanism and its diseases. Biochim Biophys
Acta, 2002; 1585: 20212.
[165] Choi, SE; Lee, SM; Lee, YJ; Li, LJ; Lee, SJ; Lee, JH; et al. Protective role of
autophagy in palmitate-induced INS-1 betacell death. Endocrinol, 2009; 150(1): 126
34.
[166] Rakatzi, I; Mueller, H; Ritzeler, O; Tennagels, N; Eckel, J. Adiponectin counteracts
cytokine- and fatty acid-induced apoptosis in the pancreatic beta-cell line INS-1.
Diabetologia, 2004; 47: 24958.
[167] Lakhani, SA; Masud, A; Kuida, K; et al. Caspases 3 and 7: key mediators of
mitochondrial events of apoptosis. Sci, 2006; 311(5762): 84751.
[168] Renault, TT; Manon, S. Bax: addressed to kill. Biochimie, 2011; 93(9): 137991.
[169] Gross, A; McDonnell, JM; Korsmeyer, SJ. BCL-2 family members and the
mitochondria in apoptosis. Genes Dev, 1999; 13: 1899911.
[170] Feldstein, AE; Canbay, A; Guicciardi, ME; Higuchi, H; Bronk, SF; Gores, GJ. Diet
associated hepatic steatosis sensitizes to Fas mediated liver injury in mice. J Hepatol,
2003; 39: 97883.
[171] Joshi-Barve, S; Barve, SS; Amancherla, K; Gobejishvili, L; Hill, D; Cave, M; et al.
Palmitic Acid Induces Production of Proinflammatory Cytokine Interleukin-8 from
Hepatocytes. Hepatol, 2007; 46: 82330.
[172] Donath, MY; Shoelson, SE. Type 2 diabetes as an inflammatory disease. Nat Rev,
2011; 11: 98107.
[173] Yorek, MA. Oxidative Stress and Diabetes-Induced Vascular Dysfunction: Role in
Diabetic Neuropathy. Oxidative Stress Appl Basic Res Clin Pract, 2014: 112.
[174] American Diabetes Association. Diagnosis and Classification of Diabetes Mellitus
American Diabetes Association. Diabetes Care, 2010; 33(Suppl 1): S62-9.
[175] Poitout, V; Robertson, RP. Minireview: Secondary beta-cell failure in type 2 diabetes
a convergence of glucotoxicity and lipotoxicity. Endocrinol, 2002; 143(2): 33942.
[176] Fonseca, SG; Gromada, J; Urano, F. Endoplasmic reticulum stress and pancreatic b-cell
death. Trends Endocrinol Metab, 2011; 22: 26674.
[177] Isfort, M; Stevens, SCW; Schaffer, S; Jong, CJ; Wold, LE. Metabolic dysfunction in
diabetic cardiomyopathy. Heart Fail Rev, 2014; 19: 3548.
[178] Rosen, P; Nawroth, PP; King, G; Moller, W; Tritschler, HJ; Packer, L. The role of
oxidative stress in the onset and progression of diabetes and its complications: a
summary of a Congress Series sponsored by UNESCOMCBN, the American Diabetes
Association, and the German Diabetes Society. Diabetes Metab Res Rev, 2001; 17:
189212.
[179] Stentz, FB; Umpierrez, GE; Cuervo, R; Kitabchi, AE. Proinflammatory cytokines,
markers of cardiovascular risks, oxidative stress, and lipid peroxidation in patients with
hyperglycemic crises. Diabetes, 2004; 53: 207986.

General Aspects of Palmitic Acid

103

[180] Mohamed, AK; Bierhaus, A; Schiekofer, S; Tritschler, H; Ziegler, R; Nawroth, PP. The
role of oxidative stress and NF-kappaB activation in late diabetic complications.
Glucose or diabetes activates. Biofactors, 1999; 10(2-3): 15767.
[181] Patel, S; Santani, D. Role of NF-kappa B in the pathogenesis of diabetes and its
associated complications. Pharmacol Rep, 2009; 61(4): 595603.
[182] Sharma, K; Ziyadeh, FN. Hyperglycemia and diabetic kidney disease: the case for
transforming growth factor-b as a key modulator. Diabetes, 1995; 44: 113946.
[183] Igarashi, M; Wakasaki, H; Takahara, N; Ishii, H; Jiang, Z-Y; Yamauchi, T; et al.
Glucose or diabetes activates p38 mitogen-activated protein kinase via different
pathways. J Clin Invest, 1999; 103(2): 18595.
[184] Koya, D; King, GL. Protein kinase C activation and the development of diabetic
complications. Diabetes, 1998; 47(6): 85966.
[185] Kawai, T; Akira, S. The roles of TLRs, RLRs and NLRs in pathogen recognition. Int
Immunol, 2009; 21: 31737.
[186] Eguchi, K; Manabe, I; Oishi-Tanaka, Y; Ohsugi, M; Kono, N; Ogata, F; et al. Saturated
Fatty Acid and TLR Signaling Link -Cell Dysfunction and Islet Inflammation. Cell
Metab, 2012; 15: 51833.
[187] Kaneto, H; Xu, G; Fujii, N; Kim, S; Bonner-Weir, S; Weir, GC. Involvement of c-Jun
N-terminal Kinase in Oxidative Stress-mediated Suppression of Insulin Gene
Expression. J Biol Chem, 2002; 277(33): 300108.
[188] Lee, YH; Giraud, J; Davis, RJ; White, MF. c-Jun N-terminal Kinase (JNK) Mediates
Feedback Inhibition of the Insulin Signaling Cascade. J Biol Chem, 2003; 278: 2896
902.
[189] Valen, G; Yan, Z; Hansson, GK. Nuclear Factor Kappa-B and the Heart. J Am Coll
Cardiol, 2001; 38(2): 30714.
[190] Cross, TG; Scheel-Toellner, D; Henriquez, NV; Deacon, E; Salmon, M; Lord, JM.
Serine/Threonine Protein Kinases and Apoptosis. Exp Cell Res, 2000; 256: 3441.
[191] Su, B; Karin, M. Mitogen-activated protein kinase cascades and regulation of gene
expression. Curr Opinion Immunol, 1996; 8: 40211.
[192] Hirosumi, J; Tuncman, G; Chang, L; Grgn, CZ; Uysal, KT; Maeda, K; et al. A
central role for JNK in obesity and insulin resistance. Nature, 2002; 420(21): 3336.
[193] Takeda, K; Kaisho, T; Akira, S. Toll-like receptors. Annu Rev Immunol, 2003; 21: 335
76.
[194] Cnop, M; Hannaert, JC; Hoorens, A; Eizirik, DL; Pipeleers, DG. Inverse Relationship
Between Cytotoxicity of Free Fatty Acids in Pancreatic Islet Cells and Cellular
Triglyceride Accumulation. Diabetes, 2001; 50(8): 17717.
[195] Jacqueminet, S; Briaud, I; Rouault, C; Reach, G; Poitout, V. Inhibition of insulin gene
expression by long-term exposure of pancreatic cells to palmitate is dependent on the
presence of a stimulatory glucose concentration. Metab, 2000; 49(4): 5326.
[196] Boden, G. Obesity, Insulin Resistance and Free Fatty Acids. Curr Opin Endocrinol
Diabetes Obes, 2011; 18(2): 13943.
[197] Jov, M; Planavila, A; Laguna, JC; Vazquez-Carrera M. Palmitateinduced interleukin 6
production is mediated by protein kinase C and nuclear-factor kB activation and leads
to glucose transporter down-regulation in skeletal muscle cells. Endocrinol, 2005; 146:
308795.

104

D. Teixeira de Almeida, M. Melo Costa and S. Feitosa

[198] Jov, M; Planavila, A; Snchez, RM; Merlos, M; Laguna, JC; Vzquez-Carrera, M.


Palmitate Induces Tumor Necrosis Factor- Expression in C2C12 Skeletal Muscle
Cells by a Mechanism Involving Protein Kinase C and Nuclear Factor-B Activation.
Endocrinol, 2006; 147(1): 55261.
[199] Itani, SI; Ruderman, NB; Schmieder, F; Boden, G. Lipid-induced insulin resistance in
human muscle is associated with changes in diacylglycerol, protein kinase C, and
IkappaB-alpha, Diabetes, 2002; 51: 200511.
[200] Weigert, C.; Brodbeck, K; Staiger, H; Kausch, C; Machicao, F; Haring, HU; et al.
Palmitate, but not unsaturated fatty acids, induces the expression of interleukin-6 in
human myotubes through proteasome dependent activation of nuclear factor-kappaB. J
Biol Chem, 2004; 279: 2394252.
[201] Jiang, H; Liang, C; Liu, X; Jiang, Q; He, Z; Wu, J; et al. Palmitic acid promotes
endothelial progenitor cells apoptosis via p38 and JNK mitogen-activated protein
kinase pathways Atherosclerosis, 2010; 210: 717.
[202] Pitocco, D; Zaccardi, F; Di Stasio, E; Romitelli, F; Santini, SA; Zuppi, C; et al.
Oxidative Stress, Nitric Oxide, and Diabetes. Rev Diabet Stud, 2010; 7(1): 1525.
[203] Reynoso, R; Salgado, LM; Caldern, V. High levels of palmitic acid lead to insulin
resistance due to changes in the level of phosphorylation of the insulin receptor and
insulin receptor substrate-1. Vascular Biochem Mol Cell Biochem: Int J Chem Biol
Health Disease, 2003; 41: 15562.

In: Palmitic Acid: Occurrence, Biochemistry and Health Effects ISBN: 978-1-63321-519-1
Editor: Lucas F. Porto
2014 Nova Science Publishers, Inc.

Chapter 5

PALMITIC ACID AS
A CARDIOMETABOLIC RISK FACTOR
Danijela Risti-Medi* and Vesna Vui
Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical
Research, University of Belgrade, Belgrade, Serbia

ABSTRACT
Current dietary recommendations are based on a reduced saturated fatty acid (SFA)
consumption to prevent cardiovascular disease (CVD). The role of individual SFA in
metabolic disease is not fully understandable. One type of SFA present in many common
foods (dairy, meat, palm and coconut oil) is palmitic acid (16:0). A number of
epidemiological studies have shown that the populations who consume large amounts of
atherogenic SFA (especially palmitic, myristic, lauric) have elevated levels of LDL and
HDL-cholesterol. Saturated fatty acid exert their atherogenic and thrombogenic effect
through increased production of LDL, very-low-density lipoproteins particles and
apolipoproteins A1, with a decrease of LDL- receptors specific activity, and an increase
in platelet aggregation. The total cholesterol/ HDL-cholesterol ratio, the best overall
indication of potential effects on coronary heart disease (CHD) risk is nonsignificantly
affected by consumption of palmitic acid (PA). Compared with lipid effects, the influence
of SFA intake on inflammation markers is less well explored. The associations between
circulating and tissue PA and dietary intake of PA are diverse and most likely reflecting
endogenous metabolism. Status of PA is not in intakeresponse relationship biomarker,
probably partly due to conversion of 16:0 to 16:1 by steaoryl-CoA-desaturase (SCD-1).
Increased SFA intake has been associated with increased SCD-1 activity in which may
predict mortality. Palmitoylation is the process involved in proteinmembrane
interactions and signal transduction. Increases in dietary intake of PA decrease fat
oxidation and daily energy expenditure with slight increases in adiposity. Evidence for
the effects of SFA, particularly PA consumption on insulin resistance, vascular function,
type 2 diabetes, and stroke is various. It is considered that circulating PA, as nonesterified
*

Corresponding author: Danijela Risti-Medi, MD. PhD. Institute for Medical Research, Centre of Research
Excellence in Nutrition and Metabolism, University of Belgrade, Tadeusa Koscuska 1, 11129 Belgrade,
Serbia. Tel:+38111303-1997; Fax: +381 11 2030-169; e-mail: dristicmedic@gmail.com.

106

Danijela Risti-Medi and Vesna Vui


fatty acids stimulate insulin resistance by decreasing phosphorylation of the insulin
receptor and insulin receptor substrate-1. In muscle cells, PA decrease oxidation of fatty
acids and glucose which elevates fatty acid and glucose levels in tissues and blood, and
decreases adiponectin production, which may both promote insulin resistance. It was
shown that 16:0 and 14:0 stimulate -cells and endothelial dysfunction. The incidence of
type 2 diabetes was associated with total SFA levels of plasma cholesterol esters (also
demonstrated for 16:0 levels independently) and phospholipids (also for 16:0 and 18:0).
In skeletal muscle phospholipids, PA has been negatively associated with insulin
sensitivity and diabetes type 2. Systematic reviews on prospective cohort studies
indicated that CHD risk has not been directly associated with SFA intake, although is
associated with a dietary habits, high in SFA-rich foods. Taken together, there is
collective convincing evidence for decreased CHD risk when replacing SFA with
polyunsaturated fats. Differences in cardiometabolic risk appear greater between food
groups and overall dietary patterns rather than between separate SFA.

INTRODUCTION
Previously, low fat intakes were traditionally recommended in the prevention of
cardiovascular disease (CVD) as a component of a health promoting diet, without much
attention to the quality of fat. However, current dietary guidelines generally put more
emphasis on the quality of fat [1-4]. Imbalances in the amounts of individual fatty acids in the
diet may have an impact on the occurrence of dyslipidemia, atherosclerosis, thrombosis,
hypertension and obesity. Saturated fatty acids (SFA) have shown to be particularly important
for development of the above mentioned diseases. However, in spite of an increasing body of
new data, the role of individual dietary SFA in metabolic diseases is not fully clarified (Micha
2010). The reachest dietary sources of SFA include fast foods, processed foods, high-fat dairy
products, red meats, and pork [1,5].
One of the most abundant SFA in many common foods (dairy, meat, palm and coconut
oil) is palmitic acid (PA, 16:0). The amount of PA is the highest in palm oil (around 50%),
but significant amounts of PA (25-26%) can also be found in butter, chicken fat, lard, beef
and lamb fat, as well as in cocoa butter. Even olive oil, which is one of basic components of
the healthy Mediterranean diet contains around 16% of PA [6]. Furthermore, PA is present in
human milk with 20-25% of total fats. Overall, PA and stearic acid (18:0) are the most
common dietary SFA and therefore they are also the major SFA in human plasma and tissues.
Their concentration in serum/plasma phospholipids and cholesterol esters reflect dietary high
fat intake.
Dietary saturated fats are of particular scientific interest because of their association with
CVD. In some countries, e.g. in Finland, there has been a decline in coronary heart disease
(CHD) mortality along with the decreased intake of saturated and total fats [7]. Some
epidemiological studies showed that total dietary fats intake is positively associated with
metabolic syndrome [8-11]. De Oliveira et al [11] have recently reported that saturated fat
intake greater than 10% of total caloric value represented a double risk for metabolic
syndrome diagnosis, with odds ratio (OR) 2.0 (1.04-3.84). This association is mostly
attributed to palmitic acid, due to the fact that excessive intake of PA increases the visceral
adipose tissue in greater proportion than other fat types [12]. Metabolic syndrome or
cardiometabolic risk refers to a cluster of metabolic abnormalities including disturbances in

Palmitic Acid As a Cardiometabolic Risk Factor

107

glucose and insulin metabolism, central obesity, dyslipidemia (high triglyceride levels, low
HDL-cholesterol and high levels of small dense LDL-particles) and hypertension [13,14].
Central to the etiology of metabolic syndrome is an interrelated triad comprising
inflammation, obesity (particularly abdominal), and aberrations in fatty acid metabolism
[15,16].

PALMITIC ACID INTAKE, ADIPOSITY AND INFLAMMATION


Low-grade systemic inflammation is an important component of most chronic noncommunicable diseases, including metabolic syndrome, CVD, diabetes type 2 and cancer
[17]. Although polyunsaturated fatty acids n-3 and n-6 series play dominant role in
inflammation, as shown by several papers (review by Ristic-Medic 18), increased serum SFA
has also been closely connected to low grade tissue inflammation. The primary cell-intrinsic
dysfunctions include lipid dysregulation (e.g. accumulation of intracellular diacylglycerols,
SFA, and ceramides) [19]. Studies in vitro indicated direct proinflammatory and pro-oxidant
effects of fatty acids [20,21]. Importantly, these effects were mostly attributed to saturated
fats, with palmitate commonly employed [21]. Mechanisms proposed by Kennedy et al [12]
related to SFA effects which contributed to the cardiometabolic risk include: 1) accumulation
of diacylglycerol (DAG) and ceramide; 2) activation of nuclear factor-kB (NFkB), protein
kinase C (PKC), and mitogen-activated protein kinases (MAPK), and subsequent induction of
inflammatory genes in white adipose tissue (WAT), immune cells, and myotubes; 3)
decreased activation of peroxisome proliferator-activated receptor- (PPAR) coactivator-1
/ and adiponectin production, which decreases the oxidation of glucose and fatty acids; and
4) recruitment of immune cells such as macrophages, neutrophils, and bone marrow-derived
dendritic cells (BMDC) to WAT and muscles.
Overconsumption of SFA (especially PA) enhances WAT expansion and adipocyte
hypertrophy and subsequent death [12]. These processes led to increased inflammatory
signaling and recruitment and activation of macrophages, neutrophils, and bone marrowderived dendritic cell. As a consequence, inflammation, impaired insulin signaling, and
insulin resistance in multiple tissues, particularly in WAT and muscles are occurred
[22,23,24].

Effect of Palmitic Acid on White Adiposity Tissue Function


Therefore, excessive palmitate intake expands WAT, but it also increases inflammation
and apoptosis through oxidative or endoplasmic reticulum stress, generation of ceramide and
reactive oxygen species (ROS), and PKC signaling. Palmitic acid induces cytokine
production in adipocytes, via activation of PKC, NFkB, and MAPK signaling with induced
interleukin-6 (IL-6) and tumor necrosis factor (TNF)- expression [12,25]. It is well known
that SFA activated toll-like receptor (TLR) signaling in murine adipocytes [26,27] and
macrophages [28,29], led to NFkB and c-jun-NH2-terminal kinase (JNK) activation and
cytokine production. Consistent with these data, TLR-4 deficiency selectively protects against
obesity induced by diets rich in palmitic acid [30]. In the conditions where palmitic acid is a

108

Danijela Risti-Medi and Vesna Vui

major component in the diet, palmitate strongly increased the expression and secretion of
TNF and IL-10 in murine 3T3L1 adipocytes, compared with monounsaturated oleic acid
and polyunsaturated docosahexanoic acid (DHA) [31]. It has been considered that in
adipocytes, palmitic acid modulated intracellular signaling and induced endoplasmic
reticulum stress by increasing C/EBP homologous protein and glucose regulatory protein 78.
Furthermore, palmitic acid alters phosphorylation of eIF2 and increases phosphorylation of
JNK and extracellular receptor kinase [32].
Experimental studies [33,34] have shown that high level of palmitic acid in the diet
impaired insulin sensitivity by reducing adiponectin secretion and impairing insulin signaling
pathways required for glucose uptake. Adiponectin is an insulin-sensitizing protein produced
by adipocytes. Reducing the levels of adiponectin appears to be the mechanism by which
palmitate caused insulin resistance in isolated rat adipocytes [35]. In accordance, experiments
in mice fed a high-fat diet showed that overexpression of adiponectin decreased insulin
resistance [36]. Furthermore, PA leads to insulin resistance due to changes of phosphorylation
level of the insulin receptor and insulin receptor substrate.
Saturated fatty acid induced inflammatory response in the interaction between adipocytes
and macrophages by the TLR4/NFkB signaling patway [26]. Supplementation with palmitic
acid induced IP-10 inflammatory gene expression in human macrophages (U937) by an
NFkB-dependent mechanism [37]. It was shown that adipocytes containing SFA have the
capability to activate macrophages to a greater extent than smaller adipocytes. It is even more
pronounced when compared adipocytes containing SFA with adipocytes enriched in
unsaturated fatty acid. For instance, SFA increased TNF mRNA levels in cultures of
adipocytes and murine macrophages, whereas unsaturated fatty acid had no effect. Factors
secreted from macrophages increase adipocyte inflammation and insulin resistance [38,39]
and high-fat feeding in mice increased TLR4 signaling in macrophages and adipocytes and
impaired insulin signaling effects [29].

Effect of Palmitic Acid in Muscle Cells


In muscle cells (for example C2C12) [40,41], palmitic acid also induce inflammation by
increasing the expression and secretion of IL-6 and TNF. Furthermore, PA leads to glucose
transporter 4 down-regulation and impaired insulin sensitivity via an NFkB/PKC pathway.
Palmitate-mediated down regulation of PPAR coactivator-1 (PGC-1) in skeletal muscle
cells involves MEK1/2 and NFkB activation [42]. It is well established that PGC-1 promotes
oxidative phosphorylation, mitochondrial gene expression, and insulin stimulated glucose
uptake [43]. However, PGC-1 expression is reduced in obesity [44]. In addition, higher
palmitic acid intake in C2C12 myotubes, increased p38 MAPK signaling, reducing PGC-1
/1 expression and activity, that would lead to decreased oxidation of fatty acid and glucose,
with increasing their accumulation in tissues and blood. Palmitate also caused the
accumulation of DAG and ceramide and reduced insulin stimulated glucose uptake in murine
L6 myotubes [45]. Taken together, these studies showed that palmitic acid are particularly
potent in recruiting and activating immune cells in WAT, increasing inflammation and
demonstrate the adverse effects of SFA on glucose uptake and utilization in muscle.

Palmitic Acid As a Cardiometabolic Risk Factor

109

DIETARY PA -EFFECTS ON CARDIOVASCULAR RISK FACTORS


Several mechanisms mediate between dietary fatty acid intake and cardiovascular disease
risk. The main mediating mechanism is probably concentration of blood cholesterol [6,
46,47]; whereas other mechanisms are related to insulin resistance, inflammation, and
endothelial function [48,49].

Lipids and Lipoproteins


The quality of dietary fats has been shown to have a significant effect on serum lipid
profile. According to several publications, substituting SFA with unsaturated fats
convincingly decreased concentrations of serum/plasma total and LDL-cholesterol in
randomized control studies (RCTs) [49-57].However, in seven of these studies concentration
of serum/plasma triglycerides did not change. Other serum or plasma lipoproteins were also
affected. Thus consumption of polyunsaturated (PUFA) or monounsaturated fatty acids
(MUFA) in place of SFA leads to lowering of serum/plasma total cholesterol, LDLcholesterol, and ApoB; slight lowering (for PUFA) of HDL-cholesterol and ApoA1; little
effect on triglycerides; and lowering of the total cholesterol/ HDL-cholesterol.
Effects of SFA consumption on serum lipids and lipoproteins further vary according to
which specific SFA is consumed [6,58,59]. Palmitic acid intake raised serum/plasma total
cholesterol and LDL-cholesterol. However, all SFA increased HDL-cholesterol as well, but
HDL-raising effects are greater as SFA chain-length decreases. This has been shown by metaanalysis of RCT studies, when different chain-length SFA were used as an isocaloric
replacement for carbohydrates [60]. The total cholesterol/HDL-cholesterol ratio, the best
overall indication of potential effects on coronary heart disease (CHD) risk is nonsignificantly
affected by consumption of palmitic acid.
Epidemiological studies have shown that the population who consume large amounts of
SFA (especially those of 12-16 carbon: lauric, myristic, and palmitic) have elevated levels of
LDL-cholesterol [61,62]. Saturated FA exert their atherogenic and thrombogenic effect
through increased production of very low-density lipoproteins (VLDL)-particles and Apo A1,
with a decrease of LDL-receptors specific activity, and an increase in platelet aggregation
[63]. Collective evidence suggests that SFA of 12-16 carbon, rather than stearic acid (18:0),
are the major activators of clotting factor VII [64]. Stearic acid has also been considered
highly atherogenic for years, but recent studies testify in favor of antiaterogenic and even
anti- carcinogenic effect of stearic acid [59,65,66]. For these reasons, recommendation by the
WHO / FAO experts for SFA (butter, cream, full-fat dairy products) intake is limited to less
than 10% of daily energy needs [1,4].
The most abundant SFA in milk fat is palmitic acid, which make up about 36% of total
fats and 44-51% of the total SFA in milk fat [67,68]. For this reason, milk and dairy products
are usually considered unhealthy, especially for people with dyslipidemia. In spite of this fact,
it has been well established that milk fat raises serum HDL-C, helping to maintain a good
HDL-cholesterol/total cholesterol ratio that is inversely related to CVD [60]. Furthermore,
palmitic and stearic acids in milk fat occupy the sn-2 position of triglycerides, which is
typically the position of unsaturated fatty acids in plant oils [69,70]. The selectivity of

110

Danijela Risti-Medi and Vesna Vui

pancreatic lipase to hydrolyze triglycerides at the sn-1 and sn-3positions leads to the
production of free fatty acids and 2-monoglyceride [69]. In this way, the unique position of
SFA in milk fat may affect postprandial metabolism, leading to prevention of
hypercholesterolemia and hypertriglyceridemia that would otherwise be associated with
consumption of saturated fat [69,68,70]. The beneficial effect of milk fat on serum lipids may
partially explain why milk fat, despite its contribution of SFA to the diet, has not been
consistently associated with higher incidence of CVD [69,72] or risk factors for cardiometabolic syndrome [72,73]. In a recent cohort study, butter and dairy intake did not predict
all-cause and ischemic heart disease mortality in men, and slightly increased risk in women,
whereas fermented full-fat milk was inversely associated with mortality in both men and
women [74] Moreover, similar findings are reported for palm oil, which also does not raise
blood cholesterol as expected based on the content of PA. It may be explained by the position
of palmitic acid in palm oil triacilglycol (10% of total PA is in the middle position). Because
triacilglycols with SFA in the sn-2 position may be absorbed more efficiently and cleared
from circulation more slowly than triacilglycols with SFA in the sn-1 and sn-3 positions,
intake of these dietary triacilglycols often leads to a more pronounced postprandial lipemia,
which is an independent risk factor for CHD [75]. Therefore, it can be assumed that PA
esterified to the sn-2 position is more atherogenic than when esterified to the sn-1 and sn-3
positions. Studies in animals and in human infants have supported this assumption since they
reported higher plasma triglycerides levels after diets with PA in the sn-2 position than after
diets containing PA in the sn-1/3 positions [76]. However, no significant differences were
found in one adult trial [77], whereas another study [78] reported larger LDL-cholesterol
concentrations caused by diets including palmitic acid in the sn-2 position in men but not in
women. Thus further research is needed to elucidate these relationships.

Insulin Resistance and Diabetes


Quality and amount of dietary fats significantly affects insulin resistance (IR), which is a
key player in development of metabolic syndrome and diabetes [79]. According to the Vessby
et al [15,49], amount can be considered more important, since excessive intake of total fat
(> 37% of daily energy intake) independently on the FA composition, may worse IR.
Furthermore, they reported that an exchange of dietary saturated for monounsaturated fats
improved insulin sensitivity. The underlying mechanism is not clarified yet, but it likely
includes interference with binding of insulin to its receptors and accumulation of triglycerides
in skeletal muscle [80]. On the contrary, total fat intake up to 30% of daily energy intake
differently influences IR, in term of the type of fatty acid consumed [15,49]. Observational
studies assessing fatty acid composition in serum or tissues (which correlates with dietary
intake) suggest that IR is associated with relatively high intakes of saturated fat (e.g. palmitic
acid) and low intakes of polyunsaturated fat (e.g. linoleic acid), findings that are supported by
recent clinical data [81,82]
The effects of SFA and MUFA on IR are different. Saturated fats increase IR when
compared to monounsaturated fats [83]. Hotamisligil et al [84] reported that reason for this
increase is that SFA activates serine kinases, thus inhibiting the insulin phosphorylation
cascade, decreasing glucose uptake and increasing glycemia. Gaster et al [85] have recently
found that PA and monounsaturated oleic acid are identically utilized in diabetic and control

Palmitic Acid As a Cardiometabolic Risk Factor

111

myotubes, although oxidation of PA is reduced in diabetic myotubes. Furthermore, there is a


difference in handling of PA and oleic acid in myotubes: PA accumulates in form of di- and
triacylglycerols, whereas oleic acid accumulates as intracellular free FA. Diacylglycerols
from palmitic acid activate protein kinase C, which has been shown to increase IR and thus
decrease glucose uptake in human skeletal muscle cells [86].
There is also a link between PA and myristic acid (14:0) and insulin synthesis. These
SFA (in particular PA) have been demonstrated to stimulate -cells in pancreas. When SFA
content in pancreatic islets increases, nitric oxide and ceramide are synthesized. A link
between serum ceramides, IR and inflammation is related to the inflammatory marker IL-6
[87]. As a consequence increased oxidative stress, inflammation and endoplasmic reticulum
stress are occurred, that lead to -cell apoptosis [88]. At the same time, insulin gene
expression is inhibited [89]
Taking into account the positive association between PA intake and IR, effect of PA on
diabetes is expected. In a recent report by FAO, SFA was considered to have a possible
positive relationship with increased risk of diabetes mellitus type 2 [1,61]. Nevertheless, in a
systematic review by van Dam et al [90] no clear associations were found. This systematic
review has shown that SFA intake was associated with a higher risk of diabetes mellitus type
2, but this association was not independent of body mass index. Few research studies have
explored the relationship between the amount of SFA in the diet and glycemic control and
CVD risk in people with diabetes. A systematic review by Wheeler et al [91] found just one
small 3-week study that compared a low-SFA diet (8% of total kcal) versus a high-SFA diet
(17% of total kcal) and found no significant difference in glycemic control and most CVD
risk measures [59,91].

DIETARY PA IN RELATION WITH CARDIOVASCULAR DISEASE RISK


Expert panel reached the conclusion that the evidence from epidemiologic, clinic and
mechanistic studies is consistent in the finding that CVD is reduced when SFAs are replaced
with PUFA in diet [92]. It is considered that replacement of only 1% of energy from SFA
with PUFA lowers LDL- cholesterol and is likely to produce a reduction in CHD incidence of
23% [93,94]. Individual SFA may have different cardiovascular effects and the effect of
particular foods on CHD cannot be predicted solely by their content of total SFA because
major SFA food sources contain other constituents that could influence CHD risk [92]. As
already mentioned, SFA with different chain length have different atherosclerotic potential.
Meta-analyses of cohort studies with self-reported SFA intakes are not associated with
CHD, stroke, or CVD [95, 96]. Two systematic reviews of prospective cohort studies and
meta-analyses, the first including 9 cohorts [97] and the second including 16 cohorts [98],
with self-reported SFA intakes found no significant association between SFA intake and
CHD risk. However, multivariable adjusted pooled analysis of individual-level data from 11
prospective cohorts over 410 years of follow-up [97] indicated that SFA consumption was
associated with higher CHD risk only in comparison to PUFA intake. In this direction, metaanalysis of eight RCTs, including participants with CHD events, found that CHD risk was
lowered by 10% when 5% energy intake of SFA substituted with PUFA [93]. That, SFA
consumption was associated with trend to lowered risk of stroke compared the highest to the

Danijela Risti-Medi and Vesna Vui

112

lowest category of SFA intake has been shown in meta-analyses of eight prospective cohort
studies [98]. Results from Women Health initiative trial [99], indicated that reduction in SFA
consumption does not appear to increase risk of stroke over 8 years. Some links between
dietary PA and cardiometabolic risk factors are presented in Figure 1.

Palmitic acid
Dietary sources:
Palm oil, dairy,
meat

Elevated LDL
Elevated HDLcholesterol

Tissue inflammation
Weight gain

Increased risk for


diabetes

Increased risk for CHD


Endotel dysfunction

Beta cell disfunction

Figure 1. Relationship between dietary intake of palmitic acid and cardiometabolic risk.

PALMITIC ACID PROPORTION IN SERUM OR TISSUES IN RELATION TO


CARDIOMETABOLIC RISK
Fatty acid composition in serum lipids reflects dietary intake and metabolic processes.
High proportions of palmitic, palmitoleic (16:1), and dihomo--linoleic (DHLA, 20:3n6)
acids and a low proportion of linoleic acid (LA, 18:2n6) in serum/plasma lipids predicts type
2 diabetes [100,101,102], myocardial infarction [103,104], stroke [105], left ventricular
hypertrophy [106], and the metabolic syndrome [107,108.109]. In a Swedish cohort, 14:0 and
16:0 in serum cholesterol esters independently predicted cardiovascular and all-cause
mortality over 33 years [110]. Patients with incident hypertensives had higher levels of
palmitic acid compared to normotensives person [111]. In overweight adolescents plasma
fatty acid composition is associated with the metabolic syndrome and low-grade
inflammation [112]. PA status in blood of patients with cardiometabolic risk from available
literature data are shown in Table 1 [15, 111-121]. As comparison, Table 2 [117,122,123]
presents PA status in plasma and erythrocytes phospholipids in healthy subjects depending on
the age and gender.
For total SFA, the associations between tissue or circulating fatty acids and dietary intake
are diverse and often weak [124,125] possibly reflecting endogenous metabolism. Plasma
16:0 levels increase with dietary intake of SFA, although not as much as expected and not in a
doseresponse manner, probably partly due to conversion of 16:0 to 16:1 by steaoryl-CoAdesaturase (SCD) [126]. Thus, the ratio of 16:1 to 16:0, which represents the estimated SCD
activity, may also be used as a marker of dietary 16:0 intake [83]. Additionally, circulating
and tissue levels of 14:0, 16:0 and 18:0 can be affected by high intakes of carbohydrate or
alcohol [124].
Warensj et al [110] showed that PA, but not stearic acid, was significantly associated
with increased mortality, especially cardiovascular disease mortality. This is in line with

Palmitic Acid As a Cardiometabolic Risk Factor

113

experimental data suggesting that palmitic acid has unique effects on several cellular
functions, such as apoptosis [127], endoplasmic reticulum stress [128], and up-regulation of
SCD-1 [129]. The lipogenic enzyme SCD catalyzes the synthesis of MUFAs, eg, oleic and
palmitoleic acids. Estimated SCD activity (16:1/16:0 ratio), together with palmitoleic acid,
has been considered as a strong predictor of mortality [126]. It may be associated with
increased lipogenesis [129], ectopic fat deposition and thereby insulin resistance
[130,131,132]. Accordingly, the estimated SCD ratio was established as an independent
predictor of directly measured insulin sensitivity over 20 years [81]. In contrast, our data on
patients with non-Hodgkin lymphoma showed very low proportion of 16:1 and activity of
SCD, especially in patients with progression of disease, but the role of SDC in cancer should
be further investigated [133].
Table 1. Palmitic acid status in patients with cardiometabolic risk
Cardiometabolic risk patients
1.Hyperlipidemic patients (n=29) [113]
2. Hyperlipidemic patients (n=39) [114]
Obesity women (n=30) [15]
I-NGT group (n=12)
II-IR group (n=18)
DM type 2 with hyperlipidemia (n=28) [115]
I-IHTG (n=14)
II-CHL (n=14)
1. Hemodialysis patients (n=35) [116]
2. Hemodialysis patients (n=37) [117]

Alcoholic cirrhosis (n=20) [118]


I. Patient with incident hypertension (n=413) [119]
II. Patient with no incident hypertension (n=1965)
III. Patient with prevalent hypertension (n=698)
IV. Patient with no prevalent hypertension (n=2383)

PA status
(source of FA, mol%)
1.serum PL 30.35 5.94
Er 25.58 4.15
2.serum PL 30.30 1.39
Er 23.64 0.90
Er 22.631.39
I-Er 22.491.67
II-Er 22.731.21
serum PL 30.01 2.70
I-serum PL 29.05 1.43
II-serum PL 30.71 3.67
1.serum PL 29.93 3.52
2. serum PL 28.09 3.34
controls 26.46 2.44
Er 21.63 1.85
control 22.42 2.59
serum PL 30.76 4.75
control 26.53 2.44
I. serum CE 10.07 0.77
II. serum CE 9.91 0.77
p< 0.001
III. serum CE 10.21 0.83
IV. serum CE 9.94 0.77
p< 0.001

Stroke (n=20) [120]

serum PL 29.2 2.57


control 37.2 2.13 p< 0.001

Danijela Risti-Medi and Vesna Vui

114

Table 1. (Continued)
Cardiometabolic risk patients

PA status
(source of FA, mol%)
I. Patients with incident CHD (n=282) [101]
I- serum PL 25.5 1.5
II-Patients with no incident CHD (n=3309)
II-serumCE 10.02 0.8
I- serum PL 25.4 1.7
II-Serum CE 10.0 0.8 p < 0.01
Patients with no MS (n=640) [121]
Er 22.7 1.2
Patients with MS (n=396)
Er 23.1 1.22 p<0.001
I- Normal weight patient (n=60) [112]
I-Serum PL 30.18 0.29
I-Serum CE 12.67 0.18
II-Overweight with no MS (n=45)
II-Serum PL 30.80 0.32
II-Serum CE 13.35 0.20
III-Overweight with MS (n=15)
III-Serum PL 31.52 0.55
III-Serum CE 13.97 0.32 p<0.001
The values are means SD; CHD, coronary heart disease; DM, diabetes mellitus; MS, metabolic
syndrome; IHTG, isolated hypertriglyceridemia; CHL, combined hyperlipidemia; CE, cholesterol
esters; PL, phospholipids; FA, fatty acid; Er, erythrocytes; IR,insulin resistence; NGT, normal
glucoso tolerance.

Table 2. Status of palmitic acid in plasma phospholipids in healthy subjects


Gender/age (y)
plasma PL
Er

MF/24 3
(n=16) [122]
26.21 2.16
21.56 0.96

MF/57 (19-74)
(n=29) [65]
26.4 2.5
n.a.

F/ 23.671.56
(n=14) [123]
27.72 1.60
23.39 0.40

MF/ 54.412
(n=37) [117] )
26.46 2.44
22.42 2.59

The values are means SD; PL. phospholipids; Er, erythrocytes; M, male; F, female;

As shown in a Swedish cohort of healthy 50-year-old men with 20 years follow-up,


proportion of 14:0 and 16:0 in serum cholesterol esters predicted the development of
metabolic syndrome, independently of other metabolic and lifestyle factors [105]. Besides
serum lipids, proportion of PA in adipose tissue is also related to insulin sensitivity. Unlikely
14:0 and 18:0, which were positively associated with insulin sensitivity, PA inversely
correlated with insulin sensitivity in 59 healthy British men and women [134]. Since the
authors excluded the effect of dietary intake for any of these SFA, they concluded that the
reason is de novo lipogenesis in adipose tissue. In human skeletal muscle phospholipids, SFA
[135,135] and especially PA [137] have been negatively associated with insulin sensitivity
and Type 2 diabetes, [138] which could partly reflect dietary intake [139]. In a Finnish cohort
study of 4 years follow-up, impaired fasting glucose and Type 2 diabetes incidence were
associated with serum nonesterified 16:0 levels, but were not associated with baseline dietary
16:0 intakes assessed from dietary records [140]. Recently published prospective follow-up
study showed that erythrocyte membrane fatty acids nominaly predict incident type 2 diabetes
[141]. In the American Atherosclerosis Risk in Communities (ARIC) study, 2909 middleaged men and women were followed for 9 years. The incidence of Type 2 diabetes was
associated with total SFA levels of plasma cholesterol esters (also observed for 16:0
independently) and phospolipids (also for 16:0 and 18:0) [101]. In a more recent 4-year case

Palmitic Acid As a Cardiometabolic Risk Factor

115

cohort study from Australia, dietary intake of 16:0 and 18:0 assessed by food frequency
questioners (FFQ) at baseline nonsignificantly predicted diabetes incidence, whereas dietary
15:0 was inversely associated with diabetes [100]. In baseline plasma phospholipids, total
SFA and 18:0 were positively associated with diabetes risk, 16:0 was nonsignificantly
associated, whereas 15:0 was negatively associated [102]. Since dietary intake correlates with
serum lipids fatty acid composition, changes in dietary habits and intake of proper fats can
prevent development of metabolic and cardiovascular diseases.

CONCLUSION
Patients with cardiometabolic risk factors represent a group at high lifetime risk for CVD.
Among other factors, nutrition markedly contributes to the development of metabolic
diseases. Although cardiometabolic risk is associated with SFA-rich foods, systematic
reviews on prospective cohort studies indicated that CHD risk has not been directly
associated with SFA intake. Nevertheless, there is convincing evidence for decreased CHD
risk when replacing SFA with polyunsaturated fats. Differences in cardiometabolic risk
appear greater between food groups and overall dietary patterns rather than between separate
SFA, even though it has been documented that palmitic acid increased the risk. Based on the
current data, it is not possible to give dietary recommendations solely based on the content of
individual SFA. Strictly controlled short-term and longer-term intervention studies are needed
to establish convincing link between palmitic acid and various alterations reported in
observational studies.

ACKNOWLEDGMENT
This work was supported by the Project III41030 financed by the Ministry of Education,
Science and Technological Development of the Republic of Serbia.

REFERENCES
[1]

[2]
[3]

[4]

Fats and fatty acids in human nutrition.: Proceedings of the Joint FAO/WHO Expert
Consultation. November 1014, 2008. Geneva, Switzerland. Ann Nutr Metab, 2009,
55: 5300.
U.S. Department of Agriculture and U.S. Department of Health and Human Services.
Dietary Guidelines for Americans, 2010. 7th ed. Washington, DC: U.S. Government
Printing Office, 2010. EFSA. Scientific opinion on Dietary Reference Values for fats,
including saturated fatty acids, polyunsaturated fatty acids, monounsaturated fatty
acids, trans fatty acids, and cholesterol. EFSA Journal. 2010, 8(3):1461.
Elmadfa I, Kornsteiner M.: Fatty acids and fatty acid requirements for adults. Ann Nutr
Metab. 2009, 55: 56-75.

116
[5]

[6]
[7]
[8]
[9]
[10]

[11]

[12]

[13]

[14]
[15]
[16]

[17]
[18]
[19]
[20]

[21]

Danijela Risti-Medi and Vesna Vui


Hu FB, Stampfer MJ, Manson JE, et al.: Dietary saturated fats and their food sources in
relation to the risk of coronary heart disease in women. Am J Clin Nutr. 1999, 70:
10011008.
Micha R, Mozaffarian D.: Saturated fat and cardiometabolic risk factors, coronary heart
disease, stroke, and diabetes: a fresh look at the evidence. Lipids. 2010, 45: 893905.
Vucic V, Tepsic J, Arsic A, et al: Fatty acid content of vegetable oils and assessment of
their consumption in Serbia. Acta Alimentaria. 2012, 41(3): 343-350.
Aro A, Becker W.: Nutrition and health in Finland. Pub Health Nutr. 2010, 13: 899999.
Freire RD, Cardoso MA, Gimeno SG, Ferreira SR.: Dietary fat is associated with
metabolic syndrome in Japanese Brazilians. Diabetes Care. 2005, 28: 1779-1785.
Chen X, Pang Z, Li K.: Dietary fat, sedentary behaviors and the prevalence of the
metabolic syndrome among Qingdao adults. Nutr Metab Cardiovasc Dis. 2009, 19: 2734.
de Oliveira Otto MC, Jacobs DR, Mozaffarian, D, et al.: Dietary intakes of saturated
fat, CVD, T2D and Metabolic Syndrome: results from the Multi-Ethnic Study of
Atherosclerosis. Am J Clin Nutr. 2012, 96:397-404.
Kennedy A, Martinez K, Chuang CC, LaPoint K, McIntosh M.: Saturated fatty acidmediated inflammation and insulin resistance in adipose tissue: mechanisms of action
and implications. J Nutr. 2009, 139:1-4.
National Cholesterol Education Program (NCEP) Expert Panel on Detection E, and
Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). Third
report of the National Cholesterol Education Program (NCEP) expert panel on
detection, evaluation and treatment of high cholesterol in adults (Adult Treatment Panel
III) final report. Circulation. 2002, 106: 3143421.
Ristic-Medic D, Vucic V. Dietary Fats And Metabolic Syndrome. J Nutrition Health
Food Sci. 2013,1 (1).
Vessby B.: Dietary fat, fatty acid composition in plasma and the metabolic syndrome.
Curr Opin Lipidol 2003, 14:159.
Takic M, Ristic-Medic D, Mandic Lj, et al.: N-3 polyunsaturated acids in erythrocyte
phospholipids are associated with insulin sensitivity in obese patients on a typical
Serbian diet. Arch Biol Sci Belgrade. 2009, 61(1): 37-43.
Vucic V.: The role of dietary polyunsaturated fatty acids in inflammation. Serbian J
Experimenl and Clinic Research, 2013 14(3), 93-99.
Ristic-Medic D, Vucic V, Takic M et al.: Polyunsaturated fatty acid in health and
disease. J Serb Chem Soc. 2013, 78: 1269-1289.
Odegaard J and Chawla A.: Pleiotropic actions of insulin resistance and inflammation
in metabolic homeostasis. Science. 2013, 339(6116): 172177.
Yuzefovych L, Wilson G, Rachek L.: Different effects of oleate vs. palmitate on
mitochondrial function, apoptosis, and insulin signaling in L6 skeletal muscle cells: role
of oxidative stress. Am J Physiol Endocrinol Metab. 2010, 299(6): E1096-1105.
Rachek LI, Musiyenko SI, LeDoux SP, Wilson GL.: Palmitate induced mitochondrial
deoxyribonucleic acid damage and apoptosis in l6 rat skeletal muscle cells.
Endocrinology. 2007, 148(1):293-299.

Palmitic Acid As a Cardiometabolic Risk Factor

117

[22] Lundman P, Boquist S, Samnegard A, et al.: high-fat meal is accompanied by increased


plasma interleukin-6 concentrations. Nutr Metab Cardiovasc Dis. 2007, 17:195202.
[23] Kien CL, Bunn JY, Ugrasbul F.: Increasing dietary palmitic acid decreases fat oxidation
and daily energy expenditure. Am J Clin Nutr. 2005, 82:3206.
[24] Riserus U.: Fatty acids and insulin sensitivity. Curr Opin Clin Nutr Metab Care. 2008,
11: 1005.
[25] Ajuwon KM, Spurlock ME.: Palmitate activates the NF-kappaB transcription factor and
induces IL-6 and TNFalpha expression in 3T3L1 adipocytes. J Nutr. 2005, 135:1841
6.
[26] Suganami T, Mieda T, Itoh M, Shimoda Y, Kamei Y, Ogawa Y.: Attenuation of
obesity-induced inflammation in C3H/HeJ mice carrying a Toll-like receptor4
mutation. Biochem Biophys Res Commun. 2007, 354: 459.
[27] Song MJ, Kim KH, Yoon JM, Kim JB.: Activation of Toll-like receptor 4 is associated
with insulin resistance in adipocytes. Biochem Biophys Res Commun. 2006, 346: 739
45.
[28] Lee JY, Zhao L, Youn HS, Weatherill AR, et al.: Saturated fatty acid activates but
polyunsaturated fatty acid inhibits Toll-like receptor 2 dimerized with Toll-like receptor
6 or 1. J Biol Chem. 2004, 279:169719.
[29] Shi H, Kokoeva M, Inouye K, Tzanneli I, Yin H, Flier J.: TLR4 links innate immunity
and fatty acid-induced insulin resistance. J Clin Invest. 2006, 116: 301525.
[30] Davis J, Gabler N, Walker-Daniels J, Spurlock M.: TLR-4 deficiency selectively
protects against obesity-induced by diets high in saturated fat. Obesity (Silver Spring)
2008, 16: 124855.
[31] Bradley RL, Fisher FF, Maratos-Flier E.: Dietary fatty acids differentially regulate
production of TNF-alpha and IL-10 by murine 3T3L1 adipocytes. Obesity (Silver
Spring). 2008, 16: 93844
[32] Guo W, Wong S, Xie W, Lei T, Luo Z.: Palmitate modulates intracellular signaling,
induces endoplasmic reticulum stress, and causes apoptosis in mouse 3T3L1 and rat
primary preadipocytes. Am J Physiol Endocrinol Metab. 2007, 293: E57686.
[33] Reynoso R, Salgado LM, Calderon V.: High levels of palmitic acid lead to insulin
resistance due to changes in the level of phosphorylation of the insulin receptor and
insulin receptor substrate. Mol Cell Biochem. 2003, 246:15562.
[34] Chavez JA, Summers SA.: Characterizing the effects of saturated fatty acids on insulin
signaling and ceramide and diacylglycerol accumulation in 3T3L1 adipocytes and
C2C12 myotubes. Arch Biochem Biophys. 2003, 419: 1019.
[35] Xi L, Qian Z,Zhou C, Sun S.: Corcetin attenuates palmitate-induced insulin
insensitivity and disordered tumor necrosis factor-alpha and adiponectin expression in
rat adipocytes. Br J Pharmacol. 2007, 151: 6107.
[36] Kim JY, Van de Wall E, Laplant M, Azzara A, Trujillo M.: Obesity associated
improvements in metabolic profile through expansion of adipose tissue. J Clin Invest.
2007, 117: 262137.
[37] Laine PS, Schwartz EA, Wang Y, et al.: Palmitic acid induces IP-10 expression in
human macrophages via NF-kappaB activation. Biochem Biophys Res Commun. 2007,
358: 1505.

118

Danijela Risti-Medi and Vesna Vui

[38] Permana PA, Menge C, Reaven PD.: Macrophage-secreted factors induce adipocyte
inflammation and insulin resistance. Biochem Biophys Res Commun. 2006, 341: 507
14.
[39] Lumeng CN, Deyoung M, Saltiel AR.: Macrophages block insulin action in adipocytes
by altering expression of signaling and glucose transport proteins. Am J Physiol
Endocrinol Metab. 2007, 292: E16674.
[40] Jove M, Planavila A, Laguna JC, Vazquez-Carrera M.: Palmitate induced interleukin 6
production is mediated by protein kinase C and nuclear-factor kB activation and leads
to glucose transporter 4 down-regulation in skeletal muscle cells. Endocrinology. 2005,
146: 308795.
[41] Jove M, Planavila A, Sanchez RM, et al.: Palmitate induce tumor necrosis factor-a
expression in C2C12 skeletal muscle cells by a mechanism involving protein kinase C
and nuclear factor-kappaB activation. Endocrinology. 2006,147(1): 552-61.
[42] Coll T, Jove M, Rodriguez-Calvo R, et al.: Palmitate-mediated downregulation of
peroxisome proliferator-activated receptor-g coactivator 1a in skeletal muscle cells
involves MEK1/2 and nuclear factor-kB activation. Diabetes. 2006, 55: 277987.
[43] Handschin C and Spiegelman B.: Peroxisome proliferator-activated receptor gamma
coactivator 1 coactivators, energy homeostasis, and metabolism. Endocr Rev. 2006, 27:
72835.
[44] Crunkhorn S, Dearie F, Mantzoros C, et al.: Peroxisome proliferator activator receptor
gamma coactivator-1 expression is reduced in obesity: potential pathogenic role of
saturated fatty acids and p38 mitogen-activated protein kinase activation. J Biol Chem.
2007, 282: 1543950.
[45] Lee JS, Pinnamaneni SK, Eo SJ, et al.: Saturated, but not n-6 polyunsaturated, fatty
acids induce insulin resistance: role of intramuscular accumulation of lipid metabolites.
J Appl Physiol. 2006, 100: 146774.
[46] Katan MB, Zock PL, Mensink RP.: Effects of fats and fatty acids on blood lipids in
humans: an overview. Am J Clin Nutr. 1994, 60: 1017S22S.
[47] Sacks FM, Katan M.: Randomized clinical trials on the effects of dietary fat and
carbohydrate on plasma lipoproteins and cardiovascular disease. Am J Med. 2002, 113
9B:13S24S.
[48] De Caterina R, Zampolli A, Del Turco S, et al.: Nutritional mechanisms that influence
cardiovascular disease. Am J Clin Nutr 2006, 83 (suppl):421S 6S.
[49] Vessby B, Uusitupa M, Hermansen K, et al.: Substituting dietary saturated for
monounsaturated fat impairs insulin sensitivity in healthy men and women: the
KANWU Study. Diabetologia. 2001, 44: 3129.
[50] Bos MB, de Vries JHM, Feskens EJM, et al.: Effect of a high monounsaturated fatty
acids diet and a Mediterranean diet on serum lipids and insulin sensitivity in adults with
mild abdominal obesity. Nutr Metab Cardiovasc Dis. 2010, 8: 591-598.
[51] Denke MA and Grundy SM.: Effects of fats high in stearic acid on lipid and lipoprotein
concentrations in men. Am J Clin Nutr. 1991, 54: 10361040.
[52] Gill JM, Brown JC, Caslake MJ, et al.: Effects of dietary monounsaturated fatty acids
on lipoprotein concentrations, compositions, and subfraction distributions and on
VLDL apolipoprotein B kinetics: dose-dependent effects on LDL and on VLDL
apolipoprotein B kinetics: dose-dependent effects on LDL. Am J Clin Nutr. 2002,
78(1): 47-56.

Palmitic Acid As a Cardiometabolic Risk Factor

119

[53] Lefevre MC, Champagne CM, Tulley RT, et al.: Individual variability in cardiovascular
disease risk factor responses to low-fat and low-saturated-fat diets in men: body mass
index, adiposity, and insulin resistance predict changes in LDL cholesterol. Am J Clin
Nutr. 2005, 82(5): 957-963.
[54] Lovejoy JC.: The influence of dietary fat on insulin resistance. Current Diabetes
Reports. 2002, 2(5): 435-440.
[55] Smith RD, Kelly CN, Fielding BA, et al.: Long-term monounsaturated fatty acid diets
reduce platelet aggregation in healthy young subjects. Br J Nutr. 2003 90(3): 597-606.
[56] Summers LK, Fielding BA, Bradshaw HA, et al.: Substituting dietary saturated fat with
polyunsaturated fat changes abdominal fat distribution and improves insulin sensitivity.
Diabetologia. 2002, 45(3): 369-377.
[57] van Dijk SJ, Feskens EJ, Bos MB, et al.: A saturated fatty acid-rich diet induces an
obesity-linked proinflammatory gene expression profile in adipose tissue of subjects at
risk of metabolic syndrome. Am J Clin Nutr. 2009, 90(6): 1656-1664.
[58] Rivellese AA, Maffettone A, Vessby B et al.: Effects of dietary saturated,
monounsaturated and n-3 fatty acids on fasting lipoproteins, LDL size and post-prandial
lipid metabolism in healthy subjects. Atherosclerosis. 2003, 167(1): 149-158.
[59] Hunter JE, Zhang J, Kris-Etherton PM.: Cardiovascular disease risk of dietary stearic
acid compared with trans, other saturated, and unsaturated fatty acids: a systematic
review. Am J Clin Nutr. 2010, 91: 4663.
[60] Mensink RP, Zock PL, Kester AD, Katan MB.: Effects of dietary fatty acids and
carbohydrates on the ratio of serum total to HDL cholesterol and on serum lipids and
apolipoproteins: a meta-analysis of 60 controlled trials. Am J Clin Nutr. 2003, 77:
114655.
[61] Smit LA, Mozaffarian D, Willett W.: Review of fat and fatty acid requirements and
criteria for developing dietary guidelines. Ann Nutr Metab. 2009; 55:4455.
[62] Skeaff CM, Miller J.: Dietary fat and coronary heart disease: summary of evidence
from prospective cohort and randomised controlled trials. Ann Nutr Metab. 2009;
55:173201.
[63] Iggman D and Risrus U.: Role of Different Dietary Saturated Fatty Acids for
Cardiometabolic Risk. Clin Lipidology. 2011, 6(2): 209-223.
[64] Miller GJ.: Dietary fatty acids and the haemostatic system. Atherosclerosis. 2005, 179:
213227.
[65] Thijessen MA, Hornstra G, Mensink RP.: Stearic, Oleic, and Linoleic Acids Have
Comparable Effects on Markers of Thrombotic Tendency in Healthy Human Subjects. J
Nutr. 2005, 135: 2805-2811.
[66] Cvetkovi Z, Vui V, Cvetkovi B, et al.: Abnormal fatty acid distribution of the
serum phospholipids of patients with non-Hodgkin lymphoma. Ann Hematol. 2010, 89
(8), 775-782.
[67] Arsic A, Prekajski N, Vucic V, et al.: Milk in human nutrition: Comparison of fatty acid
profiles. Acta Vet (Beograd), 2009, 59: 569578.
[68] USDA National Nutrient Database for Standard Reference, Release 23 (cited 2011 May
4). Available from: http://www.nal.usda.gov/fnic/ foodcomp/search/.
[69] German JB, Gibson RA, Krauss RM, et al.: A reappraisal of the impact of dairy foods
and milk fat on cardiovascular disease risk. Eur J Nutr. 2009, 48: 191203.

120

Danijela Risti-Medi and Vesna Vui

[70] Kris-Etherton PM, Griel AE, Psota TL, et al.: Dietary stearic acid and risk of
cardiovascular disease: intake, sources, digestion, and absorption. Lipids 2005; 40:
1193200.
[71] Forsythe CE, French MA, Goh YK, et al.: Cholesterolaemic influence of palmitic acid
in the sn-1, 3 v. the sn-2 position with high or low dietary linoleic acid in healthy young
men. Br J Nutr. 2007; 98: 33744.
[72] Pfeuffer M and Schrezenmeir J.: Milk and the metabolic syndrome. Obes Rev. 2007,
8:10918.
[73] Elwood PC, Pickering JE, Givens DI, Gallacher JE.: The consumption of milk and
dairy foods and the incidence of vascular disease and diabetes: an overview of the
evidence. Lipids 2010; 45: 92539.
[74] Goldbohm RA, Chorus AM, Galindo Garre F, et al.: Dairy consumption and 10-y total
and cardiovascular mortality: a prospective cohort study in The Netherlands.Am J Clin
Nutr. 2011, 93: 615627.
[75] Yli-Jokipii K, Kallio H, Schwab U, et al.: Effects of palm oil and transesterified palm
oil on chylomicron and VLDL triacylglycerol structures and postprandial lipid
response. J Lipid Res. 2001, 42: 16181625.
[76] Berry SE.: Triacylglycerol structure and interesterification of palmitic and stearic acidrich fats: an overview and implications for cardiovascular disease. Nutr Res Rev. 2009,
22(1): 3-17.
[77] Nestel P, Clifton P, Noakes M.: Effects of increasing dietary palmitoleic acid compared
with palmitic and oleic acids on plasma lipids of hypercholesterolemic men. J Lipid
Res. 1994, 35: 65662.
[78] Zock PL, de Vries JH, de Fouw NJ, Katan BM.: Positional distribution of fatty acids in
dietary triglycerides: effect on fasting blood lipoprotein concentrations in humans. Am J
Clin Nutr. 1995, 61: 4855.
[79] Weickert MO.: What dietary modification best improves insulin sensitivity and why?
Clin Endocrinol (Oxf). 2012, 77: 508-12.
[80] Thomas T and Pfeiffer AF.: Foods for the prevention of diabetes: how do they work?
Diabetes Metab Res Rev. 2011, 28(1): 25-49.
[81] Riserus U, Willett WC, Hu FB: Dietary fats and prevention of Type 2 diabetes. Prog.
Lipid Res. 2009, 48: 4451.
[82] Riserus U, Arnlv J, Berglund L.: Long-term predictors of insulin resistance: role of
lifestyle and metabolic factors in middle-aged men. Diabetes Care, 2007, 30: 2928 33.
[83] Lopez S, Bermudez B, Pacheco YM, et al.: Distinctive postprandial modulation of beta
cell function and insulin sensitivity by dietary fats: monounsaturated compared with
saturated fatty acids. Am J Clin Nutr. 2008, 88: 638-644.
[84] Hotamisligil GS.: Inflammation and metabolic disorders. Nature, 2006, 444: 860-867.
[85] Gaster AR and Henning BN. Differential Utilization of Saturated Palmitate and
Unsaturated Oleate Evidence From Cultured Myotubes. Diabetes. 2005, 54: 648656.
[86] Snook JT, Park S, Williams G, et al.: Effect of synthetic triglycerides of myristic,
palmitic, and stearic acid on serum lipoprotein metabolism. Eur J Clin Nut, 1999, 53:
597605.

Palmitic Acid As a Cardiometabolic Risk Factor

121

[87] de Mello VD, Lankinen M, Schwab U, et al. : Link between plasma ceramides,
inflammation and insulin resistance: association with serum IL-6 concentration in
patients with coronary heart disease. Diabetologia. 2009, 52 (12): 2612-5.
[88] Poitout V, Robertson RP.: Glucolipotoxicity: fuel excess and -cell dysfunction.
Endocr Rev. 2008, 29: 351366.
[89] Manco M, Calvani M, Mingrone G.: Effects of dietary fatty acids on insulin sensitivity
and secretion. Diabetes Obes Metab. 2004, 6: 402413.
[90] van Dam RM, Willett WC, Rimm EB, Stampfer MJ, Hu FB. Dietary fat and meat
intake in relation to risk of type 2 diabetes in men. Diabetes Care. 2002, 25(3): 417
424
[91] Wheeler ML, Dunbar SA, Jaacks LM, et al.: Macronutrients, food groups, and eating
patterns in the management of diabetes: a systematic review of the literature, 2010.
Diabetes Care. 2012, 35: 434445.
[92] Astrup A, Dyerberg J, Elwood P et al.: The role of reducing intakes of saturated fat in
the prevention of cardiovascular disease: where does the evidence stand in 2010? Am J
Clin Nutr. 2011, 93(4): 684688.
[93] Mozaffarian D, Micha R, Wallace SK.: Effects on coronary heart disease of increasing
polyunsaturated fat in place of saturated fat: a systematic review and meta-analysis of
randomized controlled trials. PLoS Med. 2010, 23;7(3): e1000252.
[94] Lloyd-Williams F, OFlaherty M, Mwatsama M, et al.: Estimating the cardiovascular
mortality burden attributable to the European Common Agricultural Policy on dietary
saturated fats. Bull World Health Organ. 2008, 86: 53541A.
[95] Hooper L, Summerbell CD, Higgins JP, et al.: Dietary fat intake and prevention of
cardiovascular disease: systematic review. BMJ 2001; 322: 75763.
[96] Siri-Tarino PW, Sun Q, Hu FB, Krauss RM.: Meta-analysis of prospective cohort
studies evaluating the association of saturated fat with cardiovascular disease. Am J
Clin Nutr. 2010, 91: 535546.
[97] Mente A, de Koning L, Shannon HS, Anand SS.: A systematic review of the evidence
supporting a causal link between dietary factors and coronary heart disease. Arch Intern
Med. 2009, 169(7): 659669
[98] Jakobsen MU, O'Reilly EJ, Heitmann BL, et al.: Major types of dietary fat and risk of
coronary heart disease: a pooled analysis of 11 cohort studies. Am J Clin Nutr. 2009,
89(5): 14251432
[99] Howard BV, Van Horn L, Hsia J,et al.: Low-fat dietary pattern and risk of
cardiovascular disease: the Womens Health Initiative Randomized Controlled Dietary
Modification Trial. JAMA, 2006, 295(6): 655666.
[100] Hodge AM, English DR, O'Dea K et al.: Plasma phospholipid and dietary fatty acids as
predictors of Type 2 diabetes: interpreting the role of linoleic acid. Am J Clin Nutr.
2007, 86: 189197.
[101] Wang L, Folsom AR, Zheng ZJ, Pankow JS, Eckfeldt JH.: Plasma fatty acid
composition and incidence of diabetes in middle-aged adults: the Atherosclerosis Risk
in Communities (ARIC) study. Am J Clin Nutr. 2003, 78: 9198.
[102] Vessby B, Aro A, Skarfors E, et al.: The risk to develop NIDDM is related to the fatty
acid composition of the serum cholesterol esters. Diabetes, 1994, 43: 13531357.
[103] hrvall M, Berglund L, Salminen I, et al.: The serum cholesterol ester fatty acid
composition but not the serum concentration of alpha tocopherol predicts the

122

Danijela Risti-Medi and Vesna Vui

development of myocardial infarction in 50-year-old men: 19 years follow-up.


Atherosclerosis, 1996, 127: 6571.
[104] Miettinen TA, Naukkarinen V, Huttunen JK, Mattila S, Kumlin T.: Fatty-acid
composition of serum lipids predicts myocardial infarction. Br Med J. (Clin Res Ed),
1982, 285: 9936.
[105] Wiberg, B, Sundstrom J, Arnlov J, et al.: "Metabolic risk factors for stroke and transient
ischemic attacks in middle-aged men: a community-based study with long-term followup. Stroke. 2006, 37 (12): 2898-2903.
[106] Sundstrm J, Lind L, Vessby B, et al.: Dyslipidemia and an unfavorable fatty acid
profile predict left ventricular hypertrophy 20 years later. Circulation, 2001, 103: 836
41.
[107] Vessby B, Gustafsson IB, Tengblad S, Boberg M, Andersson A.: Desaturation and
elongation of fatty acids and insulin action. Ann N Y Acad Sci. 2002; 967:18395.
[108] Vessby B, Tengblad S, Lithell H. Insulin sensitivity is related to the fatty acid
composition of serum lipids and skeletal muscle phospholipids in 70-year-old men.
Diabetologia, 1994, 37: 1044 50.
[109] Warensjo E, Riserus U, Vessby B.: Fatty acid composition of serum lipids predicts the
development of the metabolic syndrome in men. Diabetologia. 2005, 48: 19992005.
[110] Warensjo E, Sundstrom J, Vessby B, Cederholm T, Riserus U.: Markers of dietary fat
quality and fatty acid desaturation as predictors of total and cardiovascular mortality: a
population-based prospective study. Am J Clin Nutr. 2008, 88: 203209.
[111] Zheng ZJ, Folsom AR, Ma J, et al.: Plasma fatty acid composition and 6-year incidence
of hypertension in middle-aged adults: the Atherosclerosis Risk in Communities
(ARIC) Study. Am J Epidemiol. 1999, 150: 492-500.
[112] Klein-Platat C, Drai J, Oujaa M, Schlienger JS, Chantal S.: Plasma fatty acid
composition is associated with the metabolic syndrome and low-grade inflammation in
overweight adolescents. Am J Clin Nutr. 2005, 82: 1178-1184.
[113] Risti-Medi D, Risti V, Tepi V, et al.: Effect of soybean Leci-Vita product on
serum lipids and fatty acid composition in patients with elevated serum cholesterol and
triglyceride level. Nutr Res. 2003, 23(4): 465-477.
[114] Ristic-Medic D, Suzic S, Vucic V, et al.: Serum and erythrocyte membrane
phospholipid fatty acid composition in hiperlipidemia: effects of dietary intervention
and combined diet and fibrate therapy. Gen Physiol Biophys. 2009, 28: 190-199.
[115] Ristic Medic D, Ristic V, Arsic A, et al.: Effects of soybean D-LeciVita product on
serum lipids and fatty acid composition in type 2 diabetic patients with hyperlipidemia.
Nutr Metab Cardiovasc Dis. 16(6): 395-404.
[116] Rasic-Milutinovic Z, Perunicic G, Pljesa S, et al.: Effects of N-3 PUFAs
supplementation on insulin resistance and inflammatory biomarkers in hemodialysis
patients. Renal Failure, 2007, 29: 321329.
[117] Ristic, V. Tepsic J, Ristic-Medic D, et al.: Plasma and erythrocyte phospholipid fatty
acids composition in Serbian hemodialyzed patients. Renal Failure, 2006, 28: 211216.
[118] Ristic-Medic D, Takic M, Vucic V, Kostic N, Glibetic M.: Abnormalities in serum
phospholipids fatty acid profile in patients with alcoholic liver cirrhosis- a pilot study. J
Clin Biochem Nutr, 2013, 53(1): 49-54.
[119] Wang L, Manson JE, Forman JP, et al.: Dietary fatty acids and the risk of hypertension
in middle-aged and older women. Hypertension. 2010, 56(4): 598-604.

Palmitic Acid As a Cardiometabolic Risk Factor

123

[120] Glew RH, Okolie H, Huang YS, et al.: Abnormalities in the fatty-acid composition of
the serum phospholipids of stroke patients. J Natl Med Assoc. 2004, 96(6): 826-32.
[121] Kabagambe E, Tsai MY, Hopkins PN, et al.: Erythrocyte Fatty Acid Composition and
the Metabolic Syndrome. Clinical Chem. 2008, 54:154162.
[122] Tepsic J, Vucic V, Arsic A, et al.: Plasma and erythrocyte phospholipid fatty acid
profile in professional basketball and football players. Eur J Appl Physiol. 2009,
107(3): 359-365.
[123] Arsi A, Vui V, Tepi J, Mazi S, Djeli M, Glibeti M.: Altered plasma and
erythrocyte phospholipid fatty acid profile in elite female water polo and football
players. Appl Physiol Nutr Metab, 2012, 37(1): 40-47.
[124] Hodson L, Skeaff CM, Fielding BA.: Fatty acid composition of adipose tissue and
blood in humans and its use as a biomarker of dietary intake. Prog Lipid Res. 2008, 47,
348380.
[125] Baylin A, Kabagambe EK, Siles X, Campos H.: Adipose tissue biomarkers of fatty acid
intake. Am J Clin Nutr. 2002, 76: 750757.
[126] Warensj E, Ohrvall M, Vessby B.: Fatty acid composition and estimated desaturase
activities are associated with obesity and lifestyle variables in men and women. Nutr
Metab Cardiovasc Dis. 2006, 16: 128 36.
[127] Listenberger LL, Ory DS, Schaffer JE.: Palmitate-induced apoptosis can occur through
a ceramide-independent pathway. J Biol Chem. 2001; 276:148905.
[128] Wei Y, Wang D, Pagliassotti MJ.: Saturated fatty acid-mediated endoplasmic reticulum
stress and apoptosis are augmented by trans-10, cis- 12-conjugated linoleic acid in liver
cells. Mol Cell Biochem. 2007, 303: 10513.
[129] Sampath H, Miyazaki M, Dobrzyn A, Ntambi JM.: Stearoyl-CoA desaturase-1 mediates
the pro-lipogenic effects of dietary saturated fat. J Biol Chem. 2007, 282: 248393.
[130] Lelliott C, Vidal-Puig AJ.: Lipotoxicity, an imbalance between lipogenesis de novo and
fatty acid oxidation. Int J Obes Relat Metab Disord. 2004, 28(suppl 4):S22 8.
[131] Bakke SS, Moro C, Nikoli N, et al.: Palmitic acid follows a different metabolic
pathway than oleic acid in human skeletal muscle cells; lower lipolysis rate despite an
increased level of adipose triglyceride lipase. Biochim Biophys Acta. 2012,
1821(10):1323-33.
[132] Petersson H, Basu S, Cederholm T, Risrus U. et al.: Serum fatty acid composition and
indices of stearoyl-CoA desaturase activity are associated with systemic inflammation:
longitudinal analyses in middle-aged men. Br J Nutr 2008, 99(6): 1186-1189.
[133] Cvetkovi B, Vui V, Cvetkovi Z, Popovi T, Glibeti M.: Systemic alterations in
concentrations and distribution of plasma phospholipids in prostate cancer patients.
Med Oncol. 2012, 29: 809-814, 2012.
[134] Roberts R, Hodson L, Dennis AL et al.: Markers of de novo lipogenesis in adipose
tissue: associations with small adipocytes and insulin sensitivity in humans.
Diabetologia. 2009, 52: 882890.
[135] Borkman M, Storlien LH, Pan DA, et al.: The relation between insulin sensitivity and
the fatty-acid composition of skeletal-muscle phospholipids. N Engl J Med. 1993, 238
244.
[136] Manco M, Mingrone G, Greco AV, et al.: Insulin resistance directly correlates with
increased saturated fatty acids in skeletal muscle triglycerides. Metabolism, 2000, 49,
220224.

124

Danijela Risti-Medi and Vesna Vui

[137] Vessby B, Tengblad S, Lithell H.: Insulin sensitivity is related to the fatty acid
composition of serum lipids and skeletal muscle phospholipids in 70-year-old men.
Diabetologia. 1994, 37: 10441050.
[138] Salomaa V, Ahola I, Tuomilehto J et al.: Fatty acid composition of serum cholesterol
esters in different degrees of glucose intolerance: a population-based study. Metabolism
1990, 39:12851291.
[139] Corpeleijn E, Feskens EJ, Jansen EH, et al.: Improvements in glucose tolerance and
insulin sensitivity after lifestyle intervention are related to changes in serum fatty acid
profile and desaturase activities: the SLIM study. Diabetologia. 2006, 49: 2392 401.
[140] Mahendran Y, gren J, Uusitupa M, et al.: Association of erythrocyte membrane fatty
acids with changes in glycemia and risk of type 2 diabetes. Am J Clin Nutr. 2014,
99(1): 79-85.
[141] Laaksonen DE, Lakka TA, Lakka HM, et al.: Serum fatty acid composition predicts
development of impaired fasting glycaemia and diabetes in middle-aged men. Diabet
Med. 2002, 19: 456464.

In: Palmitic Acid: Occurrence, Biochemistry and Health Effects ISBN: 978-1-63321-519-1
Editor: Lucas F. Porto
2014 Nova Science Publishers, Inc.

Chapter 6

PALMITIC ACID IN HIGHER PLANT LIPIDS


R. A. Sidorov, A. V. Zhukov, V. P. Pchelkin
and V. D. Tsydendambaev*
Laboratory of Lipid Metabolism, K.A.Timiryazev Institute of Plant Physiology, Russian
Academy of Sciences, Botanicheskaya ul. 35, Moscow, Russia

ABSTRACT
Palmitic acid (C16:0) is one of the major fatty acids (FAs) forming virtually all natural
lipids. Both in eu-, and prokaryotes, C16:0 forms various lipid classes, which serve either
as the lipid background of storage fats and oils, or the hydrophobic matrix of cell
membranes, or the components of cuticle waxes and polymers. Non-esterified 16:0 does
not occur in living cells, and it is present there only as an acyl residue in various lipid
classes, such as mono-, di-, and triacylglycerols, glyco-, phospho-, and sphingolipids,
wax and steryl esters etc., where it esterifies the hydroxy groups of glycerol backbone or
other alcohols (sphingosine, higher and lower aliphatic alcohols etc.). Palmitic acid is
known to be a primary higher FA synthesized in the cell, while nearly all other FAs of
natural lipids are the products of its further modification caused by elongation,
desaturation, insertion of various functional groups, such as methyl, hydroxy, oxo, epoxy,
etc. As a saturated FA, C16:0 is used by the cell for regulating its functional state by
shifting the membrane fluidity under adverse environmental conditions and thus
providing a necessary molecular species composition of the membrane polar lipids.
Among the latter, such classes as phosphatidylinositols, phosphatidylserines, and other
highly polar lipids are particularly rich in palmitic acid. In accordance, its content in plant
lipids rises as they became less TLC-mobile, more difficultly extractable, or tightly
bound. It is evident that further screening of plant lipids as regards this index is of
considerable interest.

Corresponding author: Vladimir D. Tsydendambaev, e-mail: vdt1952@mail.ru.

126

R. A. Sidorov, A. V. Zhukov, V. P. Pchelkin et al.

INTRODUCTION
Lipids are an essential constituent of all plant cells. Fatty acids (FAs) are the mandatory
structural components of almost all lipids both storage triacylglycerols (TAGs), the seed oil
base, and membrane lipids forming every cell membrane bilayer. To date, more than 1000
different FAs are described. Such a FA diversity were found mainly in plant seed oils, and it
has been estimated that thousands more could be present throughout the plant kingdom. [1,
2]. All FA diversity is subdivided into major, minor and unusual ones [3]. Structural
membrane glycerolipids of all plant cells contain almost exclusively C16- and C18-FAs, with
none or up to three methylene-interrupted double bonds. Seven FAs with even number of
carbon atoms in the aliphatic chain lauric (C12:0), myristic (C14:0), palmitic (C16:0), stearic
(C18:0), oleic (C18:19), linoleic (C18:29,12) -linolenic (C18:39,12,15) acids, belongs to major
FAs. These FAs are widespread in nature and are the main lipid components, as a rule. The
acids homologous to major FAs with the greater or fewer number of carbon atoms or
differing in the position or configuration of double bonds and present in lower amounts in
lipids (no more than 5% of total FAs) of limited number of plant species are known as minor
FAs. Major and minor FAs are present in the lipids of most plant species; therefore, they are
called as common or usual FAs. The rest FAs deviate significantly from the common
ones in double bond configuration or position or in the presence of coupled double or triple
bonds or presence of different functional groups and also met in the plant lipids belong to
unusual FAs [3, 4]. Meanwhile, the attribution of a FA to the group of major, minor, or
unusual FAs is rather arbitrary and may be used only in a comparative study of the total lipids
in a large number of objects.
Palmitic acid occupies a distinct place among the all FA diversity. This FA in one or
another quantity present in every lipid class in all plant objects. Moreover virtually all FAs
are derived from palmitic acid by its modification, namely desaturation, elongation,
hydroxylation, oxidation, etc. Although fatty acids are major constituents of every membrane
in a cell and are also found outside cells in the cuticular lipids, their major site of synthesis is
within the plastid. In this regard, the process of lipid biosynthesis in plants is fundamentally
different from that in animals and fungi, which produce fatty acids primarily in the cytosol
[5].

Palmitic Acid Biosynthesis


The biosynthesis of FA occurs predominantly in the two subcellular compartments,
chloroplasts and endoplasmic reticulum (ER); plant mitochondria also contribute to FA
synthesis, but only in a very minor way. De novo synthesis of palmitic, stearic and FAs with
shorter chain length and also the first desaturation step of saturated FAs e.g. palmitic acid to
palmitoleic or stearic acid to oleic one occur in plastids, whereas the next desaturation steps
occurs in the ER. [4, 6].

Palmitic Acid in Higher Plant Lipids

127

FatA and FatB acyl-ACP thioesterases hydrolyzing predominantly oleoyl-ACP and diverse saturated
and unsaturated acyl ACP, respectively; ENR enoyl ACP reductase; KAR ketoacyl ACP
reductase; KAS ketoacyl synthase; MCMT malonyl-CoA : ACP malonyl transferase; HADH
hydroxyacyl ACP dehydratase.
Figure 1. FA biosynthesis in higher plants.

The synthesis of FA from acetyl-CoA starts in plastids (Figure 1) and occurs in three
stages. At the first stage the irreversible carboxylation of acetyl-CoA catalyzed by acetyl-CoA
carboxylase with the formation of malonyl-CoA occurs. Then the malonyl group is
transferred to the acyl-carrier protein (ACP) and, due to the operation of plastidial acetyl-CoA
carboxylase complex the primary substrate of FA synthetase, malonyl-ACP, is formed. [6, 7].
During the second stage, successive condensation of newly formed malonyl-CoA with
growing, bound to ACP acyl chain occurs. In such a way, by the successive addition of twocarbon fragments to the growing chain, palmitoyl-ACP is finally produced [6, 7]. Each cycle
of condensation includes four reactions. Firstly, due to the condensation of acetyl-CoA and
malonyl-CoA, 3-ketobutyl-ACP is formed catalyzed by ketoacyl synthase III (KAS III); then
3-ketobutyl-ACP is reduced to 3-oxyacyl-ACP, dehydrated to enoyl-ACP, and further
reduced to butyryl-ACP. Subsequent cycles of condensation of acyl-ACP with malonyl-CoA
are catalyzed by KAS I and proceed to the formation of myristoyl-ACP and palmitoyl-ACP
[4, 6-8]. The elongation of palmitoyl-ACP to stearoyl-ACP and its desaturation by 9desaturase to oleoyl-ACP are catalyzed by KAS II and occur also in plastids. Then, during the
third stage of FA biosynthesis, palmitoyl-ACP, stearoyl-ACP, and oleoyl-ACP are exported
into the cytosol and included into acyl-CoA and acyl-lipid pools [6, 9]. In most plants, the
process of FA elongation can continue, and due to functioning of the microsomal fatty acid
elongation (FAE) system so-called very long chain FAs (VLCFAs) including 20 and more
carbon atoms are formed [3, 6, 10].
The termination of elongation is catalyzed by ACP-thioesterases (enzymes belonging to
the class of acyl-ACP hydrolases). These enzymes hydrolyze acyl-ACP with the formation of
free FA, which can cross the plastid membrane to be reactivated outside the organelle [11].

128

R. A. Sidorov, A. V. Zhukov, V. P. Pchelkin et al.

Two main types of plant thioesterases are described: FatA preferably cleaving oleate from
ACP and FatB hydrolyzing diverse saturated and unsaturated acyl-ACP. In some plant
species, thioesterases, which are specific to acyl-ACP with short-chain acyls were found [12,
13]. The interaction between FA synthetase, 9-desaturase, and the two types of thioesterases
determines the ratio between FA produced in plastids, which will be further used by the cell
for the synthesis of glycerolipids [2].

Further Modifications of Palmitic Acid


It is well known that the major FAs of plants (and most other organisms) have a chain
length of 16 or 18 carbons and contain from one to three cis double bonds [3]. Five fatty acids
(C16:0, C18:19, C18:29,12, C18:39,12,15, and in some species, C16:39,12,15) make up over 90% of
the acyl chains of the structural glycerolipids of almost all plant membranes. The insertion of
one or several double bonds into aliphatic chain, the desaturation, is the most well studied
modification of palmitate and stearate, its elongation product (Figure 2). The first cis double
bond is inserted into aliphatic chain of C16:0 by 9-desaturase (FadB). The product aroused,
C16:19, further desaturates successively by FadC forming C16:29,12, and FAD7 and FAD8
introduce the third double bond, forming C16:39,12,15 [14]. This pathway was found only in
some plant species, however every higher plant possesses such a pathway with the another
substrate, C18:0, producing essential unsaturated C18 FAs both for membrane and storage lipid
synthesis. That is why regardless of their FA composition, angiosperms can be divided into
the so-called 16:3-plants (those containing C16:39,12,15) and the 18:3-plants which contain
little, if any, C16:39,12,15 [15, 16]. Differences between 16:3-plants and 18:3-plants are
reflected in variations in their plastidial lipid metabolism [16]. Briefly, galactolipids, the
major chloroplastic lipids, can be synthesized by two discrete pathways. The first (named the
prokaryotic pathway because it also is found in cyanobacteria) is entirely located in the
chloroplasts and leads to the synthesis of galactolipids containing tri-unsaturated C16-FAs.
The second pathway (called the eukaryotic pathway) results from the close cooperation
between endoplasmic reticulum and chloroplasts, and leads to the formation of plastid
glycerolipids containing tri-unsaturated C18-FAs. In 16:3-plants, both pathways contribute
to the synthesis of plastid lipids, so large amounts of C16:39,12,15 and C18:39,12,15 FAs are
detected in their photosynthetic tissues. In 18:3-plants, the eukaryotic pathway is the only
operative one, so little or no C16:39,12,15 is observed. Indeed, chloroplasts from 16:3-plants
possess the full equipment for the synthesis of plastid lipids [16], whereas plastids from
18:3-plants may be regarded as organelles which have lost some of their prokaryotic
features [17].
The another specific product of palmitic acid modification, which is an obligatory
component of phosphatidyl glycerol of the thylakoid membrane, is trans-3-hexadecenoic acid
(16:13tr). The synthesis of this FA from C16:0 is catalyzed by FadA (Fad4) which inserts a
trans-3 double bond into palmitate at the sn-2 position of phosphatidylglycerol [14]. One
more chloroplast desaturase, Fad5 inserts a double bond into palmitate residue on
monogalactosyldiacylglycerol and, possibly, digalactosyldiacylglycerol resulting the
formation of 7-hexadecenoate [18].

Palmitic Acid in Higher Plant Lipids

129

FAE fatty acids elongase; FAS fatty acid synthetase; KASII ketoacyl synthase II.
Figure 2. Fatty acid desaturation.

Thus, the synthesis of most monounsaturated FAs is catalyzed by soluble plastid


desaturase, which normally inserts a double bond between the 9th and 10th carbon atoms of
the acyl residue of palmitoyl- or stearoyl-ACP [1]. At the same time, some plants capable of
the synthesis of unusual monounsaturated FAs have additional desaturases, which structure is
close to that of 9-desaturase but despite this fact they introduce the double bond into another
position of the acyl chain and can manifest different substrate specificity [2]. These fatty
acids occur mainly in storage triacylglycerols (TAGs) of certain oilseed species but are
excluded from polar glycerolipids and consequently from the cell membranes. Thus, the seeds
of some plants from the family Umbelliferae, carrot and coriander in particular, contain oils
enriched in petroselinic acid (C18:16). This unusual FA is formed due to the operation of
plastid 4-desaturase introducing the double bond between the 4th and 5th carbon atoms of
the acyl in palmitoyl-ACP, conversing it into 4-hexadecenoyl-ACP, which then elongates to
petroselenic acid [19]. Petroselinic acid is the principal fatty acid of the seed oil also of most
Araliaceae and Garryaceae species [20]. Some plants (such as members of Boraginaceae,
Rununculaceae, Primulaceae, and another 7 families) contain 6-desaturase [21], and some
gymnospermous plants contain 5 desaturase [22]. The unusual 8-16:1 was found recently
in arils of fruits of some Euonymus species [23]. The seed oils of Androsace septentrionalis
and A. lactea (Primulaceae) were both found to contain cis-11-hexadecenoic and cis,cis-9,12hexadecadienoic acids [24]. These FAs are known to occur as a seed oil component in some
Proteaceae and Asclepiadaceae species.

130

R. A. Sidorov, A. V. Zhukov, V. P. Pchelkin et al.

AD I & AD III aldehyde decarbonilases, encoded by A. thaliana CER1 and CER3 genes resp.; FAE
fatty acids elongase; FAR fatty acids reductase; FAS fatty acid synthetase; S and SO
substrate and oxidized substrate respectively.
Figure 3. Some other modifications of palmitate.

In plant tissues C16:0 can undergo some other modifications (Figure 3), namely
elongation, hydroxylation, oxidation, epoxydation, reduction, oxidative decarboxylation, etc.
As a result of these modifications many different lipophilic substances are produced. Among
these substances very long-chain FAs (VLCFAs, C20), different unusual FAs (hydroxy-,
epoxy-, acetylenic, dicarboxylic), fatty aldehydes and alcohols, hydrocarbons, oxilipins, etc.
are formed. Some of them are present in plants in free form (are embedded in the complex
cuticular lipid matrix or as a components of epicuticular waxes), the others are used as a
substrates for more complex lipids and lipid polymers biosynthesis (see below).
Once synthesized, the palmitoyl moiety is transported out of the plastid to the
endoplasmic reticulum where it serves as an initial substrate for further modifications. For
instance, it can be elongated to very long chain fatty acids which contain up to 34 carbon
atoms. This process requires the sequential use of 4 reactions to add each C2 unit [25]. These
reactions are analogous to de novo synthesis (see above) but elongases, involved in synthesis
of very long chain fatty acids, are membrane-bound enzymes which use acyl-CoA substrates
and malonyl-CoA directly as the source of the C2 unit. Once elongation is complete, VLCFAs
may be reduced to fatty primary alcohols by an acyl-CoA reductase (FAR3/CER4) of the
acyl-reduction pathway, which appears to be associated with the ER. The primary fatty
alcohols generated and C16:0 acylCoA are condensed into wax esters by the bi-functional
wax synthase/acylCoA:diacylglycerol acyltransferase (WS/DGAT) enzyme, WSD1. Wax
synthase, catalyzing this reaction, is an integral membrane protein, but its site of action is not
known [26].
VLCFAs, besides, may be converted to the alkanes another component of epicuticular
waxes through fatty aldehydes by the action of membrane-associated enzymatic complex

Palmitic Acid in Higher Plant Lipids

131

comprised of two subunits of aldehyde decarbonylases [27, 28]. It was proposed that
VLCFAs activated by the long chain acyl CoA synthases in very long-chain acyl-CoAs would
be used as precursors of very long-chain alkane synthesis. A mandatory CER1/CER3
heterodimer would efficiently catalyze a two-step reaction starting with the reduction of acylCoA to a potential intermediate aldehyde subsequently decarbonylated to alkane with the loss
of one carbon potentially in carbon monoxide or formate as reported in cyanobacteria [29].
CYTB5s would interact with the di-iron catalytic core of CER1, providing electron(s)
required for the decarbonylation reaction. Additionally, palmitoyl-CoA and VLCFAs may be
converted into 2-hydroxy fatty acids, aldehydes and odd-numbered fatty acids through the
action of peroxigenases on the 2-hydroperoxy FAs which arise by the action of dehydrogenases [30].
VLCFAs form components of epidermal lipids, sphingolipids or storage lipids and are
synthesized by the acyl-CoA elongase complex [31]. The elongase is a multienzyme complex
which comprises four dissociable subunits, each possessing a unique enzymatic activity: 3ketoacyl-CoA synthase (KCS), 3-ketoacyl-CoA reductase (KCR), 3-hydroxyacyl-CoA
dehydratase and trans-2,3-enoyl-CoA-reductase (ECR); this elongase complex is membranebound which use acyl-CoA substrates and malonyl-CoA directly as the source of the C2-unit
[32]. VLCFAs are further modified to make different kinds of epicuticular wax precursors
(e.g. alkanes, fatty alcohols, aldehydes, etc., Figure 3). On the other hand, C16 and C18 FAs
are also modified to produce major monomers of lipid polymers, cutin and suberin (e.g. C16
and C18 hydroxy- and dicarboxylic FAs). The cuticle monomers are synthesized inside the
epidermal cells and deposited outside the epidermis [3]. This fact suggests the existence of
transporter(s) involved in the transportation of cuticle lipids across the plasma membrane.
Cutin is a polyester polymer composed of complex mixture of inter-esterified long chain
-hydroxy FAs and is considered as the major constituent of the plant cuticle. Cutin
monomers build up a complex, three-dimensional network by cross-links through the primary
and secondary hydroxyl and carboxyl groups [33]. C16- and C18- -hydroxy-FAs, with midchain functional groups such as epoxy- and hydroxy-, were predominant in all cutins; in some
plant cutins a small quantities of -hydroxy-C20 and C22 FAs may also occur. The other
monomers have been found in plant cutins are FAs, dicarboxylic FAs, alkanols, some
aromatic acids, and glycerol. The usual and in many cases the major constituents of most
plant cutins are different dihydroxy- derivatives of palmitic acid (C16:0di-OH). Several C16:0di-OH
isomers have been found in many cutin hydrolysates, the major ones being 10,16-C16:0di-OH
and 9,16-C16:0di-OH acids; smaller concentrations of 8,16-C16:0di-OH and 7,16-C16:0di-OH isomers
also often occur [34]. In some plant cutins a substantial quantities of 16-hydroxy-10oxohexadecanoic acid and related positional isomers were found [35].
While the cuticle lies on the outer face of the primary cell wall, suberin is located on the
inside of the primary cell wall, usually close to the plasma membrane. Plants synthesize
suberin to create a hydrophobic barrier to water and solute diffusion through cell walls during
normal development, or to provide a barrier in response to environmental stresses. A familiar
example of suberized tissue is cork, constituted by the outer bark cells (periderm) of cork oak
(Quercus suber). Suberin is also deposited in periderm walls of underground organs (i.e.
tubers, roots), and in the root endodermis. A material with composition intermediate between
suberin and cutin is often deposited surrounding the bundle sheaths of monocot leaves,
perhaps to prevent CO2 released during decarboxylation reactions from diffusing out of the
bundle sheath cells. In mature seeds, suberin has been found in seed coat layers, and sealing

132

R. A. Sidorov, A. V. Zhukov, V. P. Pchelkin et al.

off the chalazal region of the inner seed coat after disconnection from vascular tissue.
Exposure to cold, mineral stress and fungal infection are some examples where suberization
occurs as a response to external factors. It is also deposited as a wound response by injured
plant cells, even in those cells that normally synthesize cutin. [36].
The other lipid polymer, suberin, is a heteropolymer, consisting of an aliphatic polyester
associated with cross-linked polyaromatics and embedded waxes. Upon transesterification of
suberin, the monomers released include C16-C28 -hydroxy fatty acids and C16-C26 ,dicarboxylic acids, the latter of which are diagnostic monomers, unsubstituted very-longchain fatty acids (VLCFAs; C>18) and alcohols, glycerol and ferulate. Usually the major
components of suberin are -hydroxy derivatives of palmitic and/or oleic acids, but in some
cases -hydroxy C22:0 also is a dominant component [37]. Dicarboxylic FAs derived from
further oxidation of the -hydroxy-FAs are also found in suberin.
The synthesis of cutin and suberin aliphatic polyesters usually are described as distinct
pathways, with chain elongation and conversion of -hydroxy acids to dicarboxylic acids
being specific for suberin monomers. However, data from Arabidopsis has shown that
dicarboxylates are not exclusively found in suberin, and that the same gene families seem to
be involved in the synthesis of both types of polyesters. Hence, monomers with chain lengths
beyond C20, higher levels of aromatics, and primary alcohol synthesis can be considered as
specifically associated with suberin. Hydroxylated fatty acids are major constituents of plant
lipid polyesters. In plants, hydroxylation of the terminal methyl of aliphatic chain (-position)
is catalyzed by cytochrome-P450-dependent (CYP) enzymes, most of them belonging to the
CYP86 and CYP94 subfamilies. Although Arabidopsis leaf and stem cutin has an unusual
composition rich in unsaturated dicarboxylic acids, its flowers present a classical cutin
composition with high content of 10,16-dihydroxy fatty acid. CYP86A4 is involved in hydroxylation forming 16-hydroxy fatty acid, and CYP77A6 catalyzes the in-chain
hydroxylation of this monomer to produce 10,16-dihydroxy fatty acid [36]. At least two other
members of the CYP86 subfamily participate in suberin synthesis. CYP86A1 catalyzes root
suberin saturated and unsaturated C<20 monomer oxidation and is expressed in tissues where
suberization takes place; the potato CYP86A1-homologous gene plays a similar role in potato
periderm. CYP86B1 is involved in C22 and C24 fatty acid -oxidation in Arabidopsis root and
seed coat suberin [30]. Two different pathways for the synthesis of dicarboxylic acids have
been proposed. One postulates that CYP multifunctional enzymes are able to catalyze the
complete set of reactions leading to the oxidation of a terminal methyl group to the carboxyl
function, similar to tobacco CYP94A5 or Arabidopsis CYP94C1. The second route involves
two subsequent dehydrogenases that oxidize -hydroxy fatty acids to -oxo fatty acids and
then to ,-dicarboxylic acids. This was first supported by in vitro labeling experiments, and
later by genetic evidence shown in Arabidopsis cutin [36].

Where the Palmitic Acid Occur


It is well known that every cell almost never contains FA in the free form, however
some exceptions were described [38]. As a rule, their carboxyl group is esterified or otherwise
modified. In membranes, almost all the FAs are found esterified to glycerol; this class of lipid
is termed glycerolipids. Glycerolipids containing mono- and polyunsaturated FAs are the
obligatory components of all membranes of every plant cell. In particular the thylakoid

Palmitic Acid in Higher Plant Lipids

133

membranes of chloroplasts have one of the highest contents of lipids containing


polyunsaturated acyl groups (up to 90% of total FAs) known for any membrane. On the other
hand, every glycerolipid class contains palmitic acid in some content. On the other hand,
storage organs of oily plants may contain extremely high palmitic acid in their TAGs [3, 39,
40].
TAGs are the most important storage components of seeds in many plants, including
oilseeds, such as sunflower (Helianthus annuus), rapeseed (Brassica napus), soybean
(Glycine max), etc. [41]. At the same time, such plants as the olive (Olea europaea), avocado
(Persea americana), durian (Durio zibethinus), oil palm (Elaeis guineensis), sea buckthorn
(Hippophae rhamnoides) and some others, accumulate TAG not only in the seeds, but also in
juicy fruit parts outside the seeds (pericarp, hypanthium, etc.) [42-46] (see below).
The major natural resource of palmitic acid is the oil of mesocarp tissue of the African oil
palm (Elaeis guineensis) fruit, the main source of edible oil for the world. In particular it is
palm oil that gives its name to C16:0 FA. As a rule, palm oil TAGs contain 44-46% palmitic
acid and unlike to most other plant oils its concentration in sn-2 position of TAGs reach 1118% [47, 48]. The other high palmitic acid containing plant oils one can obtain from the
American oil palm (Elaeis oleifera) and the maripa palm (Attalea maripa) [49], from the
kernel of the coconut palm (Cocos nucifera) and other tropical plants such as cocoa
(Theobroma cacao) [3]. According Hilditch & Williams [39], there exist plants the juicy nonseed parts of the fruits of which contain TAGs with more than 70% palmitic acid (e.g. Sapium
sebiferum). However in plant kingdom one can find some members with extremely low
palmitate content in their TAGs, viz. Ximenia caffra, which seed oil contains ~35% unusual
VLCFAs but only 0.9% C16:0 [50].
Palmitic acid is an obligatory component not only of TAGs but of every lipid class of
every plant tissue. Thus overall FA composition of Rubiaceae leaf lipids contains 14.5-46.9%
palmitic acid [17]. The concentration of C16:0 in one or another lipid class depends on its
polarity and the more polar is lipid, the higher is palmitic acid level [51-54]. To the best of
our knowledge, Zhukov and Vereshchagin being the first to mention this fact in their study on
the composition of separate lipid fractions of different extractability [54]. After exhaustive
extraction of lipids of dry soybean seeds and separation of lipid extract obtained by Al2O3
column chromatography four fractions and nonextractable residue were obtained. A
subsequent analysis revealed that C16:0 content was higher in fractions of higher polarity, and
the highest it was in the nonextractable residue of plant tissues [54]. Later almost the same
pattern of relationship has been shown for other plant objects. For example, in hazelnut
(Corylus avellana) the palmitic acid content in TAGs was 5.8% of total FAs, whereas its
percentage in monogalactosyldiacylglycerols, PC and phosphatidylinositols (PI) was much
higher, namely 11.5, 13.1 and 16.1%, respectively [51]. In adzuki beans (Vigna angularis)
C16:0 level in TAGs, phosphatidyl ethanolamines, PC and PI was 28.0, 32.5, 25.2, and 48.2%,
respectively [55], and in broad beans (Vicia faba) its content was found to be 14.0, 17.0-18.4,
12.1-12,6, and 40.0-41,7%, respectively [56]. Many green leafy vegetables, sprouts, legumes
and other edible vegetables (cucumber, carrot, pepper, etc.) contain relatively high
concentrations of C16:0 in their lipids (15.5-43.8%) [52]. The newly differentiated vegetative
plant organs (roots, hypocotyle, epicotyle, leaves) also contain high levels of palmitic acid
(32-59% of total FAs) [53].
Palmitic acid along with VLCFAs esterified with higher fatty alcohols are known to be
the common components of epicuticular waxes whereas the esters of FAs and short-chain

134

R. A. Sidorov, A. V. Zhukov, V. P. Pchelkin et al.

aliphatic alcohols are rarely present in plant lipids. These fatty acid short-chain-alkyl esters
(FASCAE) were found in the fruits of 12 plant species of Celastraceae family [57], in the
neutral lipids of corn pollen [58], walnut fruit oil [59], callus culture from mint leaves [60],
dry rhizomes of ginseng [61], dry matter of liverwort [62]; small amounts of FASCAE were
also found in the volatile compounds of some other plant sources [57]. The concentrations of
FASCAE were low in almost all instances (<1% of total lipids). In all cases, the FASCAE
found in plants did not represent artifacts of the experiment and were represented mostly by
FA methyl esters which predominated in the FASCAE of the investigated plants, but FA
esters of other short-chain alcohols as a rule were also present. As a FA part of FASCAE
several C12-24 saturated and unsaturated fatty acyls were identified. In most cases palmitic acid
is one of the major FAs of FASCAE, and in dry rhizomes of ginseng the only FASCAE found
was methylpalmitate [61]. The qualitative and quantitative composition of various FASCAE
fractions varies considerably depending on the taxonomic position of a plant species, plant
tissue and/or organ, the extent of fruit maturity, etc.

TAGs are the Main Source of Palmitic Acid in Plants


TAG accumulation in the seeds occurs during fruit development. In its cells TAG
accumulate in the oily inclusions (oil bodies or oleosomes) discrete organelles of defined size
and composition and stored until the germination started. Whilst oil bodies appear to
superficially resemble lipid particles present in yeast and animals, their defining components
are unique to higher plants. Plant seed oil bodies are spherical organelles of between 0.6-2 m
in diameter, and comprise of a lumen in which the TAG is deposited, surrounded by a
phospholipid monolayer and an embedded layer or shell of proteins [63]. During seed
germination TAG can be used as a source of energy produced in the process of -oxidation,
or as a source of acetyl-CoA, or as a substrate in the reaction of acylation, for example, during
the synthesis of phospholipids [3, 64].
In plant seeds, glycerolipids can be synthesized in two similar pathways, which are
known as prokaryotic and eukaryotic systems. These pathways are localized in different
subcellular compartments and are characterized by a similar two-stage enzymatic conversion
of sn-glycero-3-phosphate (G3P) into phosphatidic acid (PA), but different further
conversions of PA into structural, storage, or signaling lipids [65]. Despite the closeness of
these two biosynthetic pathways, enzymes that catalyze acylation reactions are unique to each
system.
Enzymes of the prokaryotic pathway are localized in plastids, whereas enzymes of the
eukaryotic pathway in the cytosol and ER. In the prokaryotic pathway, FA acyls are directly
transferred from ACP to G3P, whereas in the eukaryotic pathway, FA are separated from
ACP by acyl-ACP thioesterases and released free FA then are transported in the cytoplasm,
where they are converted into acyl-CoA. During the synthesis of membrane and storage lipids
acyl groups are used in the ER by acyltransferases of the eukaryotic G3P pathway [66]. In
dependence on subcellular localization, these enzymes may differ in their structure, thus
forming independent clusters in phylogenetic investigations [67].

Palmitic Acid in Higher Plant Lipids

135

LPAAT acyltransferase of lysophosphatidic acid; GPAT glycerol-3-phosphate acyltransferase;


DGAT diacylglycerol acyltransferase; PPA phosphatase of phosphatidic acid.
Figure 4. Acyl_CoA_dependent pathway of TAG biosynthesis (Kennedy pathway) [69].

The accumulation of storage lipids in plants includes de novo biosynthesis of FA in the


plastid stroma with their subsequent involvement in the glycerolipid metabolism resulting in
TAG formation in the ER [68]. The two pathways of TAG biosynthesis were called as acylCoA-dependent and acyl-CoA-independent.

Acyl-CoA-Dependent Pathway of TAG Biosynthesis in Plants


In the acyl-CoA-dependent pathway of TAG biosynthesis (Figure 4), which is also
known as the Kennedy pathway, acyl-CoA is a substrate for successive reactions of glycerol
residue acylation; as a result, sn-1,2-diacylglycerols (DAG) are produced by means of
phosphatidic acid phosphatase, and a final acylation by DAG acyltransferases (DGAT)
producing TAGs [69]. Glycerol-3-phosphate (G3P) serves a source of the glycerol residue;
G3P is produced from one of the glycolysis products, dihydroxyacetone, as a result of
catalytic action of sn-glycerol-3-phosphate dehydrogenase (G3P-DH) [70].
The Kennedys pathway starts from G3P acylation. The acylation of sn-glycerol-3phosphate is catalyzed by acyl-CoA:sn-glycerol-3-phosphate acyltransferase (GPAT; EC
2.3.1.15). The second acylation is catalyzed by acyl-CoA:lyso-phosphatidic acid
acyltransferase (LPAAT; EC 2.3.1.51). It should be noted that LPAAT in plants seems to be
highly specific for unsaturated C16-C18 FAs and for this reason the percentage of saturated
FAs (palmitic, stearic, etc.) in sn-2 position of TAG molecules almost in all cases is quite low
(up to 3-4%) [40]. Phosphatidic acid phosphatase (PAP; EC 3.1.3.4) catalyzes the removal of
the phosphate group from phosphatidic acid to generate sn-1,2-diacylglycerol. The final acylCoA-dependent acylation is catalyzed by acyl-CoA:diacylglycerol acyltransferase (DGAT;
EC 3.2.1.20), using acyl-CoA as a donor of the acyl residue [7, 41, 69].

136

R. A. Sidorov, A. V. Zhukov, V. P. Pchelkin et al.

Acyl-CoA-Independent TAG Biosynthesis


As an alternative to the Kennedys pathway, FAs arising de novo in a course of FA
biosynthesis may be first integrated into the lipids of plastid membrane and/or those of the ER
and are only later accumulated as TAGs (Figure 5) [7].

AAPT aminoalcohol phosphotransferase; DGAT diacylglycerol acyltransferase; PDAT


phospholipid : diacylglycerol acyl transferase; CPT choline phosphotransferase.
Figure 5. Acyl-CoA-independent pathway of TAG biosynthesis [4, 7].

Newly synthesized FAs may be directly incorporated into phosphatidylcholines (PC),


bypassing PA and intermediate DAGs [71]. FA acyls from PC may be included into TAG via
the PC reverse conversion into DAG or through the functioning of
phospholipid:diacylglycerol acyltransferase (PDAT). This enzyme belongs to the family of
lecitine:cholesterol acyltransferases [72], which catalyze TAG formation by acyl transfer
from sn-2 position of the phospholipid molecule to DAG with phosphatidylethanolamine
(PE), a preferable donor of FA acyls in yeast and plants [73]. The PDAT activity was found
in microsomes of yeast and of some oily plant seeds [72, 73]. It was shown that arabidopsis
PDAT could use not only PE but also other phospholipids as donors of FA acyls; it could
accept diverse acyl groups with the chain length from 10 to 22 carbon atoms [74]. For
example, in yeast PDAT1 is a main enzyme involved in TAG accumulation during culture
exponential growth [75].

Palmitic Acid in Higher Plant Lipids

137

H, L, O, and P residues of hexadecenoic, linoleic, oleic, and palmitic acid, respectively.


Figure 6. Hypothetical pathway of TAG biosynthesis in the hypanthium of sea buckthorn fruit [80].

There is growing evidence of the existence of one more alternate acyl-independent


pathways of TAG formation in plants using the enzyme choline phosphotransferase or
aminoalcohol phosphotransferase. Choline phosphotransferase can catalyze TAG formation
through PC conversion into DAG, which can be later acylated by DGAT or PDAT. It has
been suggested that permanent reversible DAG transformation into PC can control, in
particular, the composition of polyunsaturated fatty acids (PUFA) in the seed oil [76], making
thereby choline phosphotransferase by the enzyme controlling the pathway, in which PUFA
become available for inclusion in the TAGs [76, 77].
Finally, it has been recently demonstrated that phospholipases (in particular,
phospholipase D) may play an important role in PC conversion into TAG [78] Due to the
suppression of phospholipase D activity by RNA interference, dilinoleyl-PC and dilinoleyl-

138

R. A. Sidorov, A. V. Zhukov, V. P. Pchelkin et al.

PE accumulated in soybean seeds at concentrations much higher than those observed in wildtype plants.

Peculiarities of TAG Biosynthesis in Plants with Oily Mesocarp


At the same time, as it was mentioned above, there are some plants (oil palm, avocado,
olive, sea buckthorn, durian, and others), in which a significant amount of oil is contained not
only in the seeds, but also in the juicy non-seed parts of the fruit arillus, pericarp, hypantium,
mesocarp, etc., TAG of which are not used by the plant itself as a source of organic matter
and energy. Such plants are called as plants with oily mesocarp (POM). With regard to TAG
composition, these juicy non-seed parts of many POM fruits contains highly saturated oil
with predominance of C16 FAs, palmitic and hexadecenoic (or 18:1 FAs, as a rule, oleic but in
some cases noticeable concentrations of its positional isomer, cis-vaccenic acid, also occur)
[42-46, 79], while the seeds of the same fruits accumulate TAG rich in polyunsaturated C18
FAs, linoleic and -linolenic [42, 80].
This feature of POM plants was studied in details on the example of the sea buckthorn
hypanthium by analyzing the changes of position-species composition of its TAG during fruit
ripening [80]. The experimental data obtained permitted the authors to suggest the hypothetic
pathway of biosynthesis of TAGs (Figure 6) containing unsaturated FA in the sn-2 position of
the molecule, which is different from the Kennedys pathway [69]. The authors suggest that
this process includes at least two stages. At the first stage, the pool 1 of free saturated and
unsaturated FAs statistically esterifies rac-1 OH groups of so far unknown hypothetical C3
precursors, thus forming the common metabolic pool of TAG precursors (pool 2) comprising
five different rac-1 components. At the second stage, the components of pool 2 are subjected
to selective sn-2 acylation at the expense of the unsaturated FA pool (pool 3); this process is
catalyzed by also so far unknown hypothetical acyltransferase X i.e., according to the
suggested hypothesis, sn-2 acylation is a terminal but not an intermediate stage of TAG
molecule biosynthesis. This stage is controlled by the amount of FA available and the degree
of affinity of separate FA to esterification of sn-2 OH groups of glycerol or its derivatives. In
addition, from the results of the performed analysis of position-species TAG composition
during fruit ripening, it follows that in the sea buckthorn hypantium a direct 12-desaturation
of the oleic acid residues, which esterifies rac-1 positions of TAG molecules occurs just after
the formation of TAG molecules [80], as this occurs in developing sunflower seeds at
temperature lowering [81].
However in other POM plants there may exist other mechanisms of TAG biosynthesis. In
particular, Tranbarger et al. [82] in their detailed study have shown that in the mesocarp tissue
of the oil palm (Elaeis guineensis) fruit three enzymes appear to participate in the acylation of
DAG to TAG. The authors ascertained that type 1 and type 2 DGAT both take part in TAG
synthesis in the oil palm mesocarp tissue. Moreover the route involving PC as the acyl donor
may make a contribution to the final acylation step of DAG, and therefore the PDAT enzyme
is also involved, whereas both enzymes recently found to contribute to PCDAG
interconversion in developing Arabidopsis seeds, phosphatidylcholine: DAG cholinephosphotransferase and CDP-choline:DAG cholinephosphotransferase, are not take part in
TAG biosynthesis in this tissue [82]. Although the literature shows that the three enzymes,
DGAT1, DGAT2, and PDAT, are all capable of catalyzing the final acylation step, their

Palmitic Acid in Higher Plant Lipids

139

individual contributions in tissues that accumulate oil in different plants may be unequal.
However the similar transcript amounts detected in the study of Tranbarger et al. suggest that
the contribution of these three enzymes could be of similar importance in the mesocarp of oil
palm [82].

REFERENCES
[1]
[2]
[3]
[4]
[5]
[6]

[7]
[8]

[9]
[10]
[11]
[12]

[13]
[14]
[15]

[16]

Millar, AA; Smith, MA; Kunst, L. All fatty acids are not equal: discrimination in plant
membrane lipids. Trends Plant Sci., 2000, 5, 95-101.
Voelker, T; Kinney AJ. Variations in the biosynthesis of seed-storage lipids. Annu.
Rev. Plant Physiol. Plant Mol. Biol., 2001, 52, 335-361.
Hitchcock, C; Nichols, B. Plant lipid biochemistry. London: Academic Press, 1971, 1387.
Sidorov, RA; Tsydendambaev, VD. Biosynthesis of fatty oils in higher plants. Russ. J.
Plant Physiol., 2014, 61, 1-18.
Orlogge, J; Browse, J. Lipid biosynthesis. Plant Cell, 1995, 7, 957-970.
Stymne, S; Griffiths, G; Stobart, K. Desaturation of fatty acids on complex-lipid
substrates. In: Stumpf PK, Mudd JB, Nes WD, editors. The Metabolism, Structure, and
Function of Plant Lipids. NY: Plenum Press, 1987, 405-412.
Durrett, TP; Benning, C; Ohlrogge, J. Plant triacylglycerols as feedstocks for the
production of biofuels. Plant J., 2008, 54, 593-607.
Tai, H; Jaworski, JG. 3-Ketoacyl-acyl carrier protein synthase III from spinach
(Spinacia oleracea) is not similar to other condensing enzymes of fatty acid synthase.
Plant Physiol., 1993, 103, 1361-1367.
Lung, SC; Weselake, RJ. Diacylglycerol acyltransferase: a key mediator of plant
triacylglycerol synthesis. Lipids, 2006, 41, 1073-1088.
Lynch, DV. Sphingolipids. In: Moore T, editor. Plant Lipids. Boca Raton, FL: CRC
Press, 1993, 285308.
Weselake, RJ; Taylor, DC; Rahman, MH; Shah, S; Laroche, A; McVetty, P; Harwood,
J. Increasing the flow of carbon into seed oil. Biotechnol. Adv., 2009, 27, 866-78.
Pollard, MR; Anderson, L; Fan, C; Hawkins, D; Davies, H. A specific acyl-ACP
thioesterase implicated in medium-chain fatty acid production in immature cotyledons
of Umbellularia californica. Arch. Biochem. Biophys., 1991, 284, 306-312.
Salas, JJ; Ohlrogge, JB. Characterization of substrate specificity of plant FatA and FatB
acyl-ACP thioesterases. Arch. Biochem. Biophys., 2002, 403, 25-34.
Hugly, S; Somerville, C. A role for membrane lipid polyunsaturation in chloroplast
biogenesis at low temperature. Plant Physiol., 1992, 99, 197-202.
Mongrand, S; Bessoule, JJ; Cabantous, F; Cassagne, C. The C16:3/ C18:3 fatty acid
balance in photosynthetic tissues from 468 plant species. Phytochemistry, 1998, 49,
1049-1064.
Heinz, E; Roughan, PG. Similarities and differences in lipid metabolism of chloroplasts
isolated from 18:3 and 16:3 plants. Plant Physiol., 1983, 72, 273279.

140

R. A. Sidorov, A. V. Zhukov, V. P. Pchelkin et al.

[17] Mongrand, S; Badoc, A; Patouille, B; Lacomblez, C; Chavent, M; Bessoule, JJ.


Chemotaxonomy of the Rubiaceae family based on leaf fatty acid composition.
Phytochemistry, 2005, 66, 549559.
[18] Wallis, JG; Browse, J. Mutants of Arabidopsis reveal many roles for membrane lipids.
Progr. Lipid Res., 2002, 41, 254-278.
[19] Cahoon, E; Ohlrogge, J. Metabolic evidence for the involvement of a 4-palmitoyl-acyl
carrier protein desaturase in petroselinic acid synthesis in coriander endosperm and
transgenic tobacco cells. Plant Physiol., 1994, 104, 827-837.
[20] Cahoon, E; Shanklin, J; Ohlrogge, J. Expression of a coriander desaturase results in
petroselinic acid production in transgenic tobacco. Proc. Natl. Acad. Sci. USA, 1992,
89, 11184-11188.
[21] Tsydendambaev, VD; Christie, WW; Brechany, EY; Vereshchagin, AG. Identification
of unusual fatty acids of four alpine plant species from the Pamirs. Phytochemistry,
2004, 65, 2697-2705.
[22] Wolff, RL; Christie, WW. Structure, practical sources (Gymnosperm seeds), gaschromatographic data (equivalent chain lengths), and mass spectrometric characteristics
of all-cis 5-olefinic acids. Eur. J. Lipid Sci. Technol., 2002, 104, 234-244.
[23] Sidorov, RA; Zhukov, AV; Pchelkin, VP; Vereshchagin, AG; Tsydendambaev, VD.
Content and fatty acid composition of neutral acylglycerols in Euonymus fruits. J. Am.
Oil Chem. Soc., 2014, 91, 805-814.
[24] Tsevegsuren, N; Aitzetmuller, K., Vosmann, K. Isomers of Hexadecenoic and
hexadecadienoic acids in Androsace septentrionalis (Primulaceae) seed oil. Lipids,
2003, 38, 1173-1178.
[25] Harwood, JL. Recent advances in the biosynthesis of plant fatty acids. Biochim.
Biophys. Acta, 1996, 1301, 7-56.
[26] Kunst, L; Samuels, AL. Biosynthesis and secretion of plant cuticular wax. Progr. Lipid
Res., 2003, 42, 5180.
[27] Bernard, A; Domergue, F; Pascal, S; Jetter, R; Renne, C; Faure, JD; Haslam, RP;
Napier, JA; Lessire, R; Joubs, J. Reconstitution of plant alkane biosynthesis in yeast
demonstrates that Arabidopsis ECERIFERUM1 and ECERIFERUM3 are core
components of a very-long-chain alkane synthesis complex. Plant Cell, 2012, 24, 3106
3118.
[28] Lee, SB; Suh, MC. Recent advances in cuticular wax biosynthesis and its regulation in
Arabidopsis. Molecular Plant, 2013, 6, 246-249.
[29] Li, F; Wu, X; Lam, P; Bird, D; Zheng, H; Samuels, L; Jetter, R; Kunst, L. Identification
of the wax ester synthase/acyl-coenzyme A: Diacylglycerol acyltransferase WSD1
required for stem wax ester biosynthesis in Arabidopsis. Plant Physiol., 2008, 148, 97
107.
[30] Hamberg, M; Sanz, A; Castresana, C. -Oxidation of fatty acids in higher plants:
Identification of a pathogen-inducible oxygenase (PIOX) as an -dioxygenase and
biosynthesis of 2-hydroperoxylinolenic acid. J. Biol. Chem., 1999, 274, 24503-24513.
[31] Cassagne, C; Bessoule, J-J; Schneider, F; Lessire, R; Sturbois, B; Moreau, P; Spinner,
C. Modulation of the very-long-chain fatty acid (VLCFA) formation in leek. In: Kader
J-K, Mazliak P, editors. Plant Lipid Metabolism. Dordrecht: Kluwer, 1995, 111-114.

Palmitic Acid in Higher Plant Lipids

141

[32] Hills, MJ; Roscoe, TJ. Synthesis of Structural and Storage Lipids by the ER. In:
Robinson DG, editor. The Plant Endoplasmic Reticulum. Berlin-Heidelberg: SpringerVerlag, 2006, 155-186.
[33] Kolattukudy, PE. Polyesters in higher plants. Adv. Biochem. Eng. Biotechnol., 2001, 71,
1-49.
[34] Holloway, PJ; Deas, AHB. Occurrence of positional isomers of dihydroxyhexadecanoic
acid in plant cutins and suberins. Phytochemistry, 1971, 10, 2781-2785.
[35] Deas, AHB; Baker, EA; Holloway, PJ. Identification of 16-hydroxyoxohexadecanoic
acid monomers in plant cutins. Phytochemistry, 1974, 13, 1901-1905.
[36] Molina, I. Biosynthesis of plant lipid polyesters. 2014 May, 5. Available from: URL
http://lipidlibrary.aocs.org/plantbio/polyesters/index.htm
[37] Kolattukudy, PE. Structure, biosynthesis, and biodegradation of cutin and suberin. Ann.
Rev. Plant Physiol., 1981, 32, 559-567.
[38] Oo, KC; Lee, KB; Ong, SS. Changes in fatty acid composition of the lipid classes in
developing oil palm mesocarp. Phytochemistry, 1986, 25, 405-407.
[39] Hilditch, TP; Williams, PN. The Chemical Constitution of Natural Fats. NY: Wiley;
1964; 1-745.
[40] Vereshchagin, AG. Biochemistry of triglycerides. M: Nauka, 1972, 1-308. (In Russian)
[41] Graham, I. Seed storage oil mobilization. Annu. Rev. Plant Biol., 2008, 59, 115-142.
[42] Berezhnaya, GA; Ozerinina, OV; Yeliseev, IP; Tsydendambaev, VD; Vereshchagin,
AG. Developmental changes in the absolute content and fatty acid composition of acyl
lipids of sea buckthorn fruits. Plant Physiol., Biochem., 1993, 31, 323-332.
[43] Gaydou, EM; Lozano, Y; Ratovohery, J. Triglyceride and fatty acid compositions in the
mesocarp of Persea americana during fruit development. Phytochemistry, 1987, 26,
15951597.
[44] Ross, J; Sanchez, J; Millan, F; Murphy, D. Differential presence of oleosins in
oleogenic seed and mesocarp tissues in olive (Olea europaea) and avocado (Persea
americana). Plant Sci., 1993, 93, 203-210.
[45] Osagie, AU; Bafor, ME. Triacylglycerols of oil palm mesocarp during fruit maturation.
Biochem. Cell Biol., 1990, 68, 313317.
[46] Berry, S. Fatty acid composition and organoleptic quality of four clones of durian
(Durio zibethinus). J. Am. Oil Chem. Soc., 1981, 58, 716-717.
[47] Barrett, CB; Dallas, MSJ; Padley, FB. The quantitative analysis of TG mixtures by TLC
on silica impregnated with silver nitrate. J. Amer. Oil Chem. Soc., 1963, 40, 580-584.
[48] Mattson, FH; Volpenhein, RA. The specific distribution of unsaturated fatty acids in the
triglycerides of plants. J. Lipid Res., 1963, 4, 392-396.
[49] Reeves, JB; Weihrauch, JL. Composition of Foods: Fats and Oils. Agriculture
handbook 8-4: Consumer and Food Economics Institute. Washington DC: U.S. Dept. of
Agriculture, Science and Education Administration; 1979; 4. Available from: OCLC
5301713.
[50] Mitei, YC; Ngila, JC; Yeboah, SO; Wessjohann, L; Schmidt, J. NMR, GCMS and
ESI-FTICR-MS profiling of fatty acids and triacylglycerols in some Botswana seed
oils. J. Amer. Oil Chem. Soc., 2008, 85, 10211032.
[51] Parcerisa, J; Richardson, DG; Rafecas, M; Codony, R; Boatella, J. Fatty acid
distribution in polar and nonpolar lipid classes of hazelnut oil (Corylus avellana L.). J.
Agric. Food Chem., 1997, 45, 3887-3890.

142

R. A. Sidorov, A. V. Zhukov, V. P. Pchelkin et al.

[52] Vidrih, R; Filip, S; Hribar, J. Content of higher fatty acids in green vegetables. Czech J.
Food Sci., 2009, 27, S125-S129.
[53] Liu, KS; Brown, EA. Fatty acid compositions in newly differentiated tissues of soybean
seedlings. J. Agric. Food Chem., 1996, 44, 1395-1398.
[54] Zhukov, AV; Vereshchagin, AG. Composition of separate fractions of the polar lipids.
Soviet J. Plant Physiol., 1980, 27, 307-314.
[55] Yoshida, H; Yoshida, N; Tomiyama, Y; Mizushina, Y. Characteristics of lipid
components, fatty acid distributions and triacylglycerol molecular species of adzuki
beans (Vigna angularis). Food Chemistry, 2009, 115, 1424-1429.
[56] Yoshida, H; Saiki, M; Yoshida, N; Tomiyama, Y; Mizushina, Y. Fatty acid distribution
in triacylglycerols and phospholipids of broad beans (Vicia faba). Food Chemistry,
2009, 112, 924-928.
[57] Sidorov, RA; Zhukov, AV; Pchelkin, VP; Vereshchagin, AG; Tsydendambaev, VD.
Occurrence of fatty acid short-chain alkyl esters of natural origin in the fruits of
Celastraceae plants. Chemistry & Biodiversity, 2013, 10, 978-988.
[58] Fathipour, A; Schlender, KK; Sell, HM. The occurrence of fatty acid methyl esters in
the pollen of Zea mays. Biochim. Biophys. Acta, 1967, 144, 476478.
[59] Rockland, LB; de Benedict, C. Occurrence of fatty acid methyl esters in walnut kernel
and other oils. J. Agric. Food Chem., 1970, 18, 228233.
[60] Suga, T; Hirata, T; Yamamoto, Y. Lipid constituents of callus tissue of Mentha spicata.
Agric. Biol. Chem., 1980, 44, 18171820.
[61] Shukla, YN; Thakur, RS. Fatty acids and esters from Panax pseudo_ginseng rhizomes.
Phytochemistry, 1985, 24, 10911092.
[62] Matsuo, A; Nakayama, M; Hayashi, S; Nagai, K. Fatty acid ethyl esters in the liverwort
Conocephalum conicum. Phytochemistry, 1980, 19, 18481849.
[63] Murphy, DJ; Vance, J. Mechanisms of lipid-body formation. Trends Biochem. Sci.,
1999, 24, 109-115.
[64] Athenstaedt, K; Daum, G. The life cycle of neutral lipids: synthesis, storage and
degradation. Cell Mol. Life Sci., 2006, 63, 1355-1369.
[65] Banas, A; Dahlqvist, A; Sthl, U; Lenman, M; Stymne, S. The involvement of
phospholipid: diacylglycerol acyltransferases in triacylglycerol production. Biochem.
Soc. Trans., 2000, 28, 703-705.
[66] Phillips, BE; Smith, CR. Stereospecific analysis of triglycerides from Mannina
emarginata seed oil. Lipids, 1972, 7, 215-217.
[67] Cagliari, A; Pinheiro-Margis, M; Loss, G; Mastroberti, A; de Araujo Mariath, J;
Margis, R. Identification and expression analysis of castor bean (Ricinus communis)
genes encoding enzymes from the triacylglycerol biosynthesis pathway. Plant Science,
2010, 179, 499-509.
[68] Lung, SC; Weselake, RJ. Diacylglycerol acyltransferase: a key mediator of plant
triacylglycerol synthesis. Lipids, 2006, 41, 1073-1088.
[69] Kennedy, E. Biosynthesis of complex lipids. Fed. Proc., 1961, 20,
934-940.
[70] Weselake, RJ; Taylor, DC; Rahman, MH; Shah, S; Laroche, A; McVetty, P; Harwood,
J. Increasing the flow of carbon into seed oil. Biotechnol. Adv., 2009, 27, 866-878.

Palmitic Acid in Higher Plant Lipids

143

[71] Bates, PD; Ohlrogge, JB; Pollard, M. Incorporation of newly synthesized fatty acids
into cytosolic glycerolipids in pea leaves occurs via acyl editing. J. Biol. Chem., 2007,
282, 3120631216.
[72] Mhaske, V; Beldjilali, K; Ohlrogge, J; Pollard, M. Isolation and characterization of an
Arabidopsis thaliana knockout line for phospholipid: diacylglycerol transacylase gene
(At5g13640). Plant Physiol. Biochem., 2005, 43, 413-417.
[73] Dahlqvist, A; Stahl, U; Lenman, M; Banas, A; Lee, M; Sandager, L; Ronne, H; Stymne,
S. Phospholipid:diacylglycerol acyltransferase: an enzyme that catalyzes the acyl-CoAindependent formation of triacylglycerol in yeast and plants. Proc. Natl. Acad. Sci.
USA, 2000, 97, 64876492.
[74] Li, R; Yu, K; Hildebrand, DF. DGAT1, DGAT2 and PDAT expression in seeds and
other tissues of epoxy and hydroxy fatty acid accumulating plants. Lipids, 2010, 45,
145-157.
[75] Oelkers, P; Cromley, D; Padamsee, M; Billheimer, JT; Sturley, SL. The DGA1 gene
determines a second triglyceride synthetic pathway in yeast. J. Biol. Chem., 2002, 277,
88778881.
[76] McMaster, CR; Bell, RM. CDP-choline:1,2-diacylglycerol choline phosphotransferase.
Biochim. Biophys. Acta, 1997, 1348, 100110.
[77] Somerville, CR; Browse, J; Jaworski, JC; Ohlrogge, J. Lipids. In: Buchanan BD;
Gruissem W; Jones RL editors. Biochemistry and Molecular Biology of Plants.
Rockville: Amer. Soc. Plant Physiol., 2000, 456-526.
[78] Lee, J; Welti, R; Schapaugh, WT; Trick, HN. Phospholipid and triacylglycerol profiles
modified by PLD suppression in soybean seed. Plant Biotechnol. J., 2011, 9, 359372.
[79] Villa-Rodrguez, JA; Molina-Corral, FJ; Ayala-Zavala, JF; Olivas, GI; GonzlezAguilar, GA. Effect of maturity stage on the content of fatty acids and antioxidant
activity of Hass avocado. Food Res. Intern., 2011, 44, 1231-1237.
[80] Vereshchagin, A; Ozerinina, O; Tsydendambaev, V. Developmental changes in the
triacylglycerol composition of sea buckthorn fruit mesocarp. J. Plant Physiol., 1999,
155, 453-461.
[81] Garces, R; Sarmiento, C; Mancha, M. Oleate from triacylglycerols is desaturated in
cold-induced developing sunflover (Helianthus annuus L.) seeds. Planta, 1994, 193,
473-477.
[82] Tranbarger, TJ; Dussert, S; Jot, T; Argout, X; Summo, M; Champion, A; Cros, D;
Omore, A; Nouy, B; Morcillo, F. Regulatory mechanisms underlying oil palm fruit
mesocarp maturation, ripening, and functional specialization in lipid and carotenoid
metabolism. Plant Physiol., 2011, 156, 564584.

In: Palmitic Acid: Occurrence, Biochemistry and Health Effects ISBN: 978-1-63321-519-1
Editor: Lucas F. Porto
2014 Nova Science Publishers, Inc.

Chapter 7

PALMITIC ACID IN INFANT NUTRITION


Fabiana Bar-Yoseph1, Yael Lifshitz1, Tzafra Cohen1
and Ita Litmanovitz2,3
1

Enzymotec Ltd., Sagi 2000 Industrial Park Kfar Baruch, Israel.


2
Departments of Neonatology,
Meir Medical Center, Kfar Saba, Israel and 3Sakler School of Medicine,
Tel Aviv University, Israel

ABSTRACT
Human breast milk provides the optimum nutrition for infants. Designed to provide
balanced nutrition, human breast milk naturally meets the needs of growing infants in the
first months after birth. In human breast milk, and in most infant formulas, approximately
50% of the energy is supplied to newborns as fat, of which more than 98% is in the form
of triglycerides. Triglyceride synthesis occurs in the mammary gland. The fatty acids are
specifically positioned to sn1, sn2 or sn3 positions on the glycerol backbone to yield the
structure-specific triglycerides that are found in human milk. Palmitic acid (C16:0) is the
predominant saturated fatty acid, comprising 17-25% of the fatty acids in mature human
milk. Surprisingly, the positioning of palmitic acid is highly conserved across
populations, and approximately 70-75% of palmitic fatty acids are esterified to the sn2
position of the triglyceride (sn2 palmitate).
Clinical and pre-clinical studies over the last three decades have provided increasing
evidence that this specific positioning of palmitic acid on the triglycerides in human milk
has a significant holistic effect on optimal infant development and well-being that is
related to the increased absorption of both palmitic acid and calcium: softer stools,
increased bone strength, increased beneficial gut flora, controlled intestinal health, and
reduced infant crying. All of these contribute to the benefits of infant wellbeing.
The overall aim of the current review is to expand the understanding of the role of
palmitic acid and its specific sn2 position in infant nutrition.

146

Fabiana Bar-Yoseph, Yael Lifshitz, Tzafra Cohen et al.

INTRODUCTION
Palmitic acid (16:0) (PA) is the most abundant and widespread natural saturated acid; it is
present in plants, animals, and microorganisms. Twenty to thirty percent of the mammalian
body lipids and 10 to 40% of vegetable oils are palmitic acid. Palm oil is a rich commodity oil
source, and over 40% of it is palmitic acid (Scrimgeour and Harwood 2007). Palmitic acid
was discovered in 1840 by Edmond Fremy (Fremy 1842) in saponified palm oil. The word
palmitic is derived from the French word palmitique, for the pith of the palm tree. As its
name indicates, it is a major component of the oil derived from palm trees (palm oil and palm
kernel oil). Recent evidence suggests that PA plays an important role beyond serving as an
energy source.
Palmitic acid is the predominant saturated fatty acid in mature human milk, comprising
17-25% of the total fatty acids, of which 70-75% is esterified to the sn2 () position of the
triglyceride (Breckenridge et al. 1969). The significance of this fatty acid and its position is
evident from the fact that this positioning is conserved in all women, regardless of their ethnic
origin or nutrition, unlike the general fatty acid profile of human milk (Jensen et al. 1978,
Jensen 1999). In contrast, the palmitic acid in vegetable oils that are commonly used in the
manufacturing of infant formulas is esterified primarily at the sn1 and sn3 positions, while the
sn2 position is predominantly occupied by unsaturated fatty acids (Jensen 1995).

THE SPECIAL STRUCTURE OF HUMAN MILK FAT


Human breast milk provides the optimal nutrition for infants. Breast milk is designed to
provide a balanced diet and meet the needs of the growing infant. Approximately 50% of
dietary calories from both human milk and infant formulas are supplied as fat. Human milk
lipids consist of triacylglycerols (TG) (98%), phospholipids (0.8%), cholesterol (0.5%), and
other lipid components (Giovannini et al. 1995).
TGs are composed of a 3 carbon glycerol to which 3 fatty acids are esterified, and the
glycerol carbons are stereo-specifically numbered as sn1, sn2, and sn3 (Innis 2011). The
mammary gland actively uptakes FAs from plasma for the de novo synthesis of TGs (Neville
and Picciano 1997), where, there is a preferential acylation of fatty acids to specific positions
in TGs, making it seem as there is a biological purpose for investing in the enzymes needed to
achieve the nonrandom positioning of TG fatty acids (Innis 2011).
Human milk fat is very well absorbed by infants despite its high saturated fatty acid
content (45% of fatty acids with a melting point above body temperature). This good
absorption is attributed at least in part to the presence of the bile salt lipase in human milk and
to the unique triglyceride synthesis and the highly specific positional distribution on the
glycerol backbone (Breckenridge et al. 1969).

NUTRIENT ABSORPTION
Infants require a bio-available source of energy and nutrients to meet the requirements of
their rapid growth, development and expanding skeletal mass. An infants nutritional

Palmitic Acid in Infant Nutrition

147

environment involves a high fat diet with frequent feedings; therefore, efficient fat absorption
is required. In breastfed infants, this is achieved using complex fat globule lipids and TG
structures that enable the efficient absorption and processing of a high-fat diet with highly
saturated fatty acids without requiring a high metabolic effort from the infant. For formulafed infants, the availability of nutrients and energy depends on the composition of the formula
(Giovannini et al. 1995).
Gastric lipase, reponible for intial TG digestion, hydrolyzes fatty acids from the sn3
position of dietary TG to release sn1,2 diacylglycerols and accounts for 10% or more of
dietary TG hydrolysis, depending on the composition of the milk or formula (Mu and Hoy
2004). Fat digestion continues with pancreatic colipase-dependent lipase (pancreatic lipase),
which has specificity for TG sn1 and sn3 ester linkages and completes TG hydrolysis to give
sn2 monoacylglycerol (2-MAG) and unesterified fatty acids, each of which are absorbed
separately. The absorption of unesterified fatty acids depends on their physical and chemical
properties (Innis 2011). The palmitic acid and stearic acid have melting points above the
intestine temperature (63C and 70C, respectively), in contrast to unsaturated fatty acids
such as oleic acid, linoleic acid and linolenic acid, which have melting points below the
intestinal temperature. Therefore, together with the pH of the intestine, unesterified longchain saturated fatty acids have an increased tendency to form insoluble fatty acid soaps that
are subsequently lost in the stools (Innis 2011). Fat absorption is influenced by the position of
the individual fatty acids on the triglyceride molecule and the importance of the positional
distribution of the fatty acids in human milk or infant formula fat applies particularly to
palmitic acid because it is one of the major constituents (Jensen et al. 1986). Palmitic acid is
absorbed from human milk as sn2 monoacylglycerol (Innis et al. 1994), and it is conserved
through digestion and absorption (Nelson and Innis 1999).

Figure 1. Soaped fatty acids in 24 hr stool collection at 6 weeks postnatal. The significance was
calculated for the two groups by the Mann Whitney test. Different letters indicate statistical significance
(p<0.05) between groups.

148

Fabiana Bar-Yoseph, Yael Lifshitz, Tzafra Cohen et al.

Clinical studies in term (Carnielli et al. 1996, Kennedy et al. 1999, Lopez-Lopez et al.
2001) and preterm infants (Carnielli et al. 1995, Lucas et al. 1997) have demonstrated the
correlation between the level of palmitic in the sn2 position on fatty acids and calcium
absorption. The reduction in calcium and fatty acid absorption is accompanied by increased
calcium soaps and, consequently, hard stools (Quinlan et al. 1995). Infants receiving the high
sn2 formula had softer stools and fewer formed and hard stools than did control regular
vegetable oil formula group (Sidnell and Greenstreet 2011, Kennedy et al. 1999, Litmanovitz
et al. 2014, and Yao et al. 2014).
Prebiotics are routinely added to infant formulas. A prebiotic is a non-digestible food
ingredient that has several potential beneficial effects on neonatal intestinal development,
including protection against infection and facilitation of nutrient absorption (Calder et al.
2006). Sn2 palmitate was recently shown to enhance fat absorption and reduce calcium soap
formation in infant formulas containing prebiotics (GOS) (Bar-Yoseph et al. 2014). In a
multicenter clinical study on Chinese term infants, sn2 palmitate formula consumption
resulted in benefits to the infant in terms of the nutrient absorption, specifically fat absorption.
Comparable with breastfeeding, the formula with sn2 palmitate reduced fat excretion,
primarily in form of calcium soap (Figure 1 (Bar-Yoseph et al. 2014) and Yao et al. 2014).

FATTY ACIDS BLOOD PROFILE


Whereas considerable data is available about the digestion and absorption of human milk
TGs, very little is known about the significance of the milk TG structures after absorption.
Assuming an average intake of 760 mL/day of human milk with 3.6 g/dL triacylglycerol
(TG), the breast-fed infant consumes approximately 27 g/d TG, representing approximately
50% of the infants daily energy intake and 35 times more TG/kg body weight than most
adults consume. Unlike usual post-weaning feeding, breast-fed infants are frequently fed,
resulting in a prolonged post-prandial state with a high lipid (milk) load (Innis 2013).
TG digestion by gastric and pancreatic lipase results in sn2 monoacylglycerols (2-MAG)
and unesterified fatty acids, which are absorbed into the enterocytes and reassembled into
TGs for secretion in chylomicrons (Innis 2011).
The effect of sn2 palmitate was shown by Carnielli et al., in a crossover clinical trial with
premature infants who were each fed for 1 week with a standard formula or a formula
containing sn2 palmitate. Infants fed with the sn2 palmitate formula had higher percentages of
palmitic acid in plasma sterol esters, triacylglycerols, and free fatty acids and they had lower
linoleic acid in triacylglycerols compared with the group receiving standard formula. These
results can be explained by the better fat absorption of the major dietary saturated fatty acids
(Carnielli et al. 1995). Previous studies have also shown high levels of palmitic acid at the
sn2 position of TG in plasma chylomicrons of breast-fed infants, showing that milk TG
structures are conserved through digestion and absorption in young infants (Innis et al. 1994,
Nelson and Innis 1999). A similar palmitic acid level in the chylomicron TG total fatty acids
was observed, indicating a physiological purpose of the milk TG structure and palmitic acid
post-absorption (Innis 2011). An additional study showed that when TGs with sn2 palmitic
acid and sn1,3 oleic acid were compared with sn2 oleic acid and sn1,3 palmitic acid, they
were associated with higher palmitic and stearic acids as well as, unexpectedly, higher

Palmitic Acid in Infant Nutrition

149

docosahexanoic acid in plasma unesterified fatty acids. This finding suggests the need for
paying further attention to the plasma unesterified fatty acids in young infants as a possible
source of fatty acids for membrane phospholipid synthesis and turnover (Innis and Nelson
2013). It is therefore possible to have the same average fatty acid composition in plasma TGs
with very different TG structures (Innis 2011).

BONE HEALTH
During the last decade, substantial efforts were made to determine the factors that
influence bone mineral accretion in healthy children. This may be attributed to the suggestion
that osteoporosis originates in childhood (Janz 2002), thereby providing optimal nutrition for
reaching the highest possible peak bone mass. The absorption of nutritional factors, such as
minerals, fats, carbohydrates, and proteins, is significantly important for normal infant growth
and development and may contribute to early bone mineral accretion (Specker 2004).
Kennedy et al., by using dual-energy X-ray absorptiometry (DEXA) to assess bone
mineralization showed in a randomized, controlled, double-blind study higher body bone
mass in infants after 12 weeks of feeding with sn2 palmitate formula (Kennedy et al. 1999),
Litmanovitz et al., showed that bone speed of sound (SOS) at 12 weeks in term newborns fed
with sn2 palmitate formula was significantly higher than that of newborns fed with regular
formula and comparable to that of breastfed newborns (Litmanovitz et al. 2013). In this study,
the SOS measurements showed a decrease in SOS in the first 12 weeks for all infants,
regardless of the type of feeding, but the decline was slighter in the sn2 palmitate and
breastfed groups than in the control group (figure 2), indicating higher bone strength in both
sn2 palmitate and breastfed groups.

Figure 2. Bone speed of sound (SOS) during the first 12 postnatal weeks for infants fed with control
formula, sn2 palmitate formula or breast milk.

150

Fabiana Bar-Yoseph, Yael Lifshitz, Tzafra Cohen et al.

This finding is in agreement with studies in both preterm infants () and term infants that
demonstrated a decrease in SOS (Eliakim et al. 2002, Litmanovitz et al. 2003, Liao et al.
2005) and DEXA (Rauch and Schoenau 2001). The reason for these phenomena is not clear.
It was suggested that the decline in bone mineral density (BMD) in healthy newborns is
associated with a relative physiological decrease in the cortical area and a redistribution of
bone tissue from the endocortical to the periosteal surface rather than with bone loss (Liao et
al. 2005). It is also possible that this decrease represents a delay between rapid linear bone
growth and mineralization.

QUALITY OF LIFE
One of the major concerns of parents is their baby's comfort and wellbeing. Various
parameters may provide comfort to the newborn infants. Those parameters include stool
characteristics, such as consistency, frequency and volume, as well as sleeping and crying
duration and their frequency during the day. Thus, comfort parameters can be the result of
neurological or gastrointestinal pathways and involve much more complex physiological
mechanisms.
Clinical studies have revealed beneficial effects of sn2 palmitate on the comfort and wellbeing of healthy term infants through observations of softer stools (Carnielli et al. 1996,
Kennedy et al. 1999). Recently, in a randomized controlled study, formula containing sn2
palmitate was shown to reduce crying in infants compared to a control formula (Bar-Yoseph
et al. 2013). Additionally, the study showed that the consumption of formula with sn2
palmitate improved infant crying patterns during the day. The crying duration and patterns in
the sn2 palmitate group were similar to those of breastfed infants, and the percentage of
crying infants was significantly lower in the sn2 palmitate group compared to the control
group during the afternoon hours (Litmanovitz et al. 2014).
Crying is a basic, instinctive response that is governed by basic neuro-chemical
mechanisms similar to those that control feeding (Brazelton 1962). Although crying is a
common spontaneous behavior, it can induce parental concern, which often results in requests
for assistance from health services (Brazelton 1962). Most infants follow a universal crying
pattern during the first few months of life, in which crying peaks at 6 weeks of age and then
declines leading up to 3 months of age (Brazelton 1962, Barr et al. 1989, St James-Roberts
and Halil 1991, St James-Roberts et al. 1993, Baildam et al. 1995, Evanoo 2007). Crying also
has a typical diurnal pattern: 40% of crying episodes occur between 1600h and 2200h, and
after the third month of life, crying episodes are distributed throughout the day. Infant crying
is usually believed to be related to abdominal discomfort (Rasquin et al. 2006). Due to the
mechanisms involved in crying, it is conceivable that diet, by modifying neurochemistry,
could alter crying that is either endogenous for no apparent reason or associated with
physiological stimuli, such as hunger/separation. Dietary approaches to reducing crying
episodes in formula-fed infants have already been introduced. Savino et al., (2006) evaluated
the efficacy of a formula containing partially hydrolyzed whey proteins, prebiotic
oligosaccharides (OS), and a high sn2-palmitic acid content (41% in the sn2 position), and the
authors observed a significant difference in crying, which most likely resulted from the
significant decrease in colic episodes (Savino, Palumeri et al. 2006). More recently, the effect

Palmitic Acid in Infant Nutrition

151

of sn2 palmitate on infant crying was re-assessed in a double blind study using infant
formulas that already contained prebiotics (GOS). This study demonstrated an additional
beneficial effect of sn2 palmitate beyond the effect of prebiotics in crying reduction (BarYoseph et al. 2014).
The authors concluded that if the reduction of crying in infants fed formula with sn2plamitate is simply due to the looser stools, one would not expect an effect using formulas
with prebiotics that result in softer stools, and there would definitely not be a diurnal pattern
of altered crying (random during the day and likely associated with time of passing stool).
Therefore, this effect could not be attributed to softer stools alone, and the mechanism had not
yet been revealed.

GASTROINTESTINAL HEALTH
Over the past two decades, easy digestion and improved nutrient absorption following
sn2 palmitate-based infant formulas have been shown in preclinical and clinical studies in
preterm (Carnielli et al. 1995, Lucas et al. 1997) and term infants (Carnielli et al. 1996,
Kennedy et al. 1999, Lopez-Lopez et al. 2001); recently, this effect was shown even in infant
formulas containing prebiotics (Bar-Yoseph et al. 2014). The benefits of sn2 palmitate on
better digestion, promote a comfortable environment for the gut, thereby indicating a possible
health benefit to the gut. The potential mechanism of sn2 palmitate in improving gut function
is not clear; however, in view of its significant contribution to the digestion process, it is
reasonable to believe that it has further effects on the gastrointestinal surroundings.
The human gut is the natural habitat for a large and dynamic microorganisms' community
(Guarner and Malagelada 2003). The structure and composition of the gut flora reflects
natural selection at both the microbial and host levels, which promotes mutual cooperation
within and functional stability of this complex ecosystem (O'Hara and Shanahan 2006). The
fetal gut is sterile colonization begins immediately after birth and is influenced by the mode
of delivery, the infant diet, hygiene levels and medication (Niers et al. 2007). Throughout life,
the gastrointestinal tract becomes heavily inhabited by a complex community of
microorganisms, which are far in excess of the eukaryotic cells of the human body
(Dethlefsen et al. 2006). The intestinal microflora is an essential organ that serves
numerous important functions for the human host, including protection against pathogens and
provision of enhanced metabolic capabilities. It acts as important source of energy and
regulates intestinal epithelial proliferation, gut maturation and modulation of the
inflammatory immune response (Lodinova et al. 1967, Dethlefsen et al. 2006, Kau et al.).
Moreover, the characteristics of the intestinal bacterial community have been associated with
allergies, late-onset autism, inflammatory bowel disease and cancer (Dethlefsen et al. 2006).
It has been shown that bifidobacteria and lactobacillus protect infants from pathogenic
intestinal microorganisms and therby decrease the incidence of infantile diarrhea (Yoshioka et
al. 1983; Huang et al. 2002). They are associated with a higher production of short-chain fatty
acids (acetic and lactic acids), which are a source of energy for colonocytes. Furthermore,
strains of bifidobacteria and lactobacillus influence gut maturation processes in infants and
have anti-inflammatory effects (Hedin et al. 2007). Specific components of the intestinal
microflora, including Lactobacilli and Bifidobacteria, are beneficial for the host, such as

152

Fabiana Bar-Yoseph, Yael Lifshitz, Tzafra Cohen et al.

promoting gut maturation and integrity, acting against pathogens and participating in immune
modulation (Calder et al. 2006, Pai and Kang 2008).
The human diet plays a major role in the gut microbiota composition and development
(Fanaro et al. 2003,Kau et al. 2011, Moore et al. 2011). Following the first days after birth,
the flora composition rapidly changes under the influence of the infant's diet; consequently,
the composition is different between breastfed and formula-fed babies (Harmsen et al. 2000,
Fanaro et al. 2003). Within 1 week after birth, bifidobacteria becomes the predominant
species in the intestine of breastfed infants, whereas formula-fed infants have a more diverse
flora without any prevalent microorganisms (Harmsen et al. 2000, Fanaro et al. 2003, Morelli
2008). Yaron et al., showed that after 6 weeks of feeding the infants fed the sn2 palmitate
formula have a profile of intestinal microorganisms similar to that found in breastfed infants,
with high Lactobacilli and Bifidobacteria compared with infants fed control formula (Yaron
et al. 2013). A similar observation was reported in a recently published study by Yao et al.,
(Yao et al. 2014). The effect may be attributed to direct or indirect mechanisms. The 2-MAG
may directly induce the adhesion and proliferation of bifidobacteria and lactobacillus and/or
inhibit other competitive bacterial species in the gut. Alternatively, the indirect mechanism
might be from the non-absorbed vegetable oil products (i.e., calcium-palmitate insoluble
complexes) that reduce the growth of the beneficial bacteria in the intestine or from fat
degradation products that activate metabolic pathways in the intestine, affecting bacterial
growth. Although the mechanism by which sn2 palmitate affects the microbiota composition
is not well understood, these data indicate the beneficial long-term health effects for infants
fed with a sn2 palmitate-containing formula.
Furthermore, the direct interaction of microflora with the intestinal mucosa might
stimulate inflammatory activity in the gut lesions in IBD (Monteleone et al. 2014). Therefore,
there might be a relationship between the beneficial effect of sn2 palmitate on the gut flora
and its beneficial effect of protection from inflammation.
Proper development of immune tolerance is necessary for the maintenance of gut
homeostasis and an efficient response against pathogens. Dysregulation of the involved
mechanisms can lead to inappropriate intestinal inflammation against microbiota, such as
inflammatory bowel disease. The immaturity of the neonatal immune system explains the
age-dependent differences of the immune responses against pathogens and the susceptibility
to different types of infection (Tourneur and Chassin 2013).
Recently, it was shown in an animal model of Mucin2 deficient mice (Muc2-/-) with
colitis (Lu et al. 2013) that sn2 palmitate controls the intestinal inflammation. Mucins are the
principal components of the intestinal mucus layer (Velcich et al. 1997), which forms a
physical barrier that protects the underlying epithelium against luminal substances and
microbes (Hecht 1999, Hollingsworth and Swanson 2004, Dharmani et al. 2009). A
deficiency in Muc2 affects the protective capacities of the mucus layer (Johansson et al.
2011), leading to the development of spontaneous colitis in Muc2-/- mice, a well-described
animal model for enterocolitis (Velcich et al. 2002, Van der Sluis et al. 2006, Lu et al. 2011).
In this study, low sn2 palmitate diet increased the incidence of erosion and mucosal
damage in the distal colon of Muc2-/- mice compared with the AIN-93G reference diet, while
the sn2 palmitate diet controled the damage by enhancing the immunosuppressive Treg
response in Muc2-/- mice, suggesting beneficial effects of sn2-palmitic acid in limiting
intestinal inflammatory diseases (Figure 3, (Lu et al. 2013)).

Palmitic Acid in Infant Nutrition

153

Moreover, the sn2 palmitate diet induced an immunosuppressive Treg response (Lu et al.
2013). Increasing evidence has shown that Treg cells have a protective role in inflammatory
diseases (Sakaguchi 2005), and patients with inflammatory bowel diseases (IBD) have
reduced Treg cell numbers compared with patients who have non-IBD inflammatory diseases
(Boden and Snapper 2008).
These data suggest that replacing the control fat with sn2 palmitate (i.e., fat containing
high levels of palmitic acid esterified to the sn2 position) may enhance the
immunosuppressive Treg response in the intestine, thereby preventing or limit intestinal
inflammation. Furthermore, sn2 palmitate might have significant beneficial effects on
necrotizing enterocolitis (NEC), which is the leading cause of morbidity, mortality and longterm complications in preterm infants (Neu and Walker 2011).

Figure 3. Morphology of the distal colon of Muc2-/- mice fed AIN93G, CF (control diet, low sn2
palmitate), or sn2 palmitate (INFAT). Distal colonic sections of mice fed with the different diets were
stained with hematoxylin and eosin. Shown are representative sections for each diet group.

Data from other fatty acids, such as LC-PUFA, with similar effects merit further
investigation. Some postulated mechanisms of this observed effect are presumably attributed
to alterations in signal transduction pathways, membrane fluidity, intraluminal bacteria, and
gene expression (Teitelbaum and Allan Walker 2001).

CONCLUSION
The sn2 palmitate research area began with a focus on increased calcium and fat
absorption and its role in bone mineralization. The increasingly broad research efforts on sn2
palmitate gradually revealed additional, and significant clinical effects, such as intestinal flora
composition, intestinal inflammation, and the infant's comfort in the form of crying patterns.
The efficient absorption of fat by breast-fed infants emphasizes the importance of the human
milk TG structures for digestion. The structure of sn2sn2 palmitate is also conserved
following digestion, entering the blood stream to reach target peripheral tissues. This
observation strongly suggests that the significance of human milk TG structures goes beyond
maintaining efficient palmitic acid absorption. As demonstrated in this review, the effect of
sn2sn2 palmitate extends to different biological benefits beyond fat and calcium absorption,
while the mechanism and the active pathways involved in those effects remain to be
elucidated.

154

Fabiana Bar-Yoseph, Yael Lifshitz, Tzafra Cohen et al.

REFERENCES
Baildam, E. M., V. F. Hillier, B. S. Ward, R. P. Bannister, F. N. Bamford and W. M. Moore
(1995). "Duration and pattern of crying in the first year of life." Dev. Med. Child Neurol.
37(4): 345-353.
Bar-Yoseph, F., Y. Lifshitz and T. Cohen (2013). "Review of sn2 palmitate oil implications
for infant health." Prostaglandins Leukot Essent Fatty Acids 89(4): 139-143.
Bar-Yoseph, F., Y. Lifshitz, T. Cohen, P. Malard, Z. Li , H. Cui, A. Zhang, H. Yang, J. Wu
and C. Xu (2014). sn2 Palmitate Reduces Crying And Soaped Fatty Acids Excretion In
Chinese Infants Fed Formula Enriched With Prebiotics: A Double-Blind Randomized
Clinical Trial. ESPGHAN 2014. Jerusalem, Israel.
Barr, R. G., M. S. Kramer, I. B. Pless, C. Boisjoly and D. Leduc (1989). "Feeding and
temperament as determinants of early infant crying/fussing behavior." Pediatrics 84(3):
514-521.
Boden, E. K. and S. B. Snapper (2008). "Regulatory T cells in inflammatory bowel disease."
Curr Opin Gastroenterol 24(6): 733-741.
Brazelton, T. B. (1962). "Crying in infancy." Pediatrics 29: 579-588.
Breckenridge, W. C., L. Marai and A. Kuksis (1969). "Triglyceride structure of human milk
fat." Can J. Biochem. 47(8): 761-769.
Calder, P. C., S. K. Etschmann, E. C. de Jong and C. Dupont (2006). "Early nutrition and
immunity progress and perspectives." 96: 774-790.
Carnielli, V. P., I. H. Luijendijk, J. B. van Goudoever, E. J. Sulkers, A. A. Boerlage, H. J.
Degenhart and P. J. Sauer (1995). "Feeding premature newborn infants palmitic acid in
amounts and stereoisomeric position similar to that of human milk: effects on fat and
mineral balance." Am. J. Clin. Nutr. 61(5): 1037-1042.
Carnielli, V. P., I. H. Luijendijk, J. B. Van Goudoever, E. J. Sulkers, A. A. Boerlage, H. J.
Degenhart and P. J. Sauer (1996). "Structural position and amount of palmitic acid in
infant formulas: effects on fat, fatty acid, and mineral balance." J. Pediatr Gastroenterol.
Nutr. 23(5): 553-560.
Dethlefsen, L., P. B. Eckburg, E. M. Bik and D. A. Relman (2006). "Assembly of the human
intestinal microbiota." Trends Ecol. Evol. 21(9): 517-523.
Dharmani, P., V. Srivastava, V. Kissoon-Singh and K. Chadee (2009). "Role of Intestinal
Mucins in Innate Host Defense Mechanisms against Pathogens." J. Innate Immun. 1(2):
123-135.
Eliakim, A., T. Dolfin, E. Weiss, R. Shainkin-Kestenbaum, M. Lis and D. Nemet (2002).
"The effects of exercise on body weight and circulating leptin in premature infants." J.
Perinatol. 22(7): 550-554.
Evanoo, G. (2007). "Infant crying: a clinical conundrum." J. Pediatr Health Care 21(5): 333338.
Fanaro, S., R. Chierici, P. Guerrini and V. Vigi (2003). "Intestinal microflora in early infancy:
composition and development." Acta Paediatr Suppl. 91(441): 48-55.
Fremy, E. (1842). "Memoire sur les produits de la saponification de lhuile de palme."
Journal de Pharmacie et de Chimie XII: 757.
Giovannini, M., E. Riva and C. Agostoni (1995). "Fatty acids in pediatric nutrition." Pediatr
Clin. North Am. 42(4): 861-877.

Palmitic Acid in Infant Nutrition

155

Guarner, F. and J. R. Malagelada (2003). "Gut flora in health and disease." Lancet 361(9356):
512-519.
Harmsen, H. J. M., A. C. M. Wildeboer-Veloo, G. C. Raangs, A. A. Wagendorp, N. Klijn, J.
G. Bindels and G. W. Welling (2000). "Analysis of Intestinal Flora Development in
Breast-Fed and Formula-Fed Infants by Using Molecular Identification and Detection
Methods." Journal of Pediatric Gastroenterology and Nutrition 30(1): 61-67.
Hecht, G. (1999). "Innate mechanisms of epithelial host defense: spotlight on intestine." Am.
J. Physiol. 277(3 Pt 1): C351-358.
Hedin, C., K. Whelan and J. O. Lindsay (2007). "Evidence for the use of probiotics and
prebiotics in inflammatory bowel disease: a review of clinical trials." Proc. Nutr. Soc.
66(3): 307-315.
Hollingsworth, M. A. and B. J. Swanson (2004). "Mucins in cancer: protection and control of
the cell surface." Nat. Rev. Cancer 4(1): 45-60.
Huang, Q., S. Abdulrahman, J. Yin and C. Zwieb (2002). "Systematic site-directed
mutagenesis of human protein SRP54: interactions with signal recognition particle RNA
and modes of signal peptide recognition." Biochemistry 41(38): 11362-11371.
Innis, S. M. (2011). "Dietary triacylglycerol structure and its role in infant nutrition." Adv
Nutr 2(3): 275-283.
Innis, S. M. (2013). "Maternal Nutrition, Genetics, and Human Milk Lipids." Current
Nutrition Reports: 1-8.
Innis, S. M., R. Dyer and C. M. Nelson (1994). "Evidence that palmitic acid is absorbed as
sn2 monoacylglycerol from human milk by breast-fed infants." Lipids 29(8): 541-545.
Innis, S. M. and C. M. Nelson (2013). "Dietary triacyglycerols rich in sn2 palmitate alter
post-prandial lipoprotein and unesterified fatty acids in term infants." Prostaglandins
Leukot Essent Fatty Acids 89(4): 145-151.
Janz, K. (2002). "Physical activity and bone development during childhood and adolescence.
Implications for the prevention of osteoporosis." Minerva Pediatr 54(2): 93-104.
Jensen, C., N. R. Buist and T. Wilson (1986). "Absorption of individual fatty acids from long
chain or medium chain triglycerides in very small infants." Am. J. Clin. Nutr. 43(5): 745751.
Jensen, R. G. (1995). "Comments on the extraction of fat from human milk for analysis of
contaminants." Chemosphere 31(9): 4197-4200; author reply 4200-4195.
Jensen, R. G. (1999). "Lipids in human milk." LIPIDS 34(12): 1243-1271.
Jensen, R. G., M. M. Hagerty and K. E. McMahon (1978). "Lipids of human milk and infant
formulas: a review." Am. J. Clin. Nutr. 31(6): 990-1016.
Johansson, M. E., D. Ambort, T. Pelaseyed, A. Schutte, J. K. Gustafsson, A. Ermund, D. B.
Subramani, J. M. Holmen-Larsson, K. A. Thomsson, J. H. Bergstrom, S. van der Post, A.
M. Rodriguez-Pineiro, H. Sjovall, M. Backstrom and G. C. Hansson (2011).
"Composition and functional role of the mucus layers in the intestine." Cell Mol. Life Sci.
68(22): 3635-3641.
Kau, A. L., P. P. Ahern, N. W. Griffin, A. L. Goodman and J. I. Gordon "Human nutrition,
the gut microbiome and the immune system." Nature 474(7351): 327-336.
Kau, A. L., P. P. Ahern, N. W. Griffin, A. L. Goodman and J. I. Gordon (2011). "Human
nutrition, the gut microbiome and the immune system." Nature 474(7351): 327-336.
Kennedy, K., M. S. Fewtrell, R. Morley, R. Abbott, P. T. Quinlan, J. C. Wells, J. G. Bindels
and A. Lucas (1999). "Double-blind, randomized trial of a synthetic triacylglycerol in

156

Fabiana Bar-Yoseph, Yael Lifshitz, Tzafra Cohen et al.

formula-fed term infants: effects on stool biochemistry, stool characteristics, and bone
mineralization." Am. J. Clin. Nutr. 70(5): 920-927.
Liao, X., W. Zhang, J. He, J. Sun and P. Huang (2005). "Bone measurements of infants in the
first 3 months of life by quantitative ultrasound: the influence of gestational age, season,
and postnatal age." Pediatr Radiol. 35: 847-853.
Litmanovitz, I., F. Bar-Yoseph, Y. Lifshitz, K. Davidson, A. Eliakim, R. Regev and D. Nemet
(2014). "Reduced Crying in Term Infants Fed High Beta-Palmitate Formula: A DoubleBlind, Randomized, Clinical Trial." BMC Pediatr 14(1): 152.
Litmanovitz, I., K. Davidson, A. Eliakim, R. H. Regev, T. Dolfin, S. Arnon, F. Bar-Yoseph,
A. Goren, Y. Lifshitz and D. Nemet (2013). "High Beta-palmitate formula and bone
strength in term infants: a randomized, double-blind, controlled trial." Calcif Tissue Int.
92(1): 35-41.
Litmanovitz, I., T. Dolfin, O. Friedland, S. Arnon, R. Regev, R. Shainkin-Kestenbaum, M.
Lis and A. Eliakim (2003). "Early physical activity intervention prevents decrease of
bone strength in very low birth weight infants." Pediatrics 112(1 Pt 1): 15-19.
Lodinova, R., V. Jouja and A. Lanc (1967). "Influence of the intestinal flora on the
development of immune reactions in infants." J. Bacteriol. 93(3): 797-800.
Lopez-Lopez, A., A. I. Castellote-Bargallo, C. Campoy-Folgoso, M. Rivero-Urgel, R. TormoCarnice, D. Infante-Pina and M. C. Lopez-Sabater (2001). "The influence of dietary
palmitic acid triacylglyceride position on the fatty acid, calcium and magnesium contents
of at term newborn faeces." Early Hum. Dev. 65 Suppl: S83-94.
Lu, P., F. Bar-Yoseph, L. Levi, Y. Lifshitz, J. Witte-Bouma, A. C. de Bruijn, A. M.
Korteland-van Male, J. B. van Goudoever and I. B. Renes (2013). "High beta-palmitate
fat controls the intestinal inflammatory response and limits intestinal damage in mucin
Muc2 deficient mice." PLoS One 8(6): e65878.
Lu, P., N. Burger-van Paassen, M. van der Sluis, J. Witte-Bouma, J. P. Kerckaert, J. B. van
Goudoever, I. Van Seuningen and I. B. Renes (2011). "Colonic gene expression patterns
of mucin muc2 knockout mice reveal various phases in colitis development." Inflamm
Bowel Dis.
Lucas, A., P. Quinlan, S. Abrams, S. Ryan, S. Meah and P. J. Lucas (1997). "Randomised
controlled trial of a synthetic triglyceride milk formula for preterm infants." Arch. Dis.
Child Fetal Neonatal Ed 77(3): F178-184.
Monteleone, G., R. Caruso and F. Pallone (2014). "Targets for new immunomodulation
strategies in inflammatory bowel disease." Autoimmun. Rev. 13(1): 11-14.
Moore, T. A., C. K. Hanson and A. Anderson-Berry "Colonization of the Gastrointestinal
Tract in Neonates." ICAN: Infant, Child, & Adolescent Nutrition 3(5): 291-295.
Moore, T. A., C. K. Hanson and A. Anderson-Berry (2011). "Colonization of the
Gastrointestinal Tract in Neonates." ICAN: Infant, Child, & Adolescent Nutrition 3(5):
291-295.
Morelli, L. (2008). "Postnatal development of intestinal microflora as influenced by infant
nutrition." J. Nutr. 138(9): 1791S-1795S.
Mu, H. and C. E. Hoy (2004). "The digestion of dietary triacylglycerols." Prog. Lipid Res.
43(2): 105-133.
Nelson, C. M. and S. M. Innis (1999). "Plasma lipoprotein fatty acids are altered by the
positional distribution of fatty acids in infant formula triacylglycerols and human milk."
Am. J. Clin. Nutr. 70(1): 62-69.

Palmitic Acid in Infant Nutrition

157

Neu, J. and W. A. Walker (2011). "Necrotizing enterocolitis." N. Engl. J. Med. 364(3): 255264.
Neville, M. C. and M. F. Picciano (1997). "Regulation of milk lipid secretion and
composition." Annu. Rev. Nutr. 17: 159-183.
Niers, L., M. Stasse-Wolthuis, F. M. Rombouts and G. T. Rijkers (2007). "Nutritional support
for the infant's immune system." Nutr. Rev. 65(8 Pt 1): 347-360.
O'Hara, A. M. and F. Shanahan (2006). "The gut flora as a forgotten organ." EMBO Rep 7(7):
688-693.
Pai, R. and G. Kang (2008). "Microbes in the gut: a digestable account of host-symbiont
interactions." Indian J. Med. Res. 128(5): 587-594.
Quinlan, P. T., S. Lockton, J. Irwin and A. L. Lucas (1995). "The relationship between stool
hardness and stool composition in breast- and formula-fed infants." J. Pediatr
Gastroenterol. Nutr. 20(1): 81-90.
Rasquin, A., C. Di Lorenzo, D. Forbes, E. Guiraldes, J. S. Hyams, A. Staiano and L. S.
Walker (2006). "Childhood functional gastrointestinal disorders: child/adolescent."
GASTROENTEROLOGY 130(5): 1527-1537.
Rauch, F. and E. Schoenau (2001). "Changes in bone density during childhood and
adolescence: an approach based on bone's biological organization." J. Bone Miner Res
16(4): 597-604.
Sakaguchi, S. (2005). "Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in
immunological tolerance to self and non-self." Nat. Immunol. 6(4): 345-352.
Scrimgeour, C. M. and J. L. Harwood (2007). The Lipid Handbook, Taylor & Francis Group,
LLC.
Sidnell, A. and E. Greenstreet "Infant nutrition review of lipid innovation in infant
formula." Nutrition Bulletin 36(3): 373-380.
Specker, B. (2004). "Nutrition influences bone development from infancy through toddler
years." J. Nutr. 134(3): 691S-695S.
St James-Roberts, I. and T. Halil (1991). "Infant crying patterns in the first year: normal
community and clinical findings." J. Child Psychol. Psychiatry 32(6): 951-968.
St James-Roberts, I., J. Hurry and J. Bowyer (1993). "Objective confirmation of crying
durations in infants referred for excessive crying." Arch. Dis. Child 68(1): 82-84.
Teitelbaum, J. E. and W. Allan Walker (2001). "Review: the role of omega 3 fatty acids in
intestinal inflammation." J. Nutr. Biochem. 12(1): 21-32.
Tourneur, E. and C. Chassin (2013). "Neonatal immune adaptation of the gut and its role
during infections." Clin. Dev. Immunol. 2013: 270301.
Van der Sluis, M., B. A. De Koning, A. C. De Bruijn, A. Velcich, J. P. Meijerink, J. B. Van
Goudoever, H. A. Buller, J. Dekker, I. Van Seuningen, I. B. Renes and A. W. Einerhand
(2006). "Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is
critical for colonic protection." Gastroenterology 131(1): 117-129.
Velcich, A., L. Palumbo, L. Selleri, G. Evans and L. Augenlicht (1997). "Organization and
regulatory aspects of the human intestinal mucin gene (MUC2) locus." J. Biol. Chem.
272(12): 7968-7976.
Velcich, A., W. Yang, J. Heyer, A. Fragale, C. Nicholas, S. Viani, R. Kucherlapati, M.
Lipkin, K. Yang and L. Augenlicht (2002). "Colorectal cancer in mice genetically
deficient in the mucin Muc2." Science 295(5560): 1726-1729.

158

Fabiana Bar-Yoseph, Yael Lifshitz, Tzafra Cohen et al.

Yao, M., E. L. Lien, M. R. Capeding, M. Fitzgerald, K. Ramanujam, R. Yuhas, R.


Northington, J. Lebumfacil, L. Wang and P. A. DeRusso (2014). "Effects of Term Infant
Formulas Containing High sn2 Palmitate with and Without Oligofructose on Stool
Composition, Stool Characteristics, and Bifidogenicity: A Randomized, Double-Blind,
Controlled Trial." J. Pediatr. Gastroenterol. Nutr.
Yaron, S., D. Shachar, L. Abramas, A. Riskin, D. Bader, I. Litmanovitz, F. Bar-Yoseph, T.
Cohen, L. Levi, Y. Lifshitz, R. Shamir and R. Shaoul (2013). "Effect of high betapalmitate content in infant formula on the intestinal microbiota of term infants." J.
Pediatr. Gastroenterol. Nutr. 56(4): 376-381.
Yoshioka, H., K. Iseki and K. Fujita (1983). "Development and differences of intestinal flora
in the neonatal period in breast-fed and bottle-fed infants." Pediatrics 72(3): 317-321.

In: Palmitic Acid: Occurrence, Biochemistry and Health Effects ISBN: 978-1-63321-519-1
Editor: Lucas F. Porto
2014 Nova Science Publishers, Inc.

Chapter 8

PROCESSING OF PALMITIC ACID AND ITS


DERIVATIVES USING SUPERCRITICAL FLUIDS
C. E. Schwarz
Department of Process Engineering, Stellenbosch University,
Private Bag X1, Matieland, South Africa

ABSTRACT
Palmitic acid, either in its triglyceride form or hydrolysed as a free fatty acid or an
ester, needs to be extracted from its source, processed and isolated to obtain useful
products. The objective of this work is to consider the use of SCF (supercritical fluid)
processing as a method to extract and process palmitic acid and/or its derivatives. A
phase behaviour analysis, in supercritical CO2, ethane and propane, at temperatures close
to the critical point of the solvent show moderate solubility of palmitic acid and
tripalmitin at pressures below 50 MPa and total solubility of methyl and ethyl palmitate at
pressures below 25 MPa. Analysis of the phase behaviour considered and studies
presented in the literature have shown that SCFs can be widely applied to the processing
of palmitic acid containing compounds. In particular SCFs can fractionate a mixture of
acids or their derivatives according to the chain length, it can de-acidify an edible oil and
it is able to fractionate a mixture containing palmitic acid and other compounds.
Additionally, SCFs can also be used to extract palmitic acid containing triglycerides from
plant material. SCFs, in particular CO2, are thus excellent alternative solvent to
traditional organic solvents and should be considered when processing palmitic acid
containing products.

SYMBOLS AND ABBREVIATIONS


Cn
DHA
EPA

Compound with a chain length of n carbon atoms


Docosahexaenoic acid
Eicosapentaenoic acid

Tel: +27 21 8084487, Fax: +27 21 8082059, Email: cschwarz@sun.ac.za

160

C. E. Schwarz
FFA
SC
SCF
SCFE
SCFF
SVE
Tr
VLE
wi

Free fatty acids


Supercritical
Supercritical fluid
Supercritical fluid extraction
Supercritical fluid fractionation
Solid-vapour equilibrium
Reduced temperature
Vapour-liquid equilibrium
Mass fraction of component i in a mixture

INTRODUCTION
Palmitic acid is a major constituent of many naturally derived oils and extracts with
applications in, amongst others, the cosmetic, pharmaceutical and surfactant industries.
Palmitic acid is a linear saturated carboxylic acid with 16 carbon atoms, hence its IUPAC
name, hexadecanoic acid.
As its common name indicates, palmitic acid is a major constituent of the oil from palm
trees. However, palmitic acid does not only occur in the products of palm trees but occurs
widely in naturally derived oils and extracts from both plant and animal sources. Plant
material sources include soybean oil [1], cocoa butter [2], palm kernel oil [3], wheat bran oil
[4], pumpkin seed oil [5], to name but a few. Additionally by-products from oil production
also contain palmitic acid, for example soybean oil and palm oil deodoriser distillates [6,7].
Fish oils also contain a significant amount of palmitic acid, as shown in the review project of
Gruger et al. [8].
Table 1. Physical properties of palmitic acid and its derivatives
Component
Palmitic acid
Methyl palmitate
Ethyl palmitate
Tripalmitin

Melting point
336 to 338 K [9]
303 to 304 K [9]
297 to 298 K [9]
337 to 338 K [10]

Boiling point
544 K [9]
469 K at 2 kPa [9]
464 K at 1.33 kPa [9]
(Not readily available)

Molecular mass
256.42 g/mol
270.45 g/mol
284.48 g/mol
807.32 g/mol

In nature, palmitic and other high molecular mass acids generally do not occur as free
fatty acids (FFAs) but rather primarily as triglycerides. In general, these triglycerides are
usually mixed, in other words the triglycerides contains different acids e.g. one palmitic acid
and two oleic acid groups. While extraction from the source material results in triglycerides,
should palmitic acid be isolated or concentrated, the triglycerides need to be hydrolysed to
acids or esters. Therefore, palmitic acid and its derivatives, while present in many sources,
also occur in combination with many other components. In order to obtain pure palmitic acid
(or its derivative), or even a product concentrated in palmitic acid, some type of separation
process is required.
As palmitic acid may occur as a triglyceride, or may be hydrolysed or esterified, this
study will thus concern itself with not only the triglyceride form but also the FFA (palmitic
acid) and the methyl and ethyl esters (methyl and ethyl palmitate) form of palmitic acid. The

Processing of Palmitic Acid

161

molecular mass and melting and boiling temperatures (where available) of palmitic acid and
its derivatives are presented in Table 1.
The physical properties of these compounds indicate that while they are liquids or solids
near their melting temperature at room temperature, they have very high boiling points, even
at vacuum. Additionally, their high molecular mass and hydrocarbon backbone nature results
in significant thermal degradation at elevated temperatures, in particular above 420 K. These
properties, together with the surfactant nature of these compounds points to difficulties in
separating these compounds from others using traditional separation methods, such as
distillation and liquid-liquid extraction.
Supercritical fluid (SCF) processing is an attractive alternative separation method that
can readily be used to separate high molecular mass thermally sensitive compounds. This
separation method is analogous to separation with a liquid organic solvent except instead of
the use of an organic solvent a high pressure gas near its critical point is used. Previously SCF
processing has been used to fractionate waxes according to their chain length [1113],
separate detergent range alkanes and alcohols [14,15], and extract components from plant and
animals sources [16], to name but a few.
The aim of this chapter is to investigate the supercritical fluid extraction (SCFE) and
supercritical fluid (SCFF) of solid matrices and liquid mixtures containing palmitic acid and
its derivatives (tripalmitin, methyl palmitate and ethyl palmitate). SC (supercritical) CO2,
ethane and propane will be considered as SC solvents with ethane, propane and other
compounds as possible co-solvents to CO2.
In particular, this chapter will focus on:
(1) The phase behaviour of palmitic acid and its derivatives in SCFs and
(2) Using the phase behaviour information, how SCFF and SCFE can be used to obtain a
product enriched in palmitic acid and/or its derivatives.
This chapter will start with a brief overview of SCF processing. Thereafter, a phase
behaviour analysis of palmitic acid, methyl palmitate, ethyl palmitate and tripalmitin in CO2,
ethane and propane, as well as CO2 together with a co-solvent will be considered. The
analysis will investigate phase equilibrium data present in the literature, trends observed the
data, a comparison between the phase behaviour of the various derivatives as well as the
various solvents, and the effect of a co-solvent. The phase behaviour data will then be used to
analyse how SCFF and SCFE can be used to obtain extracts enriched in palmitic acids or its
derivatives. In particular this study will consider how SCFF can be used to distinguish
between various fatty acids and their derivatives and also between palmitic acid and/or its
derivatives and other components present in the sources. Finally, SCFE of palmitic acid
containing triglycerides from solid matrices will be considered. The overarching aim of this
chapter is thus to show how SCFs can be used to process palmitic acid and its derivatives.

SUPERCRITICAL FLUID PROCESSING


A SCF is a component that is above both its critical temperature and pressure. Therefore,
an isothermal change in pressure or an isobaric change in temperature will not lead to a phase

162

C. E. Schwarz

change. In general, SCF processes are operated close to the critical temperature and
preferably the critical pressure of the solvent. At these conditions a SCF is neither a liquid nor
a gas but has physical and transport properties are intermediate of that of a liquid and a gas.
Additionally, in the vicinity of the critical point a small change in pressure and/or temperature
results in a large change in density, and thus solubility of components in these solvents.
Therefore, SCFs have excellent mass transfer properties and are often used as extraction or
fractionation media. Processing using SCFs are analogous to using a liquid organic solvent
with the exception that instead of a liquid solvent a high pressure gas is used as the mass
transfer medium. CO2 is the most popular SCF but low molecular mass alkanes are also
sometimes used.
While this separation method does require high pressure equipment and is generally not
the first method of choice to achieve a separation, it does hold a number of advantages.
Typically a SCF process operates a few degrees above the critical temperature of the solvent
(See Table 2) and as such thermal degradation of temperature sensitive compounds is
avoided. The nature of the solvents are such that they are generally non-toxic and in some
cases also non-flammable. In fact, the most popular SC solvent, CO2, is classified as generally
regarded as safe (GRAS). Additionally, at atmospheric temperatures and pressures these
solvents generally have a very low solubility in high molecular mass solutes resulting in a
negligible solvent residue. Lastly, due to the pressure-volume-temperature behaviour of the
solvent in the operating range, solvent-solute separation can easily be achieved through a
change in pressure and/or temperature. A complicated solvent removal process is thus not
generally required.
SCF separation processes can be divided into two main groups namely SCFF of a liquid
feed and SCFE of compounds from a solid matrix. Basic schematic representations of these
two processes are shown in Figure 1 and Figure 2 for SCFF and SCFE, respectively.
Table 2. Critical properties of selected SC solvents [17]
SC solvent
Carbon dioxide
Methane
Ethane
Propane
n-Butane
Water

Critical Temperature
304.1 K
190.4 K
305.1 K
369.8 K
425.3 K
647.1 K

Critical pressure
7.38 MPa
4.60 MPa
4.88 MPa
4.24 MPa
3.80 MPa
22.06 MPa

As pointed out by Peter and Brunner [18], SCF processing can extract and separate
compounds containing low/non-volatile and heat sensitive substances at temperatures far
lower than that of vacuum distillation. Both SCFF and SCFE are techniques that can be
applied to sources containing palmitic acid. For example, palm kernel oil can be extracted
from the kernels using SCFE and then fractionated by SCFF. In order to design such
processes one requires knowledge of the solubility of palmitic acid (or its derivate) as well as
the other components present in the SC solvent. A difference in solubility between
compounds is a necessary requirement for separation to occur.

Processing of Palmitic Acid

163

Figure 1. Schematic representation of SCFF of a liquid feed.

Figure 2. Schematic representation of SCFE from a solid matrix.

Carbon dioxide (CO2) is by far the most studied SCFs. This is mainly due to its suitable
critical temperature, its abundance and its non-toxicity. The popularity of CO2 can be seen in
number of studies considering the phase behaviour of compounds in CO2 [1922] and
fractionation and extraction studies using SC carbon dioxide [23]. However, CO2 is not
always a good solvent for high molecular mass compounds. In particular, acids and
triglycerides often require very high pressures for significant solubility in SC CO2 [24,25].

164

C. E. Schwarz

Although methyl and ethyl esters of acids have a significantly higher solubility in SC CO2
and total solubility can be attained [26], relatively high pressures (approximately 20 MPa) are
still required.
Low molecular mass alkanes (in particular ethane and propane) are also popular SCFs
and have shown improved solubility of acids in SC CO2 [27,28] with total solubility below
30 MPa at moderate temperatures. Additionally, for methyl and ethyl esters ethane and
propane also show a marked increase in solubility [29,30]. These alkanes can be used either
as solvents on their own or may be used as a co-solvent together with CO2. The critical
temperature of ethane is very close to that of CO2, hence its suitability at an alternative
solvent to CO2. On the other hand, propane has a significantly higher critical temperature
(369.8 K). However, this temperature is quite close to the melting points of palmitic acid and
tripalmitin and, as pointed out by Coorens et al. [31], may therefore be a suitable SC solvent
to process palmitic acid and its derivative. Additionally, propane in general has a higher
solubility of high molecular mass compounds compared to ethane [30], thus requiring lower
pressures and/or lower solvent to feed ratios.
In addition, besides propane and ethane, a number of studies have considered other low
molecular weight organic compounds such as ethanol and n-octane as possible co-solvents for
SCF processes. Ideally, the addition of the co-solvent generally decreases the operating
pressure significantly while hopefully not influencing the selectivity significantly.
This study will thus consider the SCFF and SCFE of sources containing palmitic acid or
its derivatives using predominantly CO2 as extraction medium. However, ethane and propane
as possible alternative solvents will also be considered, as well as the use of co-solvents.

ANALYSIS OF PHASE BEHAVIOUR OF PALMITIC ACID


AND ITS DERIVATIVES IN SCFS
The first step in designing any separation or purification process using an auxiliary
medium is to consider the phase behaviour, in this medium, of the components present in the
liquid to be fractionated or the solid from where it is to be extracted. The phase behaviour will
provide an indication of the operating conditions and a difference in phase behaviour is a prerequisite for separation to occur.
SCF processing is no different. In fact, a phase behaviour analysis is vital as it provides
an estimation of the operating pressures required and also indicates whether separation from
other components will be possible. This section will focus on the phase behaviour of palmitic
acid, methyl palmitate, ethyl palmitate and tripalmitin in SC CO2, ethane and propane and
concentrate on the data available and trends observed therein. In addition, the three solvents
will be compared and the effect of co-solvents, often used to decrease the operating pressure,
will be considered. In particular this section will focus on the phase transition pressures of the
systems studied. The phase transition pressure indicates the pressure required for total
solubility at the said temperature and composition. For the type of systems studied here, a
higher phase transition pressure leads to a lower solubility, therefore lower phase transition
pressures indicate improved solubility.

Processing of Palmitic Acid

165

Phase Behaviour Palmitic Acid and Its Derivatives in SC CO2


CO2 is the most popular SC solvent and has a critical temperature of 304.1 K [17]. As
most SC processes operate just above the critical point of the solvent, this study will focus on
the temperature range 308 to 373 K, yet studies beyond this range will also be mentioned.

Published Phase Equilibrium Data


CO2 is the most popular SC solvent and, as expected, a significant amount of data is
available for the palmitic acid and its derivatives in SC CO2.

CO2/Palmitic Acid
The systems CO2/palmitic acid has been extensive studied. Outlined in Table 3 is a
summary of the available phase equilibrium data.
Palmitic acid has a normal melting point of 336 to 338 K [9]. However, in the presence of
a SC CO2 palmitic acid undergoes a melting point depression. Bertakis et al. [46] studied the
vapour-liquid-solid phase equilibria of, amongst others, the CO2/palmitic acid system. Their
results showed that the melting point decreased significantly as the pressure increased to a
minimum of approximately 319.5 K at about 10.6 MPa, after which the melting temperature
increased slightly as the pressure increased. This information is significant at it indicates
which of the data listed in Table 3 is of vapour-solid nature and which data is of vapour-liquid
nature. As most of the data is above 10 MPa, and the melting point does not significantly
change at pressure above 10 MPa, it is assumed that all data above 320 K is of vapour-liquid
in nature, the remainder being of vapour-solid in nature.
Figure 3 shows the solubility pressure of solid palmitic acid in CO2 as a function
composition at 308.5 to 318.15 K. The data of Brunetti et al. [35] does not agree with that of
the other authors and was therefore omitted from Figure 3. Maheshwari et al. [42] also
questioned the data of Brunetti et al. [35] as they found that the data of Brunetti et al. was
consistently higher than their data as well as that of Ohgaki et al. [43]. They ascribed the
difference due to the presence of impurities in the materials. They suggested that impurities
may act as entrainers and could therefore either increase or decrease the solubility of the
molecules in CO2, depending on the impurities present.
Table 3. Literature data for the CO2 (1)/palmitic acid (2) system
Reference

Purity

Temperature
range

Pressure range

Composition range
(w2) a

Bamberger et al. [32]

(1) 99.92 %
(2) 99-100 %

313 K

8.0 to 24.8 MPa

2.86E-6 to 5.77E-3 (9)

Bharath et al. [33]

(1) 99.9 %
(2) 90 %

353.15 and
373.15 K

13.60 to
30.52 MPa

Vapour: 0.000582 to
0.0155 (5)
Liquid: 0.580 to 0.810
(7)

Brandt et al. [34]

(1) 99.995 %
(2) 99 %

313 and 318 K

10.14 to
23.35 MPa

8.361E-4 to 6.44E-3
(11)

166

C. E. Schwarz
Table 3. (Continued)

Reference

Purity

Temperature
range

Pressure range

Composition range
(w2) a

Brunetti et al. [35]

(2) 96 % min.

308.15 to
323.15 K

20 and 30 MPa

0.00902 to 0.0403 (6)

Garlapati and
Madras [36]

(1) 99.9 %
(2) 98 %

308 and 318 K

12.8 to 22.6
MPa

1.50E-3 to 6.06E-3
(10)

Garlapati and
Madras [37]

(1) 99.9 %
(2) 98 %

328 K

12.8 to 22.6
MPa

2.16E-3 to 1.23E-3 (5)

308.15 K to
328.15 K

10.0 to 35.0
MPa

3.79E-3 to 9.13E-3
(18)

Gordillo et al. [38]

Iwai et al. [39]

(1) 99.9 %
(2) 99 %

308.15 K

9.9 to 23.0 MPa

8.91E-4 to 2.89E-4 (5)

Iwai et al. [40]

(1) 99.9 %
(2) 99 %

313.2 K

15.0 MPa

3.24E-3 (1)

Kramer and Thodos


[41]

(1) 99.8 %
(2) 95 %

318 to 338 K

14.21 to
57.48 MPa

0.00302 to 0.270 (19)

Maheshwari et al.
[42]

(1) 99.9 %
(2) 99 %

308 to 328 K

13.8 to
41.4 MPa

0.0019 to 0.071 (9)

Ohgaki et al. [43]

(1) 99.99 %
(2) 99 %

298.15 and
313.15 K

7.89 to 18.7
MPa

4.08E-4 to 3.34E-3
(22)

Penedo et al. [44]

(1) 99.99 %
(2) 99.99 %

313.15 to
353.15 K

6 to 12 MPa

Vapour: 0.0087 to
0.0345 (9)
Liquid: 0.2107 to
0.3372 (9)

Schwarz and
Knoetze [24]

(1) 99.95 %
(2) 99 %

337.1 to 358.2
K

10.53 to
25.93 MPa

Vapour: 0.0142 to
0.0224 (23)
Liquid: 0.585 to 0.756
(18)

Yau et al. [45]

(1) 99.5 %
(2) 98 %

373.15 to
473.15 K

1.01 to 5.07
MPa

Vapour: 0.0000 to
0.0031 (15)
Liquid: 0.931 to 0.993
(15)

Value in brackets indicates the number of data points published

As seen, a reasonable solubility (greater than 0.2 mass %) can be attained at moderate
pressures (less than 20 MPa), indicating that SC CO2 is able to extract palmitic acid from a
solid matrix. The data also shows, as pointed out by Bamberger et al. [32], that an increase in
temperature at constant pressure leads to an increase in solubility, and, as stated by Garlapati
and Madras [37], that an increase in pressure at constant temperature also leads to an increase
in solubility.
The VLE phase behaviour of the CO2/palmitic acid system using the data of Schwarz and
Knoetze [24] is shown in Figure 4. Schwarz and Knoetze found their data for
CO2/hexadecanoic acid to be in agreement with that of Kramer and Thodos [41] and Bharath
et al. [33]. However, the data of Penedo et al. [44] appeared to be at too low pressures, most
probably due to the fact that they used an extraction type set-up instead of a phase equilibrium

Processing of Palmitic Acid

167

type set-up. Penedo et al. [44] claim that their results agree well with that of Chen et al. [47]
for the system CO2/linoleic acid and thus regarded their results as verified. However,
inspection of the graphs showing the comparison points towards a significant difference. For
all further analyses the data of Schwarz and Knoetze will be used as these authors have
regularly verified their method, they use a synthetic method which is highly accurate for
binary phase equilibrium data, and the palmitic acid used by Schwarz and Knoetze is of high
purity.
40
35
Pressure (MPa)

30
318.15 K

25

313.15 K

20

308.15 K

15
10
5
0
0.000

0.002

0.004

w2

0.006

0.008

Figure 3. Pressure composition (w2) plot for the CO2 (1)/palmitic acid (2) system at 308.15 to 318.15
K [34,36,3843].

32
28
Pressure (MPa)

24
20

358.15 K

16

348.15 K

12

338.15 K

8
4
0
0.0
Figure 4. (Continued).

0.2

0.4

(a) w2

0.6

0.8

1.0

168

C. E. Schwarz
32
28

Pressure (MPa)

24
20

358.15 K

16

348.15 K

12

338.15 K

8
4
0
0.012

0.015

0.018

0.021

0.024

(b) w2
Figure 4. Pressure composition (w2) plot for the CO2 (1)/palmitic acid (2) system at 338.15 to 358.15
K [24] (a) entire composition range and (b) detail of low molecular mass composition range.

Schwarz and Knoetze [24] found that for their VLE data an approximately linear
relationship exists between temperature and the phase transition pressure at constant
composition. This relationship has a positive gradient and indicates a higher solubility at
lower temperatures, converse to that of the solid-vapour equilibrium (SVE) phase behaviour.
This positive gradient was also found through the entire mass fraction range studied and the
authors did not find any indications of temperature inversions in this system.
Comparing Figure 3 and Figure 4(b) it is clear that the solubility of liquid phase palmitic
acid in SC CO2 is of an order of magnitude higher than that of the solid phase palmitic acid
(compare the x-axes). Thus, SCFE of palmitic acid as a solid from a matrix will, in all
likelihood, occur at much higher pressures than SCFF processes involving palmitic acid.

CO2/Methyl Palmitate
Outlined in Table 4 is a summary of data for the system CO2/methyl palmitate. Due to the
low melting point of the data (See Table 1), all data is of a vapour-liquid equilibrium (VLE)
nature.
Figure 5 shows the phase equilibrium data of the system CO2/methyl palmitate of both
Inomata et al. [48] and Lockemann [49]. Figure 5 hints towards significant differences
between the data sets, yet insufficient information is available to determine which data set us
superior. However, the data clearly shows that an increase in temperature results in an
increase in phase transition pressure. Due to the scatter in the data and slight inconsistencies
between the two sources the exact nature of the relationship between temperature and the
phase transition pressure can not be determined. Additional measurements would be required
therefore. Both sets of data do, however, indicate that total solubility can be achieved at
moderate pressures (less than 25 MPa at temperatures below 343 K).

Processing of Palmitic Acid

169

Table 4. Literature data for the CO2 (1)/methyl palmitate (2) system
Reference

Purity

Inomata et al.
[48]

Lockemann
[49]
a

(1) 99.9 %
(2) 95 %

Temperatur
e range
313.15 to
343.15 K

Pressure
range
1.01 to
18.28 MPa

(1) 99.95 %
(2) 95 %

313.15 to
333.15 K

0.98 to
13.0 MPa

Composition range
(w2) a
Vapour: 0.00165 to
0.0676 (24)
Liquid: 0.333 to
0.960 (16)
Vapour: 0.0006 to
0.1028 (16)
Liquid: 0.3041 to
0.9614 (38)

Value in brackets indicates the number of data points published

16

Pressure (MPa)

12
343.15 K
8

333.15 K
323.15 K
313.15 K

0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 5. Pressure composition (w2) plot for the CO2 (1)/methyl palmitate (2) system at 313.15 to
343.15 K [48,49].

CO2/Ethyl Palmitate
Table 5 summarises data available for the system CO2/ethyl palmitate and the data is
illustrated in Figure 6. Liang and Yeh [50] also studied the solubility of ethyl palmitate in
CO2. However, they presented their data only in figures and as a correlation (Chrastil
correlation). As such their data has not been included in this study. The data of Crampon et al.
[26] and that of Gaschi et al. [51] appear to be in agreement.
The data also indicates, as pointed out by Crampon et al., that an increase in temperature
leads to an increase in phase transition pressure and suggests that a linear relationship
between temperature and pressure exists. Gaschi et al. observed three phase behaviour at
303.15 K but not at 313.15 K and higher temperatures. Three phase regions are interesting
from a thermodynamic point of view yet in practice should be avoided due to the
complications associated therewith. Importantly, as for the CO2/methyl palmitate system,
total solubilities can be attained at moderate pressures (< 25 MPa) in the temperature range
under investigation. This affords lower operating pressures and as such lower capital and
operating costs.

170

C. E. Schwarz

CO2/Tripalmitin
A significant amount of data has been published on the VSE and VLE for the system
CO2/tripalmitin, as outlined in Table 6. To date, no detailed study has been conducted on the
melting point depression of tripalmitin in SC CO2. Due to the similar melting point of
tripalmitin and palmitic acid [9], a cut-off temperature of 320 K similar to that of palmitic
acid in CO2 is assumed. The VSE and VLE of the system CO2/tripalmitin is shown in Figure
7 and Figure 8, respectively.
Table 5. Literature data for the CO2 (1)/ethyl palmitate (2) system
Reference

Purity

Temperatur
e range

Pressure range

Composition
range (w 2) a

Crampon et al.
[26]

(1) 99.9 %
(2) 99 %

313.15 to
333.15 K

1.98 to
15.89 MPa

0.0376 to 0.931
(77)

Gashi et al.
[51]

(1) 99.9 %
(2) 95 %

303.15 to
353.15 K

3.10 to
20.86 MPa

0.05368 to 0.8645
(60)

Value in brackets indicates the number of data points published

21

Pressure (MPa)

18
15

353.15 K

12

343.15 K
333.15 K

323.15 K

313.15 K
303.15 K

3
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 6. Pressure composition (w2) plot for the CO2 (1)/ethyl palmitate (2) system at 303.15 to
353.15 K [26,51].

As for the VSE of the CO2/palmitic acid system, the VSE of the CO2/tripalmitin system
shows that an increase in temperature leads to an increase in solubility. However, there
appears to be significant discrepancies between the various sources, as seen for the 313.15 K
data. Bamberger et al. [32] found that the data of Chrastil [52] is an order of magnitude
higher than theirs and speculate that it may be due to purity. Bamberger et al. conducted
additional tests (data not given) using a lower purity tripalmitin (90%) and found that the
solubilities were much higher. The purity of the tripalmitin used by Chrastil was not given.

Processing of Palmitic Acid

171

It may therefore be that the discrepancies between the data sets are due to the purity of the
components used.
Table 6. Literature data for the CO2 (1)/tripalmitin (2) system
Reference

Purity

Bamberger
et al. [32]
Bharath et
al. [33]

(1) 99.92 %
(2) 99 %
(1) 99.9 %
(2) 90 %

Chrastil
[52]
Mnkl et
al. [53]
Nilsson and
Hudson
[54]
Ohgaki et
al. [43]
Weber et
al. [25]

(1) 99.98 %

313 to 353 K

(1) 99.995 %
(2) 99 %
Not given

336 to 363 K

(1) 99.99 %
(2) 96.5 %
(1) 99.5 %
(2) 95 %

298.15 to
313.15 K
333 and 353
K

Temperature
range
313 K
353.15 K

313.15 and
333.15 K

Pressure
range
12.2 to 29.7
MPa
5.55 to
24.31 MPa

8.11 to
25.33 MPa
3.24 to
24.75 MPa
17.2 to 31.0
MPa
8.60 to 18.2
MPa
10 to 50 MPa

Composition
range (w 2)a
5.56E-5 to 4.97E-4
(7)
Vapour: 0.00183
to 0.0286 (5)
Liquid: 0.561 to
0.787 (5)
1.02E-4 to 1.96E-3
(18)
0.700 to 0.950 (20)
2.8E-4 to 1.08E-4
(9)
8.36E-5 to 1.42E-4
(16)
Vapour: 0.00018
to 0.03340 (9)
Liquid: 0.5873 to
0.9183 (9)

Value in brackets indicates the number of data points published

40
35
Pressure (MPa)

30
313.15 K

25
20

298.15 K

15
10
5
0
0.00E+00

5.00E-04

w2

1.00E-03

1.50E-03

Figure 7. Pressure composition (w2) plot for the CO2 (1)/tripalmitin (2) system at 292.15 and 313.15
K [32,43,52,54].

172

C. E. Schwarz
50
45

Pressure (MPa)

40
35
30

363.15 K

25

353.15 K

20

343.15 K

15
10
5
0
0.0

0.2

0.4

(a) w2

0.6

0.8

1.0

50
45
40
Pressure (MPa)

35
30
25

353.15 K

20
15
10
5
0
0.000

0.006

0.012

(b) w2

0.018

0.024

Figure 8. Pressure composition (w2) plot for the CO2 (1)/tripalmitin (2) system at 343.15 to 363.15 K
[25,33,52,53] (a) entire composition range and (b) detail of low molecular mass composition range.

Despite a number of studies considering the VLE of the CO2/tripalmitin, significant


discrepancies exist between the data sets and no conclusion can be made on which set of data
is superior. However, the data of Mnkl et al. [53] do indicate that once again in the VLE
region an increase in temperature leads to an increase in phase transition pressure.
Additionally, irrespective of which source represents the phase behaviour most accurately, all
four sources show that high pressures in excess of 30 MPa are required for total solubility.
Comparing the SVE and the VLE the same trends as for the CO2/palmitic acid system are
noted. Once again significantly higher solubilities are observed for liquid tripalmitin in CO2
compared to solid tripalmitin in CO2.

Processing of Palmitic Acid

173

Analysis of Phase Equilibrium Data


All four systems behave in a generally similar manner. For VLE an increase in
temperature leads to an increase in phase transition pressure, resulting in a decrease in
solubility. For the VSE the CO2/palmitic acid system showed the opposite temperature effect
while insufficient data are available to comment on the temperature dependence of solid
tripalmitin in SC CO2. However, both the CO2/palmitic acid and CO2/tripalmitin systems
showed that the solid solubility is approximately an order of magnitude less than that of the
liquid.
Figure 9 and Figure 10 compare the phase behaviour of palmitic acid and its derivatives
in SC CO2 at 313.15 and 353.15 K, respectively.
32

Pressure (MPa)

28
24

Tripalmitin

20

Palmitic acid

16

Methyl
palmitate

12
8
4
0
0.0

0.2

0.4

(a) w2

0.6

0.8

1.0

32
28
Pressure (MPa)

24
20
Tripalmitin

16

Palmitic acid

12
8
4
0
0.000

0.002

(b) w2

0.004

0.006

Figure 9. Comparison of the pressure composition (w2) for the systems CO2 (1)/palmitic acid (2)
[34,43], CO2 (1)/methyl palmitate (2) [48,49], CO2 (1)/ethyl palmitate (2) [26,51] and CO2
(1)/tripalmitin (2) [32,43,54] systems at 313.15 K (a) entire composition range and (b) detail of low
molecular mass composition range.

174

C. E. Schwarz

The data shows that at both temperatures the esters have the highest solubility followed
by palmitic acid and that tripalmitin has the lowest solubility. It is also noted that the systems
CO2/methyl palmitate and CO2/ethyl palmitate have very similar phase behaviour and similar
phase transition pressures. This is expected due to the similarity of the molecules. However,
despite the fact that Bharath et al. [55] found that for C18 and higher esters the methyl ester
has a higher phase transition pressure than the ethyl ester, Figure 9 indicates that the phase
transition pressures are indeed very similar and from the data available in the present analysis
no outcome can be given in this regard.
32
28
Pressure (MPa)

24
20
Tripalmitin

16

Palmitic acid

12
8
4
0
0.0

0.2

0.4

0.6

0.8

1.0

(a) w2
32
28
Pressure (MPa)

24
20
Tripalmitin

16

Palmitic acid

12
8
4
0
0.000

0.005

0.010

0.015

0.020

0.025

(b) w2
Figure 10. Comparison of the pressure composition (w2) for the CO2 (1)/palmitic acid (2) [24], CO2
(1)/ethyl palmitate (2) [51]and CO2 (1)/tripalmitin (2) [25,33,52,53] systems at 353.15 K (a) entire
composition range and (b) detail of low molecular mass composition range.

Processing of Palmitic Acid

175

Importantly, however, is the fact that the solubility of methyl palmitate and ethyl
palmitate in SC CO2 is significantly higher than that of palmitic acid and tripalmitin.
Significantly lower pressures would therefore be required for processes involving methyl
palmitate or ethyl palmitate compared to those involving palmitic acid or tripalmitin.

Phase Behaviour Palmitic Acid and Its Derivatives in SC Ethane


The phase behaviour of palmitic acid and its derivatives with SC ethane is discussed
below. The same temperature range as for CO2 will be considered as ethane has a very similar
critical temperature.

Published Phase Equilibrium Data


While phase equilibrium data involving CO2 is rather abundant, very little data is
available for systems involving ethane. In fact, only the research group of Schwarz and coworkers has studied these systems.
Ethane/Palmitic Acid
Details pertaining to the ethane/palmitic acid system measurements are provided in Table
7 and the phase transition pressure, as a function of composition at various temperatures, can
be seen in Figure 11.
Schwarz and Chobanov [27] found total solubility for the system ethane/palmitic acid at
moderate pressures (< 25 MPa). They also showed that an increase in temperature leads to an
increase in phase transition pressure at constant composition, with this increase being linear in
the temperature range studied.
Table 7. Literature data for the ethane (1)/palmitic acid (2) system
Reference

Purity

Schwarz and
Chobanov [27]

(1) 99.5 %

Temperature
range
332.4 to 354.4 K

Pressure
range
9.17 to
21.20 MPa

Composition
range (w 2) a
0.0164 to 0.671

(2) 99 %
Value in brackets indicates the number of data points published

Ethane/Methyl Palmitate
As for the ethane/palmitic acid system, only a single source contains data for the
ethane/methyl palmitate system. The data is outlined in Table 8 and the phase behaviour for
the system ethane/methyl palmitate is shown in Figure 12.
Schwarz et al. [29] found that total solubility can be attained for the system
ethane/methyl palmitate at low pressures (< 15 MPa) and that, as for the ethane/palmitic acid
system, a linear relationship exists between the temperature and the phase transition pressure
at constant composition in the temperature range studied.

176

C. E. Schwarz
24

Pressure (MPa)

20
16
353.15 K
12

343.15 K
333.15 K

8
4
0
0.0

0.1

0.2

0.3

w2

0.4

0.5

0.6

0.7

Figure 11. Pressure composition (w2) plot for the ethane (1)/palmitic acid (2) system at 333, 343 and
353 K [27].

Table 8. Literature data for the ethane (1)/methyl palmitate (2) system
Reference

Purity

Temperatu
re range

Pressure range

Composition range
(w2) a

Schwarz et
al. [29]

(1) 99.95 %
(2) 99 %

311.5 to
365.5 K

5.21 to 13.8
MPa

0.0172 to 0.630 (48)

Value in brackets indicates the number of data points published

15

Pressure (MPa)

12
9

353.15 K
343.15 K

333.15 K
323.15 K
313.15 K

3
0
0.0

0.1

0.2

0.3

w2

0.4

0.5

0.6

0.7

Figure 12. Pressure composition (w2) plot for the ethane (1) /methyl palmitate (2) system at 313 to
353 K [29].

Processing of Palmitic Acid

177

Ethane/Ethyl Palmitate
Once again, only a single data source is available for the ethane/ethyl palmitate system is
available. The data is summarised in Table 9 and the phase behaviour of the ethane/ethyl
palmitate data is presented in Figure 13.
The data for the ethane/ethyl palmititate system is very similar to that of the
ethane/methyl palmitate system and the same comments thus apply.
Table 9. Literature data for the ethane (1)/ethyl palmitate (2) system
Reference

Purity

Temperature
range
312.7 to 357.3 K

Pressure range

Schwarz et
(1) 99.5 %
5.38 to 13.52
al. [30]
(2) 99 %
MPa
a
Value in brackets indicates the number of data points published

Composition
range (w 2) a
0.0155 to
0.593 (48)

15

Pressure (MPa)

12
9

353.15 K
343.15 K

333.15 K
323.15 K
313.15 K

3
0
0.0

0.1

0.2

0.3

w2

0.4

0.5

0.6

0.7

Figure 13. Pressure composition (w2) plot for the ethane (1)/ethyl palmitate (2) system at 333, 343
and 353 K [30].

Ethane/Tripalmitin
No data is available for the system ethane/tripalmitin. In order to estimate the phase
behaviour of the system ethane/tripalmitin one can approximate the data to that of an alkane
with the same molecular mass. Peters previously suggested this analogy for the
propane/tripalmitin system [56]. The oxygen atoms are located centrally in the molecule and
thus well shielded. Tripalmitin has 57 carbon and oxygen atoms in the molecular backbone
and as such the ethane/tripalmitin system may be approximated by the ethane/nheptapentacosane system as a first order approximation. While the VLE for the ethane/nheptapentacosane system has not been measured, Schwarz et al. [57] published correlations
that link the phase transition pressure with the number of carbon atoms. These correlations
are used and the approximated phase behaviour of the system ethane/tripalmitin is presented
in Figure 14. It should be noted that these correlations are based on data generated with the

178

C. E. Schwarz

solute in the liquid phase and are thus only valid above the melting point of the solute.
Additionally, data for ethane/n-heptapentacosane system would be an extrapolation and while
extrapolation may be possible [58] the data generated would therefore be more of an
indication of the phase transition pressure rather than an accurate representation. Figure 14
has been generated at 340 and 360 K, and while n-heptapentacosane may be a solid at this
temperature, tripalmitin will be in the liquid state, hence its validity.
As expected, an increase in temperature leads to an increase in phase transition pressure.
Importantly, total solubility can be attained, albeit at quite high pressures (approximately 35
MPa at 360 K).
35

Pressure (MPa)

28
21
360 K
340 K

14
7
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 14. Approximation of the ethane (1)/tripalmitin (2) pressure composition (w2) plot at 340 and
360 K based on the ethane/n-alkane correlations for 57 carbon atoms, as proposed by Schwarz et al.
[57].

Analysis of Phase Equilibrium Data


While less sources are available for the phase behaviour of palmitic acid and its
derivatives in ethane compared to CO2 as solvent, sufficient data is available to discuss the
trends present. Figure 15 compares the phase behaviour of palmitic acid, methyl palmitate,
ethyl palmitate and tripalmitin in SC ethane at 360 K. Similar comparisons result at other
temperatures.
In general similar phase behaviour trends were observed between the systems.
Tripalmitin is the least soluble in ethane followed by palmitic acid. Methyl palmitate and
ethyl palmitate show high solubility in ethane and show total miscibility at pressures below
15 MPa in the temperature range studied. As observed by Schwarz et al. [30] very little
difference exists between the phase behaviour of ethane/methyl palmitate and ethane/ethyl
palmitate systems, most probably due to the similarity in the nature of their structure.

Processing of Palmitic Acid

179

35
Tripalmitin

30

Palmitic acid

Pressure (MPa)

25

Methyl
palmitate

20
15
10
5
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 15.Comparison of the pressure composition (w2) for the systems ethane (1)/palmitic acid (2)
[27], ethane (1)/methyl palmitate (2) [29], ethane (1)/ethyl palmitate (2) [30] and ethane (1)/tripalmitin
(2) (generated as described above using the ethane/n-alkane correlation of Schwarz et al. [57]) systems
at 360 K.

Phase Behaviour Palmitic Acid and Its Derivatives in Supercritical Propane


Propane is regarded as an alternative SC solvent to or possible co-solvent with CO2,
especially for components with slightly higher melting points. Propane has a critical
temperature of 369.8 K [17] and as such SC processes with propane as SC solvent typically
operate between 373 and 408 K. This temperature range will also be the temperature range of
focus for propane as SC solvent in this study.

Published Phase Equilibrium Data


As for the phase behaviour of palmitic acid and its derivatives with ethane, not very many
studies for the phase behaviour of palmitic acid and its derivatives in propane have been
published. However, all these studies were from the research groups of Schwarz and coworkers and Peters and co-workers, and both these research groups are regarded to publish
reliable data. Sufficient data therefore exists to conduct the required analysis.
Propane/Palmitic Acid
A single source has published data for the system propane/palmitic acid. The data is
summarised in Table 10 and trends in the phase behaviour for the propane/palmitic acid
system are shown in Figure 16.
The data indicates that total solubility can be attained at low pressures (< 10 MPa) even at
high temperatures. In agreement with Figure 16, Schwarz et al. [28] found a linear
relationship between temperature and the phase transition pressure with this linear
relationship having a positive gradient. No temperature inversions were observed. Peters [56]
graphically published UCEP and LCEP data for acids in propane. This information indicates
that a three phase region is possible for the propane/palmitic acid system. Interpolation of

180

C. E. Schwarz

their graphs indicates that the three phase region occurs between approximately 370 and 374
K and therefore the observation of Schwarz et al. that no three phase regions are present
between 378 and 408 K is in agreement with the work of Peters [56].
Table 10. Literature data for the propane (1)/palmitic acid (2) system
Reference

Purity

Temperature
range

Pressure
range

Composition range
(w2) a

Schwarz et
al. [28]

(1) 99.95 %
(2) 99 %

376.4 to 409.5
K

4.18 to 9.03
MPa

0.0184 to 0.655

Value in brackets indicates the number of data points published

10

Pressure (MPa)

8
6

408.15 K
393.15 K

378.15 K

2
0
0.0

0.1

0.2

0.3

w2

0.4

0.5

0.6

0.7

Figure 16. Pressure composition (w2) plot for the propane (1)/palmitic acid (2) system at 378 to 408 K
[28].

Propane/Methyl Palmitate
Rovetto et al. [59] published phase equilibrium data for the system propane/methyl
palmitate. The data is summarised in Table 11 and Figure 17 shows the phase behaviour.
Figure 17 indicates that total solubility can be attained at very low pressures (< 8 MPa).
The data also shows that as temperature increases, the phase transition pressure increases. In
fact, the data shown in Figure 17 was obtained by fitting a linear relationship between the
temperature and phase transition pressure at constant composition on the data published by
Rovetto et al. [59]. Data between approximately 370 and 410 K was used and a fit with a
Pearsons R2 value greater than 0.997 was attained in all cases for a linear relationship
between the temperature and the phase transition pressure.

Processing of Palmitic Acid

181

Table 11. Literature data for the propane (1)/methyl palmitate (2) system
Reference

Purity

Temperature
range

Pressure
range

Composition range
(w2) a

Rovetto et
al. [59]

(1) 99.95 %
(2) 99 %

312.11 to
450.63 K

1.23 to 9.87
MPa

0.1798 to 0.7647
(84)

Value in brackets indicates the number of data points published

10

Pressure (MPa)

8
6

408.15 K
393.15 K

378.15 K

2
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 17. Pressure composition (w2) plot for the propane (1)/methyl palmitate (2) system at 378 to
408 K [59].

Propane/Ethyl Palmitate
Together with their study on the phase behaviour of the ethane/ethyl ester homologous
series, Schwarz et al. published data on the propane/ethyl ester homologous series. This is the
only known source of data for the propane ethyl/palmitate system, is summarised in Table 12
and graphically illustrated in Figure 18.
As for the propane/methyl palmitate system, total solubility can be attained at very low
pressures (< 8 MPa) for the propane/ethyl palmitate system. Similar to the observed linear
relationship between temperature and the phase transition pressure, Schwarz et al. [30] also
found a linear relationship between the phase transition pressure and temperature. They also
did not observe any three phase behaviour or indications thereof.
Table 12. Literature data for the propane (1)/ethyl palmitate (2) system
Reference

Purity

Temperature
range
377.2 to 409.6 K

Pressure
Composition range
range
(w2) a
Schwarz et
(1) 99.95 %
3.52 to 7.36 0.0201 to 0.642 (36)
al. [30]
(2) 99 %
MPa
a
Value in brackets indicates the number of data points published

182

C. E. Schwarz
8

Pressure (MPa)

4
408.15 K
2

393.15 K
378.15 K

0
0.0

0.1

0.2

0.3

w2

0.4

0.5

0.6

0.7

Figure 18. Pressure composition (w2) plot for the propane (1)/ethyl palmitate (2) system at 378 to 408
[30].

Propane/Tripalmitin
Coorens et al. [31] conducted a detailed study on the phase behaviour of the
propane/tripalmitin system. Their data is summarised in Table 13 and illustrated in Figure 19
for SVE and in Figure 20 for VLE phase behaviour.
The VSE of the propane/tripalmitin system shows that the solubility is very much
temperature and pressure dependent and that at constant pressure, significantly higher
solubility can be attained at higher temperatures. Similar observations were made for the
CO2/tripalmitin system.
The VLE phase behaviour of the propane/tripalmitin system is similar to that of the other
compounds in propane. Total solubility can be attained at moderate pressure (< 12 MPa).
Table 13. Literature data for the propane (1)/tripalmitin (2) system
Reference
Coorens et al. [31]

Purity

Temperatur
e range
317.59 to
413.77 K

Pressure
range
0.351 to
12.731 MPa

(1) 99.95 %
(2) GC
standard
Coorens et al. [31] (1) 99.95 %
314.36 to
1.051 to
(2) GC
339.37 K
13.051 MPa
standard
a
Value in brackets indicates the number of data points published

Composition
range (w 2) a
0.1258 to
0.9752 (139)
0.1258 to
0.9752 (51)

Processing of Palmitic Acid

183

15

Pressure (MPa)

12
0.975
9

0.861
0.717

0.491
0.303

0.126

0
310

315

320

325

330

335

340

Temperature (K)
Figure 19. Pressure temperature plot for the propane (1)/tripalmitin (2) system at various tripalmitin
compositions (w2) K [30].

12

Pressure (MPa)

9
408.15 K
6

393.15 K
378.15 K

0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 20. Pressure composition (w2) plot for the propane (1)/tripalmitin (2) system at 378, 393 and
408 K [30].

As for the propane/methyl palmitate data, the propane/tripalmitin data presented was
generated using a linear relationship fitted to the experimentally published data of Coorens et
al. [31]. Once again an excellent fit was obtained (R2 > 0.997 in all cases), therefore
illustrating the linear relationship between temperature and the phase transition pressure.
Coorens et al. [31] also published vapour-liquid-liquid equilibrium data showing that the
system has a three phase region between 349 and 370 K. However, compositions were not
included.

184

C. E. Schwarz

Comparing Figure 19 and Figure 20, it can be seen that the phase behaviour for the VSE
and VLE is significantly different. As for the CO2/palmitic acid and CO2/tripalmitin systems,
the VSE data shows an increase in solubility with temperature while the converse is true for
the VLE data. Additionally, the solubility of the solid in propane is again much lower than
that of the liquid in propane. Thus, while the absolute values of the pressure differ (a detailed
analysis is presented below) the same trends as for the CO2/tripalmitin system are present.

Analysis of Phase Equilibrium Data


Many of the comments made for the phase behaviour of palmitic acid and its derivatives
in CO2 and in ethane are also valid for propane as SC solvent. Figure 21 shows a comparison
of the propane/tripalmitin, propane/palmitic acid, propane/methyl palmitate and propane/ethyl
palmitate systems.
10

Pressure (MPa)

Tripalmitin
Palmitic acid

Methyl
palmitate

4
2
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 21. Comparison of the pressure composition (w2) for the systems propane (1)/palmitic acid (2)
[28], propane (1)/methyl palmitate (2) [59], propane (1)/ethyl palmitate (2) [30] and propane
(1)/tripalmitin (2) [31] systems at 393 K.

As for CO2 and ethane as SC solvents, the same trends are observed for propane as SC
solvent. Methyl palmitate and ethyl palmitate behave very similarly and are the most soluble,
followed by palmitic acid. Tripalmitin is the least soluble in propane, yet even for tripalmitin
only moderate pressures are required for total solubility.
Despite the high temperatures, relatively low pressures are required for total solubility,
therefore decreasing the capital cost investment. Propane should thus be regarded as a good
substitute or co-solvent to CO2 to reduce the processing pressures.

Processing of Palmitic Acid

185

Comparison of Supercritical Solvents


A comparison of the phase behaviour of palmitic acid, methyl palmitate, ethyl palmitate
and tripalmitin in SC CO2, ethane and propane is shown in Figure 22, Figure 23, Figure 24
and Figure 25, respectively. With exception of the tripalmitin data, the ethane and CO2 data
are at the same temperature with the propane at a similar reduced solvent temperature.
25

Pressure (MPa)

20
Propane

15

Ethane

10

Carbon
dioxide

5
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 22. Comparison, at similar reduced temperatures (Tr ~ 1.11), of the pressure composition (w2)
phase behaviour of the CO2 (1)/palmitic acid (2) at 338 K [24], ethane (1)/palmitic acid (2) at 338 K
[27] and propane (1)/palmitic acid (2) at 410 K [28] systems.

15

Pressure (MPa)

12
9

Propane
Ethane

Carbon
dioxide

3
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 23. Comparison, at similar reduced temperatures (Tr ~ 1.11), of the pressure composition (w2)
phase behaviour of the CO2 (1)/methyl palmitate (2) at 323 K [48,49], ethane (1)/methyl palmitate (2)
at 323 K [29] and propane (1)/methyl palmitate (2) at 393 K [59] systems.

186

C. E. Schwarz
15

Pressure (MPa)

12
9

Propane
Ethane

Carbon
dioxide

3
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 24. Comparison, at similar reduced temperatures (Tr ~ 1.06), of the pressure composition (w2)
phase behaviour of the CO2 (1)/ethyl palmitate (2) at 323 K [26,51], ethane (1)/ethyl palmitate (2) at
323 K [30] and propane (1)/ethyl palmitate (2) at 393 K [30] systems.

35

Pressure (MPa)

30
25
Ethane

20

Carbon dioxide

15

Propane 436 K

10

Propane 429 K

5
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 25. Comparison, at similar reduced temperatures of the pressure composition (w2) phase
behaviour of the CO2 (1)/tripalmitin (2) at 353 K (Tr = 1.16) [25,33,52,53], ethane (1)/tripalmitin (2) at
360 K (Tr = 1.18) (data generated using the ethane/n-alkane correlations for the system ethane/ntetrapentacontane, as proposed by Schwarz et al. [57]) and propane (1)/tripalmitin (2) at 429 (Tr = 1.16)
and 436 K (Tr = 1.18) [31] systems.

The results show that, as expected from the analysis above, the phase transition pressure
of propane is the lowest and as such its solubility the highest. Additionally, for palmitic acid,
methyl palmitate and ethyl palmitate the phase transition pressures are lower for propane than
for ethane, and CO2 has the highest phase transition pressure and thus the lowest solubility.

Processing of Palmitic Acid

187

For tripalmitin, it should be noted that the CO2 and ethane data are not at the same
temperature not at the same reduced temperature. The CO2 and propane at 429 K data are at
similar reduced solvent temperatures and the ethane and propane at 436 K data are at similar
reduced solvent temperatures. Tripalmitin is thus considerably more soluble in propane than
in ethane or CO2. It should be recalled (See Figure 8) that significant scatter exists for the
CO2/tripalmitin data. Before an outcome regarding a comparison of the solubility of
tripalmitin in CO2 and ethane can be given, issues relating to this scatter need to be resolved.
The accuracy of the ethane/tripalmitin approximation also needs to be verified. In all
likelihood, additional CO2/tripalmitin as well as ethane/tripalmitin data need to be measured.
The low molecular mass alkanes are thus able to dissolve palmitic acid and its derivatives
at lower pressures than CO2. This is in agreement with Mnkl et al. [53] who compared the
solubility of CO2 and in propane in a hardened rape seed oil and found that propane is able to
dissolve in the oil a lot better than the CO2. While ethane and in propane are not toxic, they
are highly flammable and as such care is required if they are used as alternative solvents or
even in high concentrations as co-solvents. However, despite the flammability issues
associated with ethane and propane, these two solvents are non-toxic and excellent
alternatives to CO2. As they are both also SC solvents, the solvent residue would be similar
than when using CO2 as solvent and the solvent recycle systems are of similar complexity to
using CO2.

The Influence of a Co-Solvent on the Phase Behaviour in Supercritical


Solvents
As seen above, at certain operating conditions high pressures (> 25 MPa) are required for
significant solubility of tripalmitin and palmitic acid in CO2. These high pressures are as a
result of CO2 not being a very good solvent for these systems. In order to circumvent the low
solubility of these components in SC CO2, either an alternative solvent to or a co-solvent with
CO2 can be used. Ethane and propane were considered in sections 3.2 and 3.3 above as
alternative solvents or co-solvents. However, in the literature other volatile organic
compounds have also been studied. Table 14 presents a summary of the main studies
involving the phase behaviour of palimitic acid and its derivatives in a SC CO2 in the
presence of a co-solvent.
The majority of the studies involve ethanol as a co-solvent. This may be due to the fact
that it is generally a good co-solvent compared to the other components considered [36,60].
Additionally, as it is often difficult to remove residual co-solvent from the high molecular
mass product, ethanol is more suitable than most other organic solvents as small amounts of
ethanol are not harmful to humans.
The effect of the co-solvent will now be investigated by firstly considering the effect that
the quantity of solvent has on the phase behaviour and secondly comparing some co-solvents,
as shown in Figure 26 to Figure 29.
Figure 26 shows that the inclusion of even 20 % (on a molar basis) of ethanol to the
solvent significantly increases the solubility of palmitic acid in the CO2. Figure 27 shows a
reduction in pressure due to the co-solvent for ethyl palmitate as solute and further indicates
than in increase in the molar ratio of ethanol to CO2 leads to an increase in solubility.
However, Gaschi et al. [51] found that while the addition of ethanol as an entrainer reduced

188

C. E. Schwarz

the phase transition pressure in the region of the critical point (as shown in Figure 27), at low
CO2 compositions this observation is reversed. Similar phase behaviour was observed for the
CO2/biodiesel/ethanol system [62].
Table 14. Literature data phase equilibria of palmitic acid or its derivatives (2) in CO2
(1) in the presence of a co-solvent (3)

Reference

System

Purity

Temperature
range

Pressure
range

Composition
range (w2, w3) a

Brandt et al.
[34]

(2) palmitic
acid
(3) ethanol

(1) 99.995 %
(2) 99 %
(3) 99.8 %

313 K

8.21 to
24.61 MPa

w2 = 0.010 to
0.1271;
w3 = 0.00525 to
0.0948 (11)

Brandt et al.
[34]

(2) palmitic
acid
(3) 2propanol

(1) 99.995 %
(2) 99 %
(3) 99.8 %

313 K

10.90 to
20.69 MPa

w2 = 0.0326 to
0.1272
w3 = 0.0110 to
0.118 (9)

Garlapati
and Madras
[36]

(2) palmitic
acid
(3) ethanol

(2) 98 %
(3) 99.9 %

308 an 318 K

12.8 to 22.6
MPa

w2 = 3.07E-3 to
0.0349;
w3 = 7.57E-3 to
0.0429 (20)

Garlapati
and Madras
[36]

(2) palmitic
acid
(3) 3methyl-1butanol

(2) 98 %
(3) 98 %

308 an 318 K

12.8 to 22.6
MPa

w2 = 2.39E-3 to
0.0145;
w3 = 0.0144 to
0.0387 (15)

Gaschi et al.
[51]

(2) ethyl
palmitate
(3) ethanol

(1) 99.9 %
(2) 95 %
(3) 99.5 %

303.15 to
353.15 K

3.52 to
19.05 MPa

w2 = 0.0530 to
0.700;
w3 = 0.0140 to
0.343 (108)

Iwai et al.
[40]

(2) palmitic
acid
(3) water

(1) 99.9 %
(2) 99 %
(3) Ultrapure

313.2 K

15.0 MPa

w2 = 0.00659
and 0.00568;
w3 = 0.00352
and 0.00391 (2)

Koga et al.
[60]

(2) palmitic
acid
(3) ethanol

(1) 99.9 %
(2) 99 %
(3) 99.5 %

308.2 K

9.9 and
19.7 MPa

w2 = 8.91E-3 to
0.0622;
w3 = 0 to 0.0879
(22)

Koga et al.
[60]

(2) palmitic
acid
(3) octane

(1) 99.9 %
(2) 99 %
(3) 98 %

308.2 K

9.9 and
19.7 MPa

w2 = 8.91E-4 to
0.0331;
w3 = 0 to 0.0184
(22)

Rosso
Comin et al.
[61]

(2) palmitic
acid
(3) ethanol

(1) 99.9 %
(2) 99 %
(3) 99.9 %

313 to 343 K

8.34 to
19.94 MPa

w2 = 0.105 to
0.705;
w3 = 0.0611 to
0.186 (34)

Value in brackets indicates the number of data points published

Processing of Palmitic Acid

189

25

Pressure (bar)

20
With Entrainer
333 K
Without
Entrainer 333K
With Entrainer
343 K
Without
Entrainer 343 K

15
10
5
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 26. Pressure composition (w2) plot for the system CO2 (1)/palmitic acid (2)/ethanol (3) at 323
and 343 K without ethanol [24] and for an ethanol to CO 2 molar ratios of 0.25 [61].

18
15
Pressure (bar)

12

No
entrainer
1:1

1:3

6
3
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 27. Pressure composition (w2) plot for the system CO2 (1)/ethyl palmitate (2)/ethanol (3) at
333.15 K for various ethanol to ethyl palmitate molar ratios [51].

190

C. E. Schwarz
24
0.73 mol %
2Me3BuOH
1.98 mol %
2Me3BuOH

Pressure (bar)

18

0.73 mol %
EtOH

12

1.98 mol %
EtOH
4.16 mo l%
EtOH

No entrainer
0
0.000

0.010

0.020

0.030

w2

0.040

Figure 28. Pressure composition (w2) plot for the system CO2 (1)/palmitic acid (2)/co-solvent (3) at
318 K for 3-methyl-1-butanol and ethanol as co-solvents at a range of co-solvent concentrations [36].

0.07
0.06
0.05
Ethanol, 9.9 MPa

w2

0.04

Ethanol, 19.7
MPa
Octane, 9.9 MPa

0.03
0.02
0.01
0.00
0.00

0.05

0.10

w3

0.15

0.20

Figure 29. Palmitic acid (w2) co-solvent (w3) concentration plot at 308 K and constant pressure for the
system CO2 (1)/palmitic acid (2)/co-solvent(3) for ethanol and octane as co-solvent [60].

Garlapati and Madras [36] and Koga et al. [60] compared various co-solvents and found
the increase in solubility to be more significant using ethanol compared to 3-methyl-1-butanol
and octane, respectively. Iwai et al. [40] considered the effect of water and found that the
solubility of palmitic acid increased with increasing molarity of water, especially near the
saturation point of water. They also found that the solubility of palmitic acid in CO2 saturated
with water was 16 times higher than that of pure CO2. (It should be noted that only 2 data
points are tabulated; the remaining data is all presented in figures.)

Processing of Palmitic Acid

191

Gl-stnda and Temelli [63] reviewed the effect of a co-solvent on the phase
behaviour of lipids in SC CO2. They found that physical interactions between the solutes and
co-solvent, such as dipole dipole, dipole induced dipole or induced dipole induced
dipole (dispersion) interactions and specific interactions such as H-bonding and charge
transfer complexes, are important contributors to the co-solvent effect. The use of a cosolvent may also lead to a change in selectivity. The magnitude of the effect of the co-solvent
is thus a combination of the solvent, the co-solvent, the solute and the operating conditions.
The use of a co-solvent can therefore significantly reduce the phase transition pressure
and thus increase the solubility of palmitic acid and its derivatives in a SC solvent. However,
while this reduction leads to a decrease in operating pressure, it comes at the cost of a more
complicated solvent recycling system, a more complicated control philosophy and increased
solvent residue in the products. The choice of the use of a co-solvent thus needs to take these
aspects, as well as the nature of the co-solvent itself, into account when the use of a cosolvent is evaluated.

SCFF OF MIXED FATTY ACIDS AND THEIR DERIVATIVES


Palmitic acid and its derivatives have, depending on the temperature and pressure,
reasonable solubility in SCFs. In particular, liquid phase components have an acceptable
solubility.
In nature, palmitic acid does not often occur as a pure component. In fact, it is usually
present as a triglyceride but even tripalmitin is seldom encountered. Usually palmitic acid is
present in combination with similar high molecular mass acids in the form of triglycerides.
Hydrolysis of these triglycerides results in a mixture of fatty acids or their esters, which in
turn need to be fractionated to obtain palmitic acid or its ester, usually ethyl palmitate or
methyl palmitate.
This section of the chapter focusses the use of SCFs as a mass transfer agent to
fractionate fatty acids and their esters. The SCF thus behaves in a similar manner to a liquid
organic solvent where it preferentially dissolves one component above another. This section
will start by considering the phase behaviour of various fatty acids in SCFs, followed by
considering typical set-ups and concludes with a summary of the fractionation of plant oils
and fish oils to obtain products of palmitic acid and its derivatives.

Phase Behaviour Analysis


As mentioned above, in order for separation to occur, a difference in phase behaviour is
required. Figure 30 and Figure 31 below consider the phase behaviour of a range of saturated
fatty acids in SC CO2 and ethane, respectively. Propane shows a similar trend to that of
ethane, albeit at lower pressures [28]. Figure 32 considers the phase behaviour of various
tryglycerides in SC CO2. Although no such comparison is possible for ethane or propane due
to a lack of data, similar trends are expected. Figure 33 shows that phase behaviour of a range
of saturated fatty acid ethyl esters in SC CO2. Schwarz and co-workers [29,30] studied the

192

C. E. Schwarz

phase behaviour of methyl and ethyl esters in SC ethane and propane and found similar
results.
30

Pressure (MPa)

25
20

Stearic acid
Palmitic acid

15

Myristic acid
Lauric acid

10

Decanoic acid
5
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 30. Pressure Composition (w2) of various CO2 (1)/linear saturated acid (2) systems at 353 K
[24].

30

Pressure (MPa)

25
Behenic acid

20

Stearic acid
15

Palmitic acid
Myristic acid

10

Lauric acid
Capric acid

5
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 31. Pressure Composition (w2) of various ethane (1)/linear saturated acid (2) systems at 353 K
[27].

Processing of Palmitic Acid

193

30

Pressure (MPa)

25
20
Tristearin
Tripalmitin

15

Trilaurin
10

Tricaprylin

5
0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 32. Pressure Composition (w2) of various CO2 (1)/triglyceride (2) systems at 353 K [25,33].

20

Pressure (MPa)

16
Ethyl
stearate

12
8

Ethyl
palmitate

Ethyl
myristate

0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 33. Pressure Composition (w2) of various CO2 (1)/ethyl ester (2) systems at 333 K [26].

The phase behaviour clearly shows that SCFs are able to distinguish between molecules
based on the number of carbon atoms present. Bharath et al. [33] found, from their phase
equilibria study, that that fatty acid and triglycerides can be fractionated based on their carbon
number using SC CO2. This is in agreement with their previous study [64] that considered the
phase behaviour of palm kernel oil and sesame oil in SC CO2, where they found that SC CO2
is able to selectively dissolve triglycerides based on their carbon number. Numerous other
studies came to similar conclusions [48,6567]. Additionally, Soares et al. [68] compared
the solubility of various fats and oils in SC CO2 and they found, in agreement with the trends
noted above, that oils with more lower molecular mass acids have a higher solubility.

194

C. E. Schwarz

As temperature and pressure affect the solubility, these effects can also be varied to
optimise the difference in solubility. Liang and Yeh [50] studied the separation of ethyl
palmitate, ethyl oleate, eicosapentaenoic acid (EPA) ethyl ester and docosahexaenoic acid
(DHA) ethyl ester. They considered the phase behaviour and used the determined coefficients
for the Chrastil equation to estimate the separation efficiency that can be attained. In general
better separation efficiencies were achieved at higher temperatures and lower pressures. They
verified their qualitative prediction through extraction of esterified fish oil and found the
results correlated well.
While it is believed that solute-solute interactions are not as large as solute-solvent
interactions, there is clear evidence that some type of solute-solute interaction is present in
SCF/high molecular mass systems. Lockemann [49] studied the phase behaviour of the
ternary system CO2/methyl myristate/methyl palmitate and found that these two components
can be separated using SC CO2. However, the separation factor, which dictates the degree or
difficulty of separation, is dependent on the composition of the feed to be separated and the
operating pressure and temperature. They found that while the composition does not
significantly affect the separation factor, better separation can be achieved at lower
composition of the component to be extracted.
In addition to saturated fatty acids with varying chain length, palmitic acid and its
derivatives usually occur in the presence of unsaturated fatty acids and their derivatives, in
particular those with 18 carbon atoms (oleic acid, linoleic acid and linolenic acid). Figure 34
compares the phase behaviour of various methyl esters of C18 acids in SC CO2.
Phase behaviour shown in Figure 34 suggests that it is also possible to achieve separation
according to the degree of saturation, but comparing Figure 33 and Figure 34, it is noted that
the difference in phase behaviour due to unsaturation is less than due to variations in the chain
length. Separation according to unsaturation, while possible, will be more difficult than
according to the chain length. The higher the degree of saturation the lower the phase
transition pressure and thus the higher the solubility. Liong et al. [65] and Nilsson et al. [67]
came to similar conclusions. Normal counter-current fractionation may not be sufficient to
separate compounds only differing in hydrocarbon backbone length and an additional
stationary phase (such as the column in a SCF chromatograph) may be required to achieve
such separations.
Gl-stnda and Temelli [66] as well as Zou et al. [70] studied the ternary system
CO2/oleic acid/linoleic acid. Gl-stnda and Temelli found that depending on the initial
composition, SC CO2 can also distinguish between the two acids. The interactions are,
however, complex and there are indications that solute-solute interactions are present. These
interactions can both enhance and decrease the partition coefficients determined based on the
binary data, depending on the temperature, pressure and acid ratios. Interestingly Zou et al.
found that while the ratio of the components in the liquid phase remains essentially the same
due to the fact that the majority of the components are in the liquid phase, the ratio is the
vapour phase is significantly different, but the difference depends on the molar ratio of the
components.

Processing of Palmitic Acid

195

15

Pressure (MPa)

12
9

Methyl
linoleate

Methyl
oleate

Methyl
stearate

0
0.0

0.2

0.4

w2

0.6

0.8

1.0

Figure 34. Pressure Composition (w2) of various CO2 (1)/C18 ethyl ester (2) systems at 313 K [48,69].

Therefore, provided the correct operating conditions are applied, SC CO2 and other
solvents are able to distinguish between acids of the same chain length but different degrees
of saturation.
The analysis presented above has focused primarity on CO2 as SC solvent, mainly due to
an abundance of information on this solvent and a lack of information on other SC solvents.
However, qualitatively similar trends are expected in other SC solvents and as such it is
expected that these solvents could also achieve the desired separation.

Separation Set-Up
The phase behaviour analysis presented above shows that SC CO2 can be used to
fractionate fatty acids and their derivatives primarily according to their chain length, but also
according to their degree of saturation. Most sources of palmitic acid contain other acids of
both longer and shorter chain length and as such a two-step separation process is required
where, in one step, components that are more soluble (generally those with less than 16
carbon atoms) are removed while, in the other step, components with that are less soluble
(generally those with more than 16 carbon atoms) are removed. Two process options, shown
in Figure 35 and Figure 36 are therefore possible. A decision as to the better process option
would depend on the feed stock composition and the associated technical and economic
analysis.

196

C. E. Schwarz

Figure 35. Possible set-up for fractionation of acids or esters according to their hydrocarbon backbone
where the light fraction is removed in the first column and heavy fraction removed in the second
column.

Figure 36. Possible set-up for fractionation of acids or esters according to their hydrocarbon backbone
where the heavy fraction is removed in the first column and the light fraction removed in the second
column.

Processing of Palmitic Acid

197

In the setup shown in Figure 35 the light fraction (i.e. components with less than 16
carbon atoms) is first removed and the raffinate from the first column is fed to the second
column where the C16 component is extracted and the raffinate ideally contains components
with 18 and more carbon atoms. By and large this option would be preferred as the solvent
usage would be less.
However, there may be occasions where it is more viable to first remove the heavy
components (C18 and greater) after which the extract is fractionated into the light components
(C14 and less) and the resultant product (C16). Such a typical process is shown in Figure 36.
It should be noted that these setups were compiled where only palmitic acid or its
derivative is the only desired product. However, in many cases palmitic acid occurs in
combination with other valuable fatty acids (e.g. oleic acid, linoleic acid, stearic acid and
even DHA and EPA) and as such more complicated separation sequences are likely.

SCFF of Palmitic Acid and Its Derivatives from Plant Extracts


A large number of plants extracts, especially from the seeds, contain palmitic acid and/or
its derivatives. These sources may include soybean oil [1], cocoa butter [2], palm kernel oil
[3], wheat bran oil [4], pumpkin seed oil [5], to name but a few. By and large the palmitic
acid occurs as triglycerides, in most cases in mixed triglycerides where the other acids present
range from C12 to generally C18 but these may be as high as C22 or even C24. In order to obtain
the palmitic acid, the triglycerides are hydrolysed to either FFA or their methyl or ethyl
esters. Therefore, in order to obtain palmitic acid or its derivatives, the mixtures need to be
fractionated, usually according the chain length.
In addition to hydrolysed triglycerides, palm fatty acid distillates also contains a
significant amount of palmitic acid. Palm fatty acid distillates is a byproduct of the palm oil
refining process and contains predominantly FFA (palmitic, oleic and linoleic, and possibly
some stearic acid)[71]. This byproduct, as well as similar byproducts from other crude oil
refining processes, are thus also potential sources of palmitic acid.
The analysis presented above shows that SCFs is able to achieve this separation.
Fractionation set-ups similar to that presented above can therefore be used to fractionate the
feed material resulting in a product rich in palmitic acid.
An excellent example of the implementation of SCFF to obtain palmitic acid from a plant
source is the work of Brunner and Machado [7,72]. They conducted a detailed analysis on the
fractionation of fatty acids from palm fatty acid distillates (99 % FFA (mainly palmitic, oleic
and linoleic acid) , 0.9 % squalene and 0.1 tocopherol) starting with a phase equilibrium
analysis through to pilot plant studies and experimental verification of the separation. They
postulated, from the phase equilibrium studies, that squalene and palmitic acid would be
preferentially extracted and verified their postulation experimentally. They also considered a
pseudo-binary mixture separation where palmitic acid is to be separated from oleic and
linoleic acid and showed, using separation factors that this is possible. On pilot plant scale
they showed that such a separation is feasible and balanced yield and extract quality. At their
optimum conditions (373 K, 29 MPa, extract to raffinate ratio of 1.2) they obtained an extract
where the palmitic acid content was enriched from 52.5 % in the feed to 74.4 % in the extract
and the oleic and linoleic acid content enriched from 46.3 % in the feed to 59.0 % in the
raffinate. Squalene was also enriched in the extract from 0.6 % in the feed to 1.2 % in the

198

C. E. Schwarz

extract. They thus showed it is possible to separate high molecular mass acids based on the
number of carbon atoms on the hydrocarbon backbone.
Using the same concept, only applying it to triglycerides Asep et al. [2] fractionated
cocoa butter according to the molecular mass of the triglyceride. Their triglycerides contained
mainly palmitic, stearic and oleic acid and triglycerides containing more palmitic acid were
preferentially extracted. Acetone, ethanol and isopropanol were used as co-solvents to
increase the extraction yield. Interestingly, an increase in co-solvent concentration also
showed an increase in palmitic acid selectivity while it did not influence the selectivity of the
other acids significantly. Ethanol was found to be the best polar co-solvent.

Palmitic Acid As a By-Product from Fish Oil Fractionation


Fish oils are also rich in fatty acids. Gruger et al. [8] studied the composition of various
fish oils in terms of their fatty acid content. In general, fish oils contain slightly longer chain
fatty acids than those of plant material origin. The study if Gruger et al. showed that the oils
have between 9.5 and 33.4 % of palmitic acid with palmitic acid being the lowest major
constituent in fish oils. The majority of literature information on the fractionation of fatty
acids from fish oils is centred on obtaining the polyunsaturated fatty acids (or their esters).
However, while the main focus of the fractionation of fish oils is to obtain unsaturated fatty
acids, in particular EPA and DHA [73], fractionation of fish oils can produce palmitic acid as
a by-product.
Staby et al. [74] considered the phase behaviour of fish oil in SC CO2 and determined Kvalues for the various constituents. They found that the K-values depend primarily on the
operating temperature and pressure as well as the hydrocarbon backbone chain length but not
so specifically on the number of double bonds present. This is in agreement with phase
equilibria observations where there is a much larger difference in the phase behaviour
between the acids of differing chain length than acids of the same chain length and differing
degrees of saturation.
Staby and Mollerup [75] conducted a review on the fractionation of fish oils according to
their molecular mass using SC CO2. They proposed a process whereby the fish oil can be
fractionated into 4 fractions. Three extraction columns are used in series and each column
should contain a stripping and an enrichment section. The lightest fraction obtained in their
process contains C10 to C16 acids. One could modify this set-up slightly through the addition
of another column and in such also obtain a C16 fraction, as suggested in Figure 37.
The fish oil could be fractionated into five fractions, of which a low chain length, a
stream rich in palmitic acid, a stream rich in stearic, oleic and linoleic acid (C18 acids) and a
stream rich in EPA (C20 acid) and DHA (C22 and C24 acids) are produced. It should, however,
be noted that although the set-up proposed in Figure 37 is probably the most suitable in
general, there may be cases where the column sequencing would be different, such as the
difference between the two sequences shown in Figure 35 and Figure 36. The optimum
column sequencing would be feed stream and separation sequence specific and as such would
not be discussed further here.
Riha and Brunner [76,77] considered the separation of a pseudo 5 component system of
fatty acid ethyl esters of fish oil origin. The focus of the fractionation process was to separate

Processing of Palmitic Acid

199

low molecular mass esters (C14 to C18) from high molecular mass esters (C18 to C20). The high
molecular mass ethyl ester products were achieved at 95 % recovery and 95 % purity. They
thus showed that practically fatty acid ethyl esters can also be separated according to their
molecular mass using SC fluids.

Figure 37. Possible set-up for obtaining palmitic acid or the ester thereof from fish oil using SCFF
(Modification of process proposed by Staby and Mollerup [75]).

SCFF TO SEPARATE PALMITIC AND OTHER FATTY ACIDS AND


THEIR DERIVATIVES FROM OTHER COMPONENTS
SCFF of oils containing palmitic acid and/or its derivatives is not limited only to studies
involving the fractionation of these oils according to their fatty acid content. In fact, as far
back as 1949 Passino [78] published one of the early works on the processing of oils using
high pressure/liquefied gases. In particular he used high pressure propane to process
Mahendra oil, Sardine oil, Soybean oil, Linseed oil, Cod-liver oil and Tall oil. The oils were
improved by removing the colour molecules and ash, and, where applicable, the oil was then
fractionated. In 1978, Peter and Brunner [18] showed experimentally that SC propane, in the

200

C. E. Schwarz

presence of acetone as an entrainer, is able to separate mono-, di- and triglycerides. Later
Peter and Ender [79] showed that monoglycerides can be separated from a mixture of
glycerides using CO2 with propane as an co-solvent. A product containing 99.5 %
monoglycerides was obtained from a glyceride mixture containing 55 % monoglycerides.
Outlined below is a brief discussion on how SCFs can be used to achieve fractionations
involving palmitic acid where the required separation is not (only) between fatty acids of
differing chain length, but rather obtaining palmitic acid from a mixture of a wide range of
compounds. In particular, this part of the chapter will focus on the reduction of FFA content
of oils and the separation of palmitic acid from tocopherols, sterols and other components.

Reduction in Free Fatty Acid Content of Oils


In general, edible oils are preferred to have a low FFA content. In fact, for olive oils the
International Olive Council has included the maximum FFA content in extra virgin (< 0.8
mass %), virgin (< 2.2 mass %) and ordinary olive oil (< 3.3 mass %). SCFF is an excellent
method to reduce the FFA content of oil and as such to improve the quality of the oil
increasing its retail value. SCFF is highly suitable due to the fact that the SC solvent is able to
preferentially dissolve the FFA above the triglycerides present. Such a process would operate
at low temperatures thus limiting thermal degradation and using SC CO2 would omit the use
of organic solvents and unacceptable solvent residues.

Phase Behaviour Analysis


The FFAs (amongst others, palmitic acid) are to be separated from the oil, which contains
predominantly triglycerides. Figure 10, for CO2 as solvent, and Figure 21, for propane as
solvent, show that palmitic acid and tripalmitin have a significantly different solubility in the
SCFs and that the acid preferentially dissolves compared to the triglyceride. While the plots
in Figure 10 and Figure 21 illustrate the difference for palmitic acid and tripalmitin, similar
difference are noted for other fatty acids and their corresponding triglycerides.
Published Studies
A number of studies have, on pilot plant scale, shown that this separation is indeed
possible. Bondoli et al. [80] considered the upgrading of olive oil where the primary aim was
to remove the FFA from the oil. While they did not consider the individual components they
did show that a product with a significant reduction in FFA can be attained. In the extract
high enrichment ratios for squalene, FFA and monoglycerides were obtained. Similarly List et
al. [81] showed experimentally that the FFA content of soybean oil can be reduced using SC
CO2. Ziegler and Liaw [82] used near and SC CO2 to remove FFA from edible oil. A
synthetic mixture containing refined soybean oil with added oleic, linoleic and palmitic acid
as well as pyrazine and its derivatives was used. The acids were concentrated in the extract
with the highest degree of concentration being that of palmitic acid.
Ooi et al. [83] considered the fractionation of palm oil using SC CO2 to produce an
improved product. Their results show that a raffinate with a reduced FFA, monoglyceride and
diglyceride content is achieved and slight fractionation of the triglycerides are obtained.
Similar results were obtained with the use of ethanol as a co-solvent with the exception that

Processing of Palmitic Acid

201

the process operated at a lower pressure. They did not consider a detailed acid content of the
triglycerides, but did show fractionation is possible by separating them into fractions with 48,
50, 52 and 54 carbon atoms.
While literature studies on the removal of FFAs have focussed almost exclusively on CO2
as SC solvent, the work by Peter and co-workers [18,79] on the fractionation of mono-, diand triglycerides using propane or CO2 with propane as co-solvent, shows that it would also
be possible to remove FFAs from oils using other SC solvents.

Separation of Fatty Acids from Tocopherol, Sterols and Other Components


Palmitic acid and other fatty acids very seldom occur in a mixture that does not contain
other chemical compounds. In fact, components such as squalene, tocopherols and sterols are
often present in combination with the fatty acid. The question now arises as to the presence of
these compounds and their distribution between the extract and raffinate phases.

Phase Behaviour Analysis


Figure 38 shows the phase behaviour of palmitic acid in combination with other
compounds often found together with palmitic acid. Importantly, Figure 38 shows that there
exists a significant difference in the phase behaviour between the various compounds (or the
groups of molecules that they represent). Separation would this be possible using a two-step
process where the components more soluble than palmitic acid are removed in the one
column and palmitic acid removed from the components that are less soluble in a second
column. Similar set-ups such as those shown in Figure 35 and Figure 36 may be used to
achieve these separations.
30

Pressure (MPa)

25
20

Cholesterol

15

alpha-tocopherol
Palmitic acid

10

Squalene
5
0
0.00

0.01

w2

0.02

0.03

Figure 38. Comparison of the pressure composition (w2) phase behaviour of the CO2 (1)/squalene (2)
[84], CO2 (1)/palmitic acid (2) [24], CO2 (1)/alpha-tocopherol (2) [85] and CO2 (1)/cholesterol (2) [86]
systems at 333 K.

202

C. E. Schwarz

Published Studies
Araujo et al. [6] modelled the SCFF with SC CO2 of Soybean Oil distillates, containing
FFAs, squalene, tocopherols, sterols and triglycerides, using the Peng Robinson equation of
state. Their results show that squalene and the FFAs present themselves in the extract, the
remainder of the product in the raffinate. However, the squalene has a much higher partition
coefficient and therefore separation from the FFAs is possible in a second column. In their
study the FFAs were lumped together and they did not consider palmitic acid alone. However,
the concept should be applicable to palmitic acid, as suggested by Figure 38.
Stoldt and Brunner [87] considered the phase behaviour of various deacidified palm oils
and soybean oil deodoriser distillates. They focused their study in the compounds other than
the acids/triglycerides present and showed that the other components can all be concentrated
either in the extract or the raffinate of a SCF process. However, improved purity palmitic acid
fractions are obtained as by-products in their process, showing that palmitic acid can also be
recovered from these sources.

SCFE OF PALMITIC ACID CONTAINING OILS


FROM SOLID MATRICES
In the above sections, the focus of the applications of SCF processing was to fractionate a
liquid-like stream using SCFs as extraction medium, similar to liquid-liquid extraction.
However, SCFs has another, often larger, application, namely the extraction of oils and other
compounds from a solid matrix. In fact, one of the earliest commercial applications of SCF
processing is the SCFE of caffeine from coffee to produce decaffeinated coffee [88].
Traditionally components are extracted from solid matrices using liquid organic solvents such
as hexane and methylene chloride. However, these organic solvents result in an unacceptable
solvent residue and thermal degradation occurs during solvent removal. SCF extraction,
especially using SC CO2, is an attractive alternative method.

SCFE as a Possible Extraction Medium


Both solid palmitic acid and solid tripalmitin have acceptable, although not high,
solubilities in SC CO2 and therefore it is possible to obtain extracts of these compounds using
SC CO2. A semi-batch setup, as proposed in Figure 2, can thus be used to obtain oil
containing fatty acids, amongst others palmitic acid, from solid matrices.
Salgin and Korkmaz [5] considered the SCFE of pumpkinseed oil. They found that the
oil they obtained did not differ significantly from that obtained using hexane extraction in
terms of fatty acid content. They came to the conclusion that SCFE is a green process and can
be used to recover healthy oil from pumpkin seeds.
Jung et al. [4] conducted extraction with near and SC CO2 of wheat bran and found that
the major fatty acids present in the extracted oil were palmitic, oleic, linoleic and -linolenic
acids. They only considered the yield change with time, not the compositional change.
However, as seen in 6.2 below, partial fractionation of the oil may be possible.

Processing of Palmitic Acid

203

zkal and co-workers [89,90] studied the extraction of palmitic acid containing oil from
apricot kernels. They studied the effect of, amongst others, temperature, pressure and CO2
flow rate on the extraction yield and built a mass transfer model to describe the extraction.
Their results show that irrespective of temperature, pressure or solvent flow rate, at very long
extraction times the same yield is obtained. However, particle size is important as higher
yields at long extraction times were obtained for smaller particles. zkal and co-workers
concentrated their efforts on the extraction yields and, save the optimum point, did not
consider the composition of their extracts. The operating parameters may influence the
sequence in which the components are extracted and would thus warrant a separate study.

Possibility of Partial Fractionation


Although fatty acids occur in nature predominantly in the form of mixed triglycerides,
some degree of fractionation of the triglycerides based on their total mass may be achieved
using SCFE if the extraction conditions are correctly chosen and varied. Additionally, a
prerequisite for fractionation during extraction would also be that the triglycerides present
vary in molecular mass. Fractionation would thus achieved by the fact that the lower
molecular mass triglycerides are more soluble in SC CO2 and are thus preferentially
extracted.
Salgin and Korkmaz [5] found in their study in the SCFE of pumpkinseed oil that fatty
acid profile of the oil obtained did not differ significantly as the process conditions changed.
This may be due to how the triglycerides are combined or could even be attributed to the
experimental conditions. Partial fractionation is thus not always possible.
There are, however, a number of studies that have shown that partial fractionation is
indeed possible. In most of these studies the temperature and pressure have been kept
constant.
Hassan et al. [3] used SC CO2 extraction to obtain fractions of oils, containing lauric
acid, myristic acid, palmitic acid and oleic acid amongst others, from Palm kernels. Their
results show that at the beginning of the extraction lower acids are removed and later higher
acids are removed.
Joki et al. [1] extracted soybean oil from soybeans and analysed the oil for the acid
content. Statistically significant changes in the acid content were observed as the experiment
continued. The palmitic acid content of the earlier fractions were higher than that of the latter
fractions while the reverse was true for the higher acids.
Nik Norulaini et al. [91] considered the extraction of palm kernels with SC CO2 and
analysed the fractions for their acid content. They found that SCFE can be applied in such a
way that the earlier fractions are richer in lower molecular mass acids and the later fractions
are richer in higher molecular mass acids.

Use of Entrainers
As for SCFF entrainers can be used in SCFE to increase the solubility and/or decrease the
operating pressure. SCFE is by and large more concerned with extraction rather than

204

C. E. Schwarz

fractionation and thus the loss of selectivity sometimes encountered with the use of entrainers
is far out-weighed by the increase in solubility and/or decrease in operating pressure. A large
number of studies have been published on the SCFE using a co-solvent with a selection of
these focussing on products that contain palmitic or similar acids (or rather their
triglycerides).
Bimakr et al. [92] considered the extraction of seed oil from winter melon using SC CO2
and ethanol as a co-solvent with a 10:1 CO2 to ethanol mass ratio. While they did not
investigate the effect of the co-solvent per se, they did conclude that their optimised SCFE
process yields similar results to that of traditional solvent extraction using ethanol. Their
optimised the SCFE process did yield a slightly lower saturated fatty acid contents, with
palmitic acid being approximately 40 % less. However, only the yield was optimised and the
only sample for which analysis was presented was that of the optimised condition. The
process was not optimised for a specific component and it may thus be that higher palmitic
acid content, possibly even higher than that of the traditional ethanol extraction, may be
obtained.
Mendes et al. [93] used ethanol as co-solvent (10 mol %) to increase the lipid yield
during the SC extraction of Spirulina. The CO2 modified with ethanol gave lipid yields
comparable with those of traditional organic solvents. Mendes et al. focussed their research
on the extraction of -linoleinic acid but did also analyse the palmitic acid content of their
samples. However, no comments regarding palmitic acid were given and too little information
is presented to provide definitive conclusions.
zkal et al. [89] investigated the effect of parameters on the SCFE of apricot kernels.
Amongst others, they considered the effect of the use of and the amount of ethanol as cosolvent. They found that an increase in the amount of co-solvent lead to an increase in the
fatty acid yield. The fatty acid contents was not presented for all samples, only the maximum
yield sample and a composition similar to that using traditional organic solvent extraction was
obtained.
In another study on the SC CO2 of extraction of apricot kernels zkal et al. [90]
developed a mass transfer model. They used the model to predict, amongst others, the effect
of the co-solvent and found that while the co-solvent initially increases the extraction yield at
a very long times the extraction yield with or without the co-solvent was the same. The effect
of the co-solvent therefore appears to be to reduce the amount of solvent required and thus
also the extraction time rather than the absolute maximum extraction that can be attained.

CONCLUSION
This chapter has shown that SCF processing is a viable method to process products
containing palmitic acids and its derivatives (tripalmitin, ethyl palmitate and methyl
palmitate). This study was conducted by investigating the solubility of these compounds in
SCFs, in particular CO2, ethane and propane, followed by an investigation as to how SCFF
and SCFE can be applied based on the phase behaviour and verified using published studies.
This study concludes that SCF processing is able to (i) extract palmitic acid and/or its
derivatives from a solid matrix, (ii) fractionate a fatty acid mixture or a mixture of its
derivatives mainly according to the chain length and (iii) provided sufficient difference in the

Processing of Palmitic Acid

205

phase behaviour is present, is able to separate palmitic acid from other components. The
fundamental basis for the possibility of the extraction and separation is that palmitic acid and
its derivatives have a reasonable solubility in SC solvent. While the solubility in CO2 may be
low at times, the solubility can be improved through the use of co-solvents, or ethane or
propane may be used as alternative solvents.
This study has shown that on technical level SCF processing of palmitic acid containing
products is a viable method. In particular, a more environmentally friendly and generally
regarded as safe solvent is used resulting in a product with minimal, if any, solvent residue.
This study was conducted by conceptually considering the separation of palmitic acid or
its derivatives. While examples were mentioned, a more global approach was taken to prove
the technical viability. In order to implement SCF processing technology, the separation
required needs to be investigated based on the raw materials. In addition, an economic and
energy analysis would be required to provide a final outcome as to the viability of the process
as a whole.

NOMENCLATURE
Details pertaining to the acids (and their derivatives) studied in this works are presented
in Table 15.
Table 15. Nomenclature and structure of acids mentioned in this work
Common
name

IUPAC ID

Number of
carbon atoms

Degree of unsaturation

Capric acid

Decanoic acid

10

None

Lauric acid

Dodecanoic acid

12

None

Myristic acid

Tetradecanoic acid

14

None

Palmitic acid

Hexadecanoic acid

16

None

Stearic acid

Octadecanoic acid

18

None

Oleic acid

Octadecenoic acid

18

Single double bond in


varying positions

Linoleic acid

Octadecadienoic
acid

18

Two double bonds,


usually in position 9 and
12

Linolenic acid

Octadecatrienoic
acid

18

Three double bonds

EPA

Eicosapentaenoic_a
cid

20

Five double bonds,


usually from position 3
onwards

Behenic acid

Docosanoic acid

22

None

DHA

Docosahexaenoic
acid

22

Six double bonds, usually


from position 3 onwards

206

C. E. Schwarz

REFERENCES
[1]
[2]
[3]
[4]
[5]
[6]
[7]
[8]
[9]
[10]
[11]
[12]
[13]
[14]
[15]
[16]
[17]
[18]
[19]
[20]
[21]
[22]
[23]
[24]
[25]
[26]
[27]
[28]
[29]
[30]

Jokic, S.; Sudar, R.; Svilovic, S.; Vidovic, S.; Bilic, M.; Velic, D.; Jurokovic, V. Czech
J. Food Sci., 2013, 31, 116125.
Asep, E. K.; Jinap, S.; Jahurul, M. H. A.; Zaidul, I. S. M.; Singh, H. Innovative Food
Sci. Emerging Technol., 2013, 20, 152160.
Hassan, M.; Rahman, N. N. A.; Ibrahim, M.; Omar, A. M. Sep. Purif. Technol., 2000,
19, 113120.
Jung, G.-W.; Uddin, M. S.; Kwon, T. K.; Chun, B.-S. Afr. J. Biotechnol., 2010, 9,
77027709.
Salgn, U.; Korkmaz, H. J. Supercrit. Fluids, 2011, 58, 239248.
Araujo, M. E.; Machado, N. T.; Meireles, M. A. A. Ind. Eng. Chem. Res., 2001, 40,
12391243.
Brunner, G.; Machado, N. T. J. Supercrit. Fluids, 2012, 66, 96110.
Gruger, E. H.; Nelson, R. W.; Stansby, M. E. J. Am. Oil. Chem Soc., 1964, 41, 662
667.
Green, D. W.; Perry, R. H. Perrys Chemical Engineers Handbook; 8th Edition.;
McGraw-Hill: New York, NY, U.S.A., 2004.
Hong, J.; Hua, D.; Wang, X.; Wang, H.; Li, J. J. Chem. Eng. Data, 2010, 55, 297302.
Crause, J. C.; Nieuwoudt, I. J. Supercrit. Fluids, 2003, 27, 3954.
Nieuwoudt, I.; Crause, C.; du Rand, M. J. Supercrit. Fluids, 2002, 24, 4755.
Crause, J. C.; Nieuwoudt, I. Ind. Eng. Chem. Res., 2000, 39, 48714876.
Bonthuys, G. J. K.; Schwarz, C. E.; Burger, A. J.; Knoetze, J. H. J. Supercrit. Fluids,
2011, 57, 101111.
Schwarz, C. E.; Bonthuys, G. J. K.; van Schalkwyk, R. F.; Laubscher, D. L.; Burger, A.
J.; Knoetze, J. H. J. Supercrit. Fluids, 2011, 58, 352359.
Temelli, F. J. Supercrit. Fluids, 2009, 47, 583590.
Poling, B. E.; Prausnitz, J. M.; OConnell, J. P. The Properties of Gases and Liquids;
5th Edition.; McGraw-Hill: New York, NY, U.S.A. 2001.
Peter, S.; Brunner, G. Angew. Chem. Int. Ed. Engl., 1978, 17, 746750.
Dohrn, R.; Brunner, G. Fluid Phase Equilib., 1995, 106, 213282.
Christov, M.; Dohrn, R. Fluid Phase Equilib., 2002, 202, 153218.
Dohrn, R.; Peper, S.; Fonseca, J. M. S. Fluid Phase Equilib., 2010, 288, 154.
Fonseca, J. M. S.; Dohrn, R.; Peper, S. Fluid Phase Equilib., 2011, 300, 169.
Brunner, G. J. Supercrit. Fluids, 2009, 47, 574582.
Schwarz, C. E.; Knoetze, J. H. J. Supercrit. Fluids, 2012, 66, 3648.
Weber, W.; Petkov, S.; Brunner, G. Fluid Phase Equilib., 1999, 158160, 695706.
Crampon, C.; Charbit, G.; Neau, E. J. Supercrit. Fluids, 1999, 16, 1120.
Schwarz, C. E.; Chobanov, K. G. J. Supercrit. Fluids, 2014, 87, 4049.
Schwarz, C. E.; Zamudio, M.; Knoetze, J. H. J. Chem. Eng. Data, 2011, 56, 1116
1124.
Schwarz, C. E.; Hahn, M. L.; de Villiers, A. J.; Knoetze, J. H. Fluid Phase Equilib.,
2011, 311, 3644.
Schwarz, C. E.; Schlechter, C.; Knoetze, J. H. J. Supercrit. Fluids, 2013, 79, 1926.

Processing of Palmitic Acid

207

[31] Coorens, H. G. A.; Peters, C. J.; De Swaan Arons, J. Fluid Phase Equilib., 1988, 40,
135151.
[32] Bamberger, T.; Erickson, J. C.; Cooney, C. L.; Kumar, S. K. J. Chem. Eng. Data, 1988,
33, 327333.
[33] Bharath, R.; Yamane, S.; Inomata, H.; Adschiri, T.; Arai, K. Fluid Phase Equilib.,
1993, 83, 183192.
[34] Brandt, L.; Elizalde-Solis, O.; Galicia-Luna, L. A.; Gmehling, J. Fluid Phase Equilib.,
2010, 289, 7279.
[35] Brunetti, L.; Daghetta, A.; Fedell, E.; Kikic, I.; Zanderighi, L. J. Am. Oil Chem. Soc.,
1989, 66, 209217.
[36] Garlapati, C.; Madras, G. J. Chem. Eng. Data, 2008, 53, 29132917.
[37] Garlapati, C.; Madras, G. J. Chem. Thermodynamics, 2010, 42, 193197.
[38] Gordillo, D.; Pereyra, C.; Martnez de la Ossa, E. J. J. Chem. Eng. Data, 2004, 49, 435
438.
[39] Iwai, Y.; Fukuda, T.; Koga, Y.; Arai, Y. J. Chem. Eng. Data, 1991, 36, 430432.
[40] Iwai, Y.; Uno, M.; Nagano, H.; Arai, Y. J. Supercrit. Fluids, 2004, 28, 193200.
[41] Kramer, A.; Thodos, G. J. Chem. Eng. Data, 1988, 33, 230234.
[42] Maheshwari, P.; Nikolov, Z. L.; White, T. M.; Hartel, R. J. Am. Oil. Chem. Soc., 1992,
69, 10691076.
[43] Ohgaki, K.; Tsukahara, I.; Semba, K.; Katayama, T. Int. Chem. Eng., 1989, 29, 302
308.
[44] Penedo, P. L.; Coelho, G. L. V.; Mendes, M. F. Braz. J. Chem. Eng., 2009, 26, 137
147.
[45] Yau, J.-S.; Chiang, Y.-Y.; Shy, D.-S.; Tsai, F.-N. J. Chem. Eng. Jpn., 1992, 25, 544
548.
[46] Bertakis, E.; Lemonis, I.; Katsoufis, S.; Voutsas, E.; Dohrn, R.; Magoulas, K.; Tassios,
D. J. Supercrit. Fluids, 2007, 41, 238245.
[47] Chen, C.-C.; Chang, C. J.; Yang, P. Fluid Phase Equilib., 2000, 175, 107115.
[48] Inomata, H.; Kondo, T.; Hirohama, S.; Arai, K.; Suzuki, Y.; Konno, M. Fluid Phase
Equilib., 1989, 46, 4152.
[49] Lockemann, C. A. Chem. Eng. Process, 1994, 33, 171187.
[50] Liang, J. H.; Yeh, A.-I. J. Am. Oil Chem. Soc., 1991, 68, 687692.
[51] Gaschi, P. S.; Mafra, M. R.; Ndiaye, P. M.; Corazza, M. L. J. Chem. Thermodynamics,
2013, 57, 1421.
[52] Chrastil, J. J. Phys. Chem., 1982, 86, 30163021.
[53] Mnkl, P.; Wubbolts, F.; De Loos, T. W.; Jansens, P. J. J. Supercrit. Fluids, 2006,
39, 15.
[54] Nilsson, W. B.; Hudson, J. K. J. Am. Oil Chem. Soc., 1993, 70, 749754.
[55] Bharath, R.; Inomata, H.; Arai, K.; Shoji, K.; Noguchi, Y. Fluid Phase Equilib., 1989,
50, 315327.
[56] Peters, C. J. In Supercritical Fluids: Fundamentals for Application; Kiran, E.; Levelt
Sengers, J. M. H., Eds.; Kluwer Academic Publishers: Dordrecht, NL, 1994; pp. 117
145.
[57] Schwarz, C. E.; Nieuwoudt, I.; Knoetze, J. H. J. Supercrit. Fluids, 2008, 46, 226232.
[58] Schwarz, C. E.; Nieuwoudt, I.; Knoetze, J. H. J. Supercrit. Fluids, 2007, 41, 327334.
[59] Rovetto, L. J.; Bottini, S. B.; Peters, C. J. J. Supercrit. Fluids, 2004, 31, 111121.

208

C. E. Schwarz

[60] Koga, Y.; Iwai, Y.; Hata, Y.; Yamamoto, M.; Arai, Y. Fluid Phase Equilib., 1996, 125,
115128.
[61] Rosso Comim, S. R.; Franceschi, E.; Borges, G. R.; Corazza, M. L.; Vladimir Oliveira,
J.; Ferreira, S. R. S. J. Chem. Thermodynamics, 2010, 42, 348354.
[62] Arajo, O. A. S.; Silva, F. R.; Ramos, L. P.; Lenzi, M. K.; Ndiaye, P. M.; Corazza, M.
L. J. Chem. Thermodynamics, 2012, 47, 412419.
[63] Gl-stndag, .; Temelli, F. J. Supercrit. Fluids, 2005, 36, 115.
[64] Bharath, R.; Inomata, H.; Adschiri, T.; Arai, K. Fluid Phase Equilib., 1992, 81, 307
320.
[65] Liong, K. K.; Foster, N. R.; Ting, S. S. T. Ind. Eng. Chem. Res., 1992, 31, 400404.
[66] Gl-stnda, .; Temelli, F. J. Supercrit. Fluids, 2006, 38, 275288.
[67] Nilsson, W. B.; Seaborn, G. T.; Hudson, J. K. J. Am. Oil Chem. Soc., 1992, 69, 305
308.
[68] Soares, B. M. C.; Gamarra, F. M. C.; Paviani, L. C.; Gonalves, L. A. G.; Cabral, F. A.
J. Supercrit. Fluids, 2007, 43, 2531.
[69] Chang, C. J.; Lee, M.; Li, B.; Chen, P. Fluid Phase Equilib., 2005, 233, 5665.
[70] Zou, M.; Yu, Z. R.; Kashulines, P.; Rizvi, S. S. H.; Zollweg, J. A. J. Supercrit. Fluids,
1990, 3, 2328.
[71] Ab Gapor Md Lipid Technology, 2010, 22, 1113.
[72] Machado, N. T.; Brunner, G. Cinc. Tecnol. Aliment., 1997, 17, 354360.
[73] Perretti, G.; Motori, A.; Bravi, E.; Favati, F.; Montanari, L.; Fantozzi, P. J. Supercrit.
Fluids, 2007, 40, 349353.
[74] Staby, A.; Forskov, T.; Mollerup, J. Fluid Phase Equilib., 1993, 87, 309340.
[75] Staby, A.; Mollerup, J. Fluid Phase Equilib., 1993, 91, 349386.
[76] Riha, V.; Brunner, G. J. Supercrit. Fluids, 1999, 15, 3350.
[77] Riha, V.; Brunner, G. J. Supercrit. Fluids, 2000, 17, 5564.
[78] Passino, H. J. Ind. Eng. Chem., 1949, 41, 280287.
[79] Peter, S.; Ender, U. Fat Sci. Technol., 1989, 91, 260266.
[80] Bondioli, P.; Mariani, C.; Lanzani, A.; Fedeli, E.; Mossa, A.; Muller, A. J. Am. Oil
Chem. Soc., 1992, 69, 477480.
[81] List, G. R.; King, J. W.; Johnson, J. H.; Warner, K.; Mounts, T. L. J. Am. Oil Chem.
Soc., 1993, 70, 473476.
[82] Ziegler, G.; Liaw, Y.-J. J. Am. Oil Chem. Soc., 1993, 70, 947953.
[83] Ooi, C. K.; Bhaskar, A.; Yener, M. S.; Tuan, D. Q.; Hsu, J.; Rizvi, S. S. H. J. Am. Oil
Chem. Soc., 1996, 73, 233237.
[84] Martinez-Correa, H. A.; Gomes, D. C. A.; Kanehisa, S. L.; Cabral, F. A. J. Food Eng.,
2010, 96, 4350.
[85] Pereira, P. J.; Goncalves, M.; Coto, B.; de Azevedo, E. G.; da Ponte, M. N. Fluid Phase
Equilib., 1993, 91, 133143.
[86] Yun, S. L. J.; Liong, K. K.; Gurdial, G. S.; Foster, N. R. Ind. Eng. Chem. Res., 1991,
30, 24762482.
[87] Stoldt, J.; Brunner, G. Fluid Phase Equilib., 1998, 146, 269295.
[88] Williams, D. F. Chem. Eng. Sci., 1981, 36, 17691788.
[89] zkal, S. G.; Yener, M. E.; Bayndrl, L. LWT-Food Sci. Technol.. 2005, 38, 611616.
[90] zkal, S. G.; Yener, M. E.; Bayndrl, L. J. Supercrit. Fluids, 2005, 35, 119127.

Processing of Palmitic Acid

209

[91] Nik Norulaini, N. .; Md Zaidul, I. .; Anuar, O.; Mohd. Omar, A. Sep. Purif. Technol.,
2004, 35, 5560.
[92] Bimakr, M.; Rahman, R.; Taip, F.; Adzahan, N.; Sarker, M.; Ganjloo, A. Molecules,
2013, 18, 9971014.
[93] Mendes, R. L.; Reis, A. D.; Palavra, A. F. Food Chem., 2006, 99, 5763.

In: Palmitic Acid: Occurrence, Biochemistry and Health Effects ISBN: 978-1-63321-519-1
Editor: Lucas F. Porto
2014 Nova Science Publishers, Inc.

Chapter 9

PALMITIC ACID IN TUNISIAN OLIVE


OIL: UPDATING AND PERSPECTIVE
Ghayth Rigane1,2, and Ridha Ben Salem1,
1

Laboratoire de Chimie Organique-Physique UR11ES74,


Facult des Sciences de Sfax, Dpartement de Chimie,
Sfax, Universit de Sfax, Tunisie
2
Dpartement de Physique-Chimie, Facult Des Sciences
et Techniques de Sidi Bouzid, Sidi Bouzid,
Universit de Kairouan, Tunisie

ABSTRACT
In this review the major saturated fatty acid, palmitic acid, of Virgin Olive Oil
(VOO) was studied. This oil is one of the oldest known vegetable oils and it plays a
fundamental role in human nutrition around the Mediterranean basin. This nature juice is
the only edible oil of great production obtained by physical methods from the fruit Olea
europaea L. Consideration of VOO as a natural functional fat is related to the presence of
palmitic acid. Updating of its levels in Virgin olive oils throughout the Tunisian olive oil
as well as information on expecting levels in other products from olive tree establish our
view point. Studies on levels palmitic acid upon maturity stage in the oil are also
discussed.
Major analytical practices are given in brief. Palmitic acid (C16:0) is the principal
saturated fatty acid in olive oil, responsible for its figeability at low temperature.
Few are the exceptions as palmitic acid content depends heavily on the genetic
factor. Palmitic fatty acids, important for the nutritional properties of an olive oil, showed
a crucial rule in the characterization of olive oils.

Keywords: Palmitic acid, olive oil, saturated fatty acid composition, storage olive oil
Ghayth Rigane: Laboratoire de Chimie Organique-Physique UR11ES74, Facult des Sciences de Sfax,
Dpartement de Chimie, B.P 1171 3038, Sfax, Universit de Sfax, Tunisie.
Corresponding author: Ridha Ben Salem. Laboratoire de Chimie Organique-Physique UR11ES74, Facult des
Sciences de Sfax, B.P 1171 3038 Sfax, Universit de Sfax, Tunisie. E-mail : ridha.bensalem@voila.fr

212

Ghayth Rigane and Ridha Ben Salem

1. INTRODUCTION
Olives (Olea europea L.) are the most widespread and valuable plant in Mediterranean
countries. Chehab et al. (2013) mentioned that world-wide production of olive oil during the
last 20 years increased by almost 70% (from 1.7 to 2.8 million tons). Olive oils makes up a
small proportion (<3.5%) of the volume in the world vegetable oil market. However, in terms
of product value, only olive oil has a 15% share of world trade (Luchetti, 2000). According to
the International Olive Council (IOC) statistics, there is an equilibrium exists between olive
oil production and consumption worldwide, which is interesting taking into account the
increase in production the last decade.
Olive oil production currently reaches the 3 Mt (Tsimidou, 2012).
Olives (Olea europaea L.) are a major part of the agriculture and gastronomy in many
countries in Europe, North Africa, and Asia Minor surrounding the Mediterranean Sea. They
are one of the main sources of fat in the Mediterranean diet. The oval-shaped olives can be
consumed as table olives or as (virgin) olive oil. The edible part of the olive is called pulp and
the hard central part, which usually is discarded as waste, is called the kernel (Rigane et al.,
2013 a; b; Moghaddam et al., 2012).
Fatty acids, which are made up by long chains of carbon atoms, are extremely useful in
the characterization of olive oils. The health-related benefits of olive oil have also partly been
attributed to the high amount of oleic acid present in this type of vegetable oil (Moghaddam et
al., 2012). The virgin olive oil composition depends on numerous factors such as the
interaction between the cultivar and the environment, cultivation techniques, fruit ripeness
and the oil extraction system.
The influence of the cultivar on oil quality is being considered with increasing interest
because its determination could be used to produce mono-varietal virgin olive oils, thus
increasing product value (Haddada et al., 2008).

2. LEVELS OF PALMITIC ACID IN TUNISIAN OLIVE OIL


In Tunisia, the olive oil sector plays an important role in the economy, providing both
employment and export revenues. Olive trees cover an area of 1.6 million ha with an annual
oil production of 170 000 t (Rigane et al., 2011). The last decade, Tunisian autochthonous
varieties were extensively studied for their potential with regard to VOO quality
characteristics. The oils obtained from healthy fruits using laboratory scale extraction, most of
the time with the aid of an Abencor type mill and extractor (Oueslati et al., 2009). Thus, the
amounts of bioactive phytochemicals reported are rather high in comparison to what is
reported for VOOs from the main producing countries. This illusive picture does not reflect
necessarily the real content of this nutrient in the commercial products sold locally or of those
exported.
The characterization of virgin olive oils from six Tunisian cultivars, namely Chtoui, Ain
Jarboua, Jarboui, Regregui, Rekhami and Neb Jmel, grown in Nebeur (a region of the Kef)
was carried out by Haddada et al. (2008). They reported that palmitic acid is the major
saturated fatty acid in olive oil; its content ranged between 13.46 and 16.33%. On the other
hand, Rigane et al. (2013a, b) had studied three rare olive cultivars, such as, Jemri-

Palmitic Acid in Tunisian Olive Oil: Updating and Perspective

213

Bouchouka, Gemri-Dhokar and Dhokar olive oils grown under the same growth conditions in
the south of Tunisia. In these works, they mentioned that Dhokar olive oil was characterized
by its high palmitic acid level which means this oil freezes at low temperatures (19.37 %). In
addition, Oueslati et al., (2009) have studied four olive oils from southern of Tunisia. They
found that Chemlali variety yields oils of rather high concentration of palmitic acids (13.8716.06 %). Compared to some secondary Tunisian VOOs (Baccouri et al., 2007; Mana et al.,
2007) Chemlali Tataouine, Fakhari Douirat and Zarrazi Douirat varieties produced oils with
excellent fatty acid (FA) composition, i.e., low palmitic acid content (minimum of 8.67%)
which was lower than the upper limit of 5% established for extra virgin olive oil (EVOO)
(EEC, 2003).
Some of these FA ratios may prove useful in chemometric studies to discriminate oils on
the basis of origin (Aparicio et al., 1999) because they showed great variability depending on
the crop seasons, maturity index and the genetic factor.
To our knowledge such data are limited or missing. Among the many autochthonous
varieties the major ones are Chemlali (central and southern Tunisia) and Chtoui (northern
Tunisia), the former being more resistant to lack of rainfall. Some interesting data for the
evolution of palmitic acid during maturation are given by Damak et al. (2008). In this report,
this research team mentioned that during maturation, as fruit ripened, the content of palmitic
acids slightly decreased from 16.230.28% to 9.350.65%. Furthermore, it is well-known
that, in addition to the maturity stage, fatty acid composition could be affected by
environmental factors such as rainfall and geographical origin (Ben Temime et al., 2006).
While, Issaoui et al. (2008) studied the effect of harvesting time on the FA composition of the
five cultivars including: Zarrazi Zarzis, Jeddaria Chaal, Chemchali Chouamekh, Chemlali
Zarzis and Chemchali Gafsa. They found that the decreases in the level of palmitic acid are
variety dependent (55.92% of variability), and there was a significant cultivar kind (P <
0.001) and harvest timing (42.65% of variability; p < 0.001) effect in all varieties studied.
Hence, Zarrazi Zarzis show a decline of about one-half at the last part of maturation (from
12.83 to 7.74%); in contrast, a slight decrease was shown for the cultivars Chemlali
Chouamekh and Chemlali Zarzis (from 19.02 to 13.81% and from 19.06 to 13.61%,
respectively). the decrease of the palmitic acid during maturation was explained by GomezRico et al. (2005): In fact, the increase in oleic acid content is a result of the active
biosynthesis of TAGs, which takes place through fruit ripening, involving a fall in the relative
percentage of the oils palmitic acid content.
On the other hand, Bedbabis et al. (2010) reported the effect of waste water irrigation on
the extra virgin olive oil quality from the Tunisian cultivar Chemlali. The objective of this
work was to verify if an irrigation with Treated Waste Water (TWW) over 4 years affects the
Extra Virgin olive oil quality from Tunisian cv. Chemlali. Moreover, the amounts of
palmitic (C16:0) varied from 16.28 to 19.92%. The variability of fatty acid composition of the
sampled oil was within the normal range expected for olive oils (Codex, 1989). Differences
between our data and those reported for other varieties indicated that the olive cultivar affects
oil fatty acid composition, confirming previous studies (Inglesse et al., 1996; Ranalli et al.,
1997; Patumi et al., 1999; Dhifi et al., 2004; Ben Temime et al., 2006; Baccouri et al., 2007).
In addition, Gharsallaoui et al., (2011) studied the effect of irrigation with TWW on
Chemlali olive oil quality. This study focuses also on the determination of the quality of
oils extracted from hand picked olives and from olives fallen under the trees irrigated with
reclaimed water. Regarding palmitic acid, the difference between the treatments was not

214

Ghayth Rigane and Ridha Ben Salem

significant with values ranging between 19.88 and 20.23%. Moreover, it has been note that
the percentages of this acid in the samples under investigation were very close to the upper
limit set by the IOC (International Oleicol Council) (20%) and even exceed the limit for the
WW-Fl plot. These values are similar to those found for Chemlali variety grown in the Sfax
region and in the South of Tunisia (Dabbou et al., 2010; Issaoui et al., 2010).
Several studies have reported that irrigation can affect the fatty acid composition (Ranalli
et al., 1997; Aparicio and Luna, 2002). At the end of our experiment, palmitic, palmitoleic,
stearic and linoleic acids were found in high concentrations in oils coming from TWW
irrigated plot, respect to acid contents in oil from trees grown under rain-fed conditions. In
addition, these results indicated that irrigation with TWW seems to stimulate these fatty acid
enzymes synthetase.

3. ANALYTICAL ASPECTS OF PALMITIC ACID DETERMINATION


The analytical method for the determination of the palmitic acid was expressed as relative
percent of total area of fatty acid. The fatty acid composition of the oils was determined by
gas chromatography (GC) as fatty acid methyl esters (FAMEs). FAMEs were prepared by
saponification/ methylation with sodium methylate according to the EEC 2568/9119.
The chromatographic analysis was performed with a gas chromatograph using a capillary
column Stabilwax 50 m x 0.32 mm and 0.25 m film thickness (Restek Corporation,
Bellefonte, PA, US).
The column temperature was isothermal at 180 C and injector and FID temperatures
were set at 230 C and 250 C, respectively. The carrier gas was nitrogen. The injection
volume was 1 mL (Bouaziz et al., 2010; Manai-Djebali et al., 2012; Rigane et al., 2013). On
the other hand, Issaoui et al. (2008) focused on the changes of pomological characteristics, oil
content, FA composition and sugars occurring during ripening in some Tunisian olive
varieties studied for the first time in an attempt to establish an optimum harvest period.
To attend these objectives, they analysed the fatty acid composition by the extraction of
the lipophilic fraction was carried out with the procedure described by Allen and Good (1971)
and Hannachi et al. (2007).
The FAmethyl esters (FAMEs) were prepared by dissolving 0.1 g of oil in 2 mL of
heptanes and a solution of KOH (0.2 N) in methanol in which FAMEs were obtained.
Individual FAMEs were separated and quantified by gas chromatography (GC). The
temperature was programmed to increase from 170 to 270 C at a rate of 5C min-1.

CONCLUSION
As can be seen from this review, the level of palmitic acid in olive oil, as the major
saturated acid form, is the result of a complex interaction of various factors. From the existing
literature concerning the characterization of palmitic acid in the Tunisian olive oils, it is clear
that the main focus has been directed toward the two most important varieties: Chtoui and
Chemlali.
Currently, most cultivars may yield virgin olive oils with more than 19 %.

Palmitic Acid in Tunisian Olive Oil: Updating and Perspective

215

Nevertheless, bearing in mind the diversity of Tunisian olive germplasm, a widening of


the study to other varieties is called for. GC methodologies are established for quantitative
determination of palmitic acid.

ACKNOWLEDGMENTS
The authors thank the Tunisian Ministry of Higher Education and Scientific Research for
financial support and are grateful to Professor Mohamed Rigane for useful discussions about
the English.

REFERENCES
Allen, C. and Good, P. (1971). Acyl lipids in photosynthetic systems. In: Methods in
Enzymology, Vol. 23 (S. P. Clowic and N. O. Kaplan, eds.) pp. 523547, Academic
Press, New York, NY.
Aparicio, R. and Luna, G. (2002). Characterisation of monovarietal virgin olive oils.
European Journal of Lipid Science and Technology, 104, 614627.
Aparicio, R., Roda, L., Albi, M. A., and Gutirrez, F. (1999). Effect of various compounds on
virgin olive oil stability measured by Rancimat. Journal of Agriculture and Food
Chemistry, 47, 41504155.
Baccouri, B., Ben Temime, S., Taamalli, W., Daoud, D., MSallem, M., and Zarrouk, M.
(2007). Analytical characteristics of virgin olive oils from two new varieties obtained by
controlled crossing on Meski variety. Journal of Food Lipids, 14, 1934.
Bedbabis S., Bchir Ben Rouina and Makki Boukhris. (2010). The effect of waste water
irrigation on the extra virgin olive oil quality from the Tunisian cultivar Chemlali.
Scientia Horticulturae, 125, 556561.
Ben Temim, S., Campeol, E., Luigi Cioni, P., Daoud, D., and Zarrouk, M. (2006). Volatile
compounds from Chtoui olive oil and variations induced by growing area. Food
Chemistry, 99, 315325.
Chehab, H., Issaoui, M., Flamini, G., Mechri, B., Attia, F., Pier Luigi, C., Boujnah, D.,
Hammami, M. (2013). Oil quality and aroma composition of Chemlali olive trees (Olea
europaea L.) under three irrigation regimes. African Journal of Agricultural Research,
8(48), 6291-6299.
Codex, 1989. stan 33. Norme codex pour les huiles dolive vierges et raffines et pour lhuile
de grignons dolive raffine Codex stan 33-1981 (Rv. 1-1989).
Dabbou, S., Rjiba, I., Nakbi, A., Gazzah, N., Issaoui, M., and Hammami, M. (2010).
Compositional quality of virgin olive oils from cultivars introduced in Tunisian arid
zones in comparison to Chemlali cultivars. Horticultural Science, 124, 122127.
Damak, N., Bouaziz, M., Ayadi, M., Sayadi, S., and Damak, M. (2008). Effect of the
maturation process on the phenolic fractions, fatty acids and antioxidant activity of the
Chetoui olive fruit cultivar. Journal of Agriculture and Food Chemistry, 56, 15601566.

216

Ghayth Rigane and Ridha Ben Salem

Dhifi, W., Hamrouni, I., Ayachi, S., Chahed, T., Saidani, M., and Marzouk, B., 2004.
Biochemical characterization of some Tunisian olive oils. Journal of Food Chemistry, 11,
287296.
EEC (2003). Characteristics of olive and olive-pomace oils and on their analytical methods.
Official Journal of the European Communities, L295, 5777. Regulation EEC/2568/91,
Regulation EEC/ 1989/03.
Gharsallaoui, M., Benincasa, C., Ayadi, M., Perri, E., Khlif, M., and Gabsi, S. (2011). Study
on the impact of wastewater irrigation on the quality of oils obtained from olives
harvested by hand and from the ground and extracted at different times after the
harvesting. Scientia Horticulturae, 128, 2329.
Haddada, F. M., Krichene, D., Manai, H., Oueslati, I., Daoud, D., and Zarrouk, M. (2008).
Analytical evaluation of six monovarietal virgin olive oils from northern Tunisia.
European Journal of Lipid Science and Technology, 110, 905913.
Hannachi, H., Msallem, M., Ben Elhadj, S., and El Gazzah, M. (2007). Influence du site
gographique sur les potentialits agronomiques et technologiques de lolivier (Olea
europaea L.) en Tunisie. Comptes Rendus Biologies, 330, 135142.
Inglesse, P., Barone, E. and Gullo, G. (1996). The effect of complementary irrigation on fruit
growth, ripening pattern and soil characteristics of olive (Olea europaea L) cv. Carolea.
The Journal of Horticultural Science and Biotechnology, 71, 257263.
Issaoui, M., Mechri, B., Echbili, A., Dabbou, S., Yangui, A., Belguith, H., Trigui, A., and
Hammami, M. (2008). Chemometric characterization of five tunisian varietals of olea
europaea l. olive fruit according to different maturation indices. Journal Food Lipids, 15:
277-296.
Issaoui, M., Flamini, G., Brahmi, F., Dabbou, S., Ben Hassine, K., Taamali, A., Chehab, H.,
Ellouz, M., and Zarrouk, M., Hammami, M. (2010). Effect of the growing area conditions
on differentiation between Chemlali and Chtoui olive oils. Food Chemistry, 119, 220
225.
Luchetti, F. (2000). Introduction: In: Handbook of Olive Oil: Analysis and Properties (Eds.,
Harwood, J. Aparicio, R.) Aspen Publishers, Inc., Gaithersburg, MD, US, pp. 1-16.
Mana, H., Haddada, F. M., Trigui, A., Daoud, D., and Zarrouk, M. (2007). Compositional
quality of virgin olive oil from two new Tunisian cultivars obtained through controlled
crossings. Journal of the Science of Food and Agriculture, 87, 600606.
Manai-Djebali, H., Krichene, D., Ouni, Y., Gallardo, L., Sanchez, J., Osorio, E., Daoud, D.,
Guido, F., and Zarrouk, M. (2012). Chemical profiles of five minor olive oil varieties
grown in central Tunisia. Journal of Food Composition and Analysis, 27, 109119.
Oueslati, I., Anniva, C., Daoud, D., Tsimidou, M. Z., and Zarrouk, M. (2009). Virgin olive oil
(VOO) production in Tunisia: The commercial potential of the major olive varieties from
the arid Tataouin zone. Food Chemistry, 112, 733741.
Patumi, M., DAndria, R., Fontanaza, G., Morelli, G., Gioro, P., and Sorrentino, G. (1999).
Yield and oil quality of intensively trained trees of three cultivars of olive (Olea europaea
L.) under different irrigation regimes. Journal of Horticultural Science and
Biotechnology, 74, 729737.
Ranalli, A., de Mattia, G., Ferrante, M. L., and Giansante, L. (1997). Incidence of olive
cultivation area on the analytical characteristics of the oil. Note 1. La Riv. Ital. Sostanze
Grass 74, 501508.

Palmitic Acid in Tunisian Olive Oil: Updating and Perspective

217

Rigane, G., Boukhris, M., Bouaziz, M., Sayadi, S., and Ben Salem, R. (2013a). Analytical
evaluation of twomonovarietal virgin olive oils cultivated in the south of Tunisia: JemriBouchouka and Chemlali-Tataouin cultivars. Journal of Agriculture and Food Chemistry,
93, 1242-1248.
Rigane, G., Ayadi, M., Boukhris, M., Sayadi, S., and Bouaziz, M. (2013b). Characterisation
and phenolic profiles of two rare olive oils from southern Tunisia: Dhokar and GemriDhokar cultivars. Journal of Agriculture and Food Chemistry, 93, 527-534.
Rigane, G., Ben Salem, R., Sayadi, S., and Bouaziz, M. (2011). Phenolic composition,
isolation and structure of a new deoxyloganic acid derivative from Dhokar and GemriDhokar olive cultivars. Journal of Food Science, 76(7), 965-973.
Tsimidou, M. Z. (2012). Virgin olive oil (voo) and other olive tree products as sources of tocopherol, updating and perspective. Editors: Angel Catala: Tocopherol: Sources, Uses
and Health Benefits.

INDEX
#
21st century, 94

A
accounting, 70
acetone, 68, 86, 200
acetylcholine, 5, 13
acidity, 25, 71
active site, 34
active transport, 32
acylation, 74, 134, 135, 138, 146
AD, 97, 100, 119, 130
adaptation, 157
adenine, 30, 85
adenosine, 6, 36, 79
adenosine triphosphate, 6
adhesion, 20, 91, 152
adipocyte, 34, 107, 108, 118
adiponectin, ix, 106, 107, 108, 117
adipose, 3, 11, 21, 35, 37, 38, 40, 48, 49, 52, 53, 58,
61, 73, 78, 79, 83, 86, 90, 99, 106, 107, 114, 116,
117, 119, 123
adipose tissue, 3, 11, 21, 37, 38, 40, 48, 49, 52, 54,
58, 61, 73, 78, 79, 83, 86, 90, 99, 106, 107, 114,
116, 117, 119, 123
adiposity, ix, 105, 119
adolescents, 112, 122
ADP, 85
adults, 33, 39, 42, 115, 116, 118, 121, 122, 148
adverse effects, 47, 108
Africa, 71
age, 21, 112, 114, 150, 152, 156
agriculture, 212
Agrobacterium, 38
AIDS, 39
alcohols, ix, 125, 130, 131, 132, 133, 161

aldehydes, 130, 131


algae, viii, 17, 21, 34
alkane, 131, 140, 177, 178, 179, 186
alpha-tocopherol, 201
alters, 58, 108
American Heart Association, 9, 10, 14
amino, 20, 90
amino acid, 20
angiotensin II, 5
anorexia, 34
anorexia nervosa, 34
antimicrobial agent, vii, 17, 26, 38
antioxidant, 57, 94, 95, 143, 215
aorta, 5, 12, 13
apoptosis, 26, 33, 37, 39, 40, 43, 90, 91, 92, 102,
104, 107, 111, 113, 116, 117, 123
appetite, 33
aquaculture, 60
aqueous solutions, 83
Arabidopsis thaliana, 143
arginine, 2
aromatics, 132
arterioles, 3, 11
artery(s), vii, 1, 9, 10, 11, 12
Asia, 59, 61, 94, 97, 212
Aspergillus terreus, 26
assessment, 95, 116
atherosclerosis, 4, 6, 48, 59, 60, 106
atherosclerotic plaque, 9
atoms, 20, 126, 129, 177, 195, 197
atopic dermatitis, 41
ATP, 4, 5, 6, 8, 9, 13, 20, 27, 64, 79, 83, 84, 85, 100
attachment, 28, 81
attribution, 126
autism, 151

220

Index

B
bacteria, vii, 17, 21, 25, 26, 28, 152, 153
bacterial cells, 37
Bangladesh, 38
base, 68, 126
basilar artery, 5
beauty products, vii, 1
beef, 22, 42, 49, 50, 52, 57, 58, 59, 61, 68, 73, 92,
106
beneficial effect, 78, 110, 148, 150, 151, 152, 153
benefits, vii, x, 1, 17, 35, 145, 148, 151, 153, 212
bile, 146
biochemical action, 24, 25
biochemical processes, 27
biochemistry, vii, viii, 1, 17, 18, 27, 34, 93, 94, 99,
100, 139, 156
biodegradation, 141
biodiesel, 20, 36, 188
biological processes, 9
biological signaling, vii, 1
biological systems, vii, 1, 2, 20
biomarkers, 31, 34, 122, 123
biomolecules, 18
biosynthesis, 13, 28, 37, 79, 81, 88, 126, 127, 130,
135, 136, 137, 138, 139, 140, 141, 142, 213
biosynthetic pathways, 29, 134
biotechnology, 18, 20, 38
biotin, 79, 80
birds, 52
birth weight, 156
Black Sea, 51
bleaching, 71, 94
blends, 77, 96, 97
blood, viii, ix, 2, 3, 9, 12, 32, 36, 37, 45, 46, 47, 62,
86, 87, 88, 92, 101, 106, 108, 109, 110, 112, 118,
120, 123, 153
blood pressure, 3, 12
blood stream, 86, 153
blood vessels, 3
blood-brain barrier, 33, 36
bloodstream, 83, 86
body fat, 57
body mass index, 111, 119
body weight, 148, 154
bonds, 24, 66, 68, 126, 128
bone, x, 93, 107, 145, 149, 150, 153, 155, 156, 157
bone growth, 150
bone marrow, 107
bone mass, 149
Botswana, 141
bowel, 88
bradykinin, 4

brain, 11, 28, 32, 35, 40, 41, 42, 46, 83, 86, 100
Brazil, 63, 71, 72, 75, 95, 97
breast milk, viii, x, 63, 78, 145, 146, 149
breastfeeding, 148
building blocks, 79
by-products, 160, 202

C
Ca2+, 4, 5
cacao, 133
caffeine, 202
calcium, x, 3, 4, 9, 12, 57, 58, 59, 78, 98, 145, 148,
152, 153, 156
calcium channel blocker, 4, 9, 12
caloric intake, 87, 88
cancer, 13, 107, 113, 151, 155, 157
capillary, 214
carbohydrate(s), 18, 19, 34, 39, 41, 47, 66, 109, 112,
118, 119, 149
carbohydrate metabolism, 41
carbon, vii, 1, 2, 3, 7, 8, 14, 20, 24, 26, 35, 66, 68,
75, 79, 81, 82, 85, 86, 99, 109, 126, 127, 129,
130, 131, 136, 139, 142, 146, 159, 160, 163, 177,
178, 193, 194, 195, 197, 198, 201, 205, 212
carbon atoms, 68, 85, 126, 127, 129, 130, 136, 159,
160, 177, 178, 193, 194, 195, 197, 198, 201, 205,
212
carbon dioxide, 35, 86, 163
carbon monoxide, 131
carboxyl, 7, 9, 18, 81, 131, 132
carboxylic acid, 84, 160
carcinogenicity, 37, 58
cardiac arrest, 9
cardiac muscle, 83
cardiomyopathy, 102
cardiovascular disease(d), viii, 2, 4, 6, 12, 19, 33, 34,
37, 41, 45, 46, 47, 63, 97, 105, 106, 109, 112,
115, 118, 119, 120, 121
cardiovascular function, 4
cardiovascular risk, 46, 102
carotene, 75, 94
carotenoids, 71, 73
cascades, 103
catabolism, 32
catalysis, 27
catalyst, 76, 78
catfish, 55, 56, 60
cattle, 52, 54, 59, 60, 61
CBS, 64
C-C, 18
cell culture, 90
cell death, 3, 9, 10, 14, 90, 102

Index
cell line, 102
cell membranes, ix, 20, 26, 27, 86, 125, 129
cell organelles, 25
cell organization, 27
cell signaling, 20, 28, 37
cell surface, 70, 92, 155
central nervous system, vii, 1
central obesity, 37, 107
ceramide, 39, 107, 108, 111, 117, 123
cerebral blood flow, 3
challenges, 18, 94
channel blocker, 3
cheese, 46, 50, 74, 95
chemical(s), 24, 34, 38, 39, 40, 68, 70, 71, 72, 74,
76, 77, 92, 97, 147, 150, 201
chemical characteristics, 97
chemical properties, 24, 70, 71, 72, 147
chemical reactions, 68, 92
chemokines, 91, 92
chicken, 49, 96, 106
childhood, 149, 155, 157
children, 40, 149
Chile, 73
chloroform, 68
chloroplast, 128, 139
cholesterol, viii, ix, 6, 31, 34, 36, 39, 41, 45, 46, 47,
49, 52, 59, 61, 70, 72, 74, 86, 87, 88, 89, 92, 100,
105, 106, 107, 109, 111, 112, 114, 115, 116, 119,
121, 122, 124, 136, 146, 201
cholesterolaemia, viii, 45, 48
choline, 136, 137, 138, 143
chromatography, 37, 133, 214
chronic diseases, 62
circulation, vii, 1, 2, 4, 5, 6, 66, 110
cirrhosis, 89, 113
classes, ix, 26, 57, 87, 125, 141
classification, 42
cleavage, 85
climates, 71
clinical trials, 118, 155
clusters, 134
CNS, 10
CO2, x, 79, 81, 84, 131, 159, 161, 162, 163, 164,
165, 166, 167, 168, 169, 170, 171, 172, 173, 174,
175, 178, 179, 182, 184, 185, 186, 187, 188, 189,
190, 191, 192, 193, 194, 195, 198, 200, 201, 202,
203, 204, 205
cocoa, viii, 63, 71, 72, 77, 92, 95, 106, 133, 160,
197, 198
cocoa butter, viii, 63, 71, 72, 77, 92, 95, 106, 160,
197, 198
coconut oil, vii, ix, 1, 47, 76, 77, 87, 105, 106
coenzyme, vii, 1, 2, 80, 83, 84, 88, 98, 140

221

coffee, 202
colic, 150
colitis, 152, 156, 157
collagen, 89
colon, 152, 153
colonization, 41, 151
color, 75, 94
combustion, 89
commercial, 202, 212, 216
commodity, 146
communication, 33
community, 66, 122, 151, 157
complement, 34
complexity, 187
complications, 12, 91, 102, 103, 153, 169
compounds, x, 7, 28, 35, 66, 71, 84, 134, 159, 161,
162, 163, 164, 182, 194, 200, 201, 202, 204, 215
comprehension, 18
condensation, 79, 80, 81, 82, 127
conductance, 4, 30
configuration, 66, 126
conflict, 10
conflict of interest, 10
Congress, 57, 102
conservation, 56
conserving, 49
constant rate, 53
constituents, 6, 18, 41, 66, 111, 126, 131, 132, 142,
147, 198
Constitution, 58, 141
construction, 66
consumers, 49
consumption, viii, 34, 48, 58, 59, 73, 86, 87, 88, 89,
92, 105, 109, 110, 111, 116, 120, 148, 150, 212
consumption patterns, 59
control group, 149, 150
controlled trials, 39, 119, 121
controversial, 87
convergence, 102
COOH, 18, 26, 68
cooking, 66, 71, 76
cooling, 96
cooperation, 128, 151
coordination, 27, 28
coronary arteries, 6, 11
coronary artery disease, 6, 10
coronary heart disease, ix, 42, 47, 49, 62, 87, 105,
106, 109, 114, 116, 119, 121
correlation(s), 30, 54, 55, 148, 169, 177, 178, 179,
186
cortical neurons, 15
cosmetic(s), vii, 1, 18, 20, 38, 70, 73, 76, 160
cost, 60, 70, 77, 92, 94, 184, 191

222

Index

cotton, 77
covalent bond, 28
CPT, 8, 64, 84, 136
crises, 102
critical value, 88, 92
crop, 213
crude oil, 197
crystalline, 24, 77
crystallization, 71, 96
CT, 64, 79, 80
cues, vii, 17
cultivars, 73, 95, 212, 213, 214, 215, 216, 217
cultivation, 212, 216
culture, 37, 39, 102, 134, 136, 215
culture conditions, 37
cuticle, ix, 125, 131
cutin, 131, 132, 141
CVD, viii, 19, 33, 105, 106, 107, 109, 111, 115, 116
cycles, 8, 85, 127
cysteine, vii, 17, 20, 81
cytochrome, 3, 11, 83, 90, 132
cytokines, 34, 90, 91, 102
cytoplasm, 8, 79, 134

D
dairy products, viii, 45, 48, 63, 68, 74, 106, 109
data set, 168, 171, 172
database, 95
deaths, 6, 9
decomposition, 71
decoupling, 27
Deer, 50
defects, 90
deficiency, 91, 94, 107, 117, 152
deformation, 27
degradation, 8, 30, 83, 142, 152
degumming, 71
dehydration, 80, 82
dendritic cell, 107
deoxyribonucleic acid, 116
Department of Agriculture, 67, 93
Department of Health and Human Services, 115
deposition, 61, 62, 113
depression, 55, 165, 170
derivatives, x, 38, 90, 131, 132, 138, 159, 160, 161,
164, 165, 173, 175, 178, 179, 184, 187, 188, 191,
194, 195, 197, 199, 200, 204, 205
detection, 116
detergents, 35, 70
detoxification, 89
developed countries, 49
developmental process, 15

diabetes, ix, 4, 33, 39, 91, 100, 102, 103, 106, 107,
110, 111, 114, 116, 120, 121, 124
diabetic kidney disease, 103
diabetic patients, 122
diacylglycerol, 91, 104, 107, 117, 130, 135, 136,
142, 143
diarrhea, 151
diet, viii, 31, 33, 45, 46, 47, 48, 49, 52, 54, 55, 56,
57, 59, 61, 62, 63, 69, 78, 86, 87, 88, 89, 100,
101, 106, 108, 110, 111, 116, 118, 119, 122, 146,
147, 150, 151, 152, 153, 212
diet composition, 54
dietary fat, 37, 39, 52, 54, 57, 59, 60, 61, 62, 87, 100,
106, 109, 110, 118, 119, 120, 121, 122
Dietary Guidelines, 115
Dietary Guidelines for Americans, 115
dietary habits, ix, 106, 115
dietary intake, ix, 5, 6, 28, 54, 93, 105, 110, 112,
114, 123
dietary lipid sources, viii, 45
dietary sources, viii, 42, 45, 106
dietary supplementation, 52
diffusion, 90, 131
digestibility, 60, 74
digestion, 57, 59, 61, 78, 93, 120, 147, 148, 151,
153, 156
digestive system, viii, 45
discomfort, 150
discrimination, 139
diseases, viii, 6, 20, 34, 49, 63, 90, 102, 106, 107,
115, 153
disorder, 26, 30
dispersion, 97, 191
displacement, 27
distillation, 71, 161, 162
distribution, viii, 18, 21, 22, 28, 34, 39, 57, 63, 78,
98, 119, 120, 123, 141, 142, 146, 147, 156, 201
diversification, 39
diversity, 18, 126, 215
DNA, 26
docosahexaenoic acid, 4, 5, 66, 67, 96, 194
DOI, 98, 101
donors, 3, 11, 136
double blind study, 151
double bonds, 66, 126, 128, 198, 205
down-regulation, 103, 108, 118
drugs, 40
dry matter, 54, 95, 134
duodenum, 59
dyslipidemia, 106, 107, 109

223

Index

E
ecosystem, 151
editors, 139, 140, 143
egg, 46, 51, 56, 57, 59
eicosapentaenoic acid, 12, 13, 41, 67, 194
electron(s), 9, 26, 27, 32, 84, 85, 90, 131
elongation, ix, 6, 8, 14, 31, 48, 61, 82, 99, 122, 125,
126, 127, 128, 130, 132
e-mail, 105, 125
employment, 212
emulsions, 61
encoding, 142
endogenous synthesis, 31
endosperm, 140
endothelial dysfunction, ix, 4, 106
endothelial NO synthase, 13
endothelium, 3, 4, 5, 6, 11, 12, 13, 83
energy, ix, x, 8, 18, 20, 26, 27, 28, 29, 30, 31, 33, 34,
37, 38, 42, 51, 57, 66, 72, 75, 77, 79, 83, 86, 87,
89, 98, 105, 109, 110, 111, 117, 118, 134, 138,
145, 146, 148, 149, 151, 205
energy expenditure, ix, 26, 31, 33, 38, 98, 105, 117
engineering, 93
environment, 21, 37, 51, 88, 147, 151, 212
environmental conditions, ix, 125
environmental factors, viii, 45, 213
environmental stress(s), 131
enzymatic activity, 131
enzyme(s), viii, 6, 3, 8, 11, 19, 20, 24, 26, 27, 34, 48,
49, 59, 61, 63, 69, 75, 78, 79, 81, 82, 84, 85, 86,
88, 91, 99, 100, 113, 127, 130, 132, 134, 136,
137, 138, 139, 142, 143, 146, 214
EPA, 4, 12, 41, 64, 159, 194, 197, 198, 205
epidemiologic, 111
epidermis, 131
epinephrine, 79
epithelial cells, 55
epithelium, 86, 152
equilibrium, 160, 161, 165, 166, 168, 175, 180, 183,
197, 212
equipment, 128, 162
erosion, 152
erythrocytes, 112, 114
ESI, 141
essential fatty acids, 4, 61
ester, vii, viii, x, 1, 2, 7, 14, 18, 22, 63, 68, 69, 83,
121, 140, 147, 159, 174, 181, 191, 193, 194, 195,
199
ethanol, 68, 164, 187, 188, 189, 190, 198, 200, 204
etiology, 101, 107
eukaryotic, vii, 1, 2, 17, 28, 128, 134, 151
eukaryotic cell, vii, 1, 2, 28, 151

Europe, 72, 212


evidence, viii, ix, x, 12, 28, 33, 34, 35, 45, 47, 89,
91, 106, 109, 111, 115, 116, 119, 120, 121, 132,
137, 140, 145, 146, 153, 194
evolution, 213
excretion, 6, 148
exercise, 14, 154
experimental condition, 203
exports, 70
exposure, 10, 33, 38, 90, 103
extraction, 73, 95, 133, 155, 160, 161, 162, 163, 164,
166, 194, 198, 202, 203, 204, 205, 212, 214
extracts, 28, 41, 160, 161, 197, 202, 203

F
FAD, 8, 64, 84, 85
families, 129, 132
family members, 102
FAS, 64, 79, 81, 83, 129, 130
fast food, 106
fasting, 31, 42, 83, 86, 114, 119, 120, 124
fasting glucose, 114
fat intake, 106, 110, 121
feedstocks, 139
fermentation, 59, 95
fetal growth, 33, 38
fiber, 51
fibrate, 122
fibrinolysis, 62
fibrosis, 89
financial, 215
financial support, 215
Finland, 106, 116
fish, viii, 4, 5, 6, 45, 46, 48, 51, 52, 55, 56, 57, 58,
59, 60, 61, 191, 194, 198, 199
fish oil, 4, 5, 6, 52, 56, 57, 60, 191, 194, 198, 199
flammability, 187
flavor, 74
flora, x, 145, 151, 152, 155, 157
flowers, 23, 36, 132
fluid, x, 159, 160, 161
fluid extract, 160, 161
food, viii, ix, 18, 20, 40, 41, 45, 48, 49, 50, 51, 57,
58, 60, 63, 66, 70, 71, 74, 76, 77, 92, 94, 95, 106,
111, 115, 116, 121, 148
food industry, viii, 63, 71, 76
food products, 74, 77, 95
food spoilage, 41
foodborne pathogens, viii, 17
football, 123
formation, vii, 6, 17, 20, 39, 78, 127, 128, 135, 136,
137, 138, 140, 142, 143, 148

224

Index

formula, 68, 78, 97, 98, 147, 148, 149, 150, 151,
152, 156, 157, 158
fragility, 60
fragments, 127
France, 57
free energy, 81
freshwater, 51, 58, 61
fruits, 23, 36, 70, 73, 97, 129, 133, 134, 138, 140,
141, 142, 212
FTICR, 141
fungal infection, 132
fungi, 26, 39, 126
fungus, 21, 25, 30, 34
fusion, 39, 40

G
ganglion, 2, 11
gastrointestinal tract, 151
gel, vii, 1
gene expression, 88, 91, 103, 108, 111, 119, 153,
156
genes, 88, 91, 107, 130, 142
genetic factors, 57
genetic marker, 91
genome, 18
geographical origin, 213
germination, 134
gestational age, 156
ginseng, 134, 142
gland, x, 55, 74, 145, 146
glucagon, 79
gluconeogenesis, 31, 39, 86
glucose, viii, ix, 33, 39, 63, 79, 86, 91, 92, 103, 106,
107, 108, 110, 118, 124
glucose tolerance, 124
glutathione, 89
glycerol, viii, ix, x, 48, 61, 63, 66, 70, 73, 75, 76, 77,
92, 125, 128, 131, 132, 135, 138, 145, 146
glycogen, 89
glycolysis, 79, 135
God, 72
Gori, 95
GRAS, 162
grass, 52, 57, 60
growth, 25, 27, 33, 37, 38, 41, 42, 57, 59, 60, 61, 77,
136, 146, 149, 152, 213, 216
growth temperature, 41
guidelines, 106, 119
Guinea, 35

H
habitat, 151
hair, 42
hardness, 71, 78, 98, 157
harvesting, 95, 213, 216
H-bonding, 191
HE, 95
health, vii, x, 1, 9, 17, 18, 33, 34, 35, 38, 39, 42, 47,
49, 52, 57, 58, 59, 78, 98, 99, 106, 116, 145, 150,
151, 152, 154, 155, 212
health care, 9
health care costs, 9
health effects, vii, 18, 42, 152
health promotion, 35
health services, 150
heart disease, 14, 59, 110
heart failure, 33, 39
helminthes, viii, 17
hematology, 58
heme, 11
hemodialysis, 122
hepatocellular carcinoma, 89
hepatocytes, 31, 36, 39, 40, 90, 102
hepatoma, 33, 43
hexane, 202
high fat, 86, 106, 147
hippocampus, 9
HIV, 34, 39, 40
HIV-1, 39, 40
HM, 95, 96, 98, 124, 142
homeostasis, vii, 17, 27, 33, 35, 83, 116, 118, 152
homocysteine, 31, 42
hormone(s), 33, 89
host, 21, 28, 36, 151, 155, 157
House, 59
housing, 58
human body, 151
human health, vii, 17, 38, 47, 66, 76
human milk, x, 38, 75, 77, 78, 96, 98, 106, 145, 146,
147, 148, 153, 154, 155, 156
human subjects, 36, 37
Hunter, 119
hybrid, 60
hydrocarbons, 41, 130
hydrogen, 24, 68
hydrogen bonds, 68
hydrogenation, 71, 97
hydrolysis, 34, 48, 83, 147
hydrophobicity, 18, 20
hydroxide, 69
hydroxyl, 7, 66, 69, 75, 81, 131
hydroxyl groups, 66, 75

225

Index
hygiene, 151
hypercholesterolemia, viii, 12, 63, 87, 110
hyperglycemia, 90
hyperlipidemia, 6, 113, 114, 122
hypertension, 4, 5, 6, 11, 106, 107, 113, 122
hypertriglyceridemia, 33, 110, 114
hypertrophy, 107, 112, 122
hypothalamus, 33
hypothesis, 89, 102, 138
hypoxia, 13

I
IBD, 152, 153
ID, 36, 205
ideal, 87
identification, 22
identity, 3, 38
IL-8, 31, 90
immune modulation, 152
immune reaction, 156
immune response, 33, 34, 91, 151, 152
immune system, 152, 155, 157
immunity, 154
immunomodulation, 156
improvements, 117
impurities, 71, 165
in vitro, 3, 6, 9, 10, 36, 55, 92, 107, 132
in vivo, viii, 6, 9, 10, 12, 13, 45, 91, 92, 99
incidence, ix, 87, 106, 110, 111, 114, 120, 121, 122,
151, 152
individuals, 34
Indonesia, 70
induction, 92, 107
industrialized countries, viii, 45, 49
industry(s), 18, 20, 38, 40, 68, 71, 72, 73, 74, 76, 77,
160
infancy, 154, 157
infants, x, 38, 57, 75, 77, 78, 96, 98, 110, 145, 146,
147, 148, 149, 150, 151, 152, 153, 154, 155, 156,
157, 158
infection, 34, 35, 39, 148, 152
inflammation, ix, 38, 83, 89, 91, 102, 105, 107, 108,
109, 111, 116, 117, 118, 121, 123, 152, 153, 157
inflammatory bowel disease, 151, 152, 153, 154,
155, 156
inflammatory disease, 102, 152, 153
ingestion, 93
inhibition, 5, 14, 19, 24, 26, 27, 30, 40, 42, 79
inhibitor, 2, 9, 11, 39
initiation, 34, 85, 90
injury, 3, 11, 89, 90, 100, 102
innate immunity, 117

INS, 102
insertion, ix, 25, 27, 125, 128
insulin, viii, ix, 4, 33, 34, 36, 37, 38, 39, 63, 79, 90,
91, 92, 98, 101, 103, 104, 105, 107, 108, 109,
110, 111, 113, 114, 116, 117, 118, 119, 120, 121,
122, 123, 124
insulin resistance, viii, ix, 33, 34, 36, 38, 63, 90, 91,
92, 98, 101, 103, 104, 105, 107, 108, 109, 110,
113, 116, 117, 118, 119, 120, 121, 122
insulin sensitivity, ix, 39, 91, 106, 108, 110, 113,
114, 116, 117, 118, 119, 120, 121, 123, 124
insulin signaling, 92, 107, 108, 116, 117
integrity, 25, 26, 28, 37, 152
interference, 110, 137
interferon, 31
interferon-, 31
intervention, 9, 37, 115, 122, 124, 156
intestinal flora, 153, 156, 158
intestine, 69, 78, 88, 147, 152, 153, 155
intracellular calcium, 5
intravenously, 33
investment, 184
ion channels, 20
ions, 26
IP-10, 108, 117
iron, 131
irrigation, 213, 214, 215, 216
ischemia, vii, 1, 2, 3, 5, 9, 10, 11
isolation, 43, 217
isomers, 131, 141
isozymes, 91
Israel, 145, 154
issues, viii, 45, 187
Italy, 45

K
K+, 3, 4, 5, 10
keratinocytes, 34
kidney, 50
kill, 26, 102
kinetics, 34, 118
KOH, 214

L
labeling, 132
lactation, 54, 58
lactic acid, 151
lactobacillus, 151, 152
Lactobacillus, 23, 38, 78
L-arginine, 2

226

Index

Latin America, 72
lauric acid, viii, 31, 36, 45, 47, 77, 203
LDL, viii, ix, 6, 13, 34, 45, 47, 59, 65, 70, 86, 87, 88,
89, 92, 105, 107, 109, 110, 111, 118, 119
lead, 26, 49, 78, 79, 89, 104, 108, 111, 117, 152,
161, 191, 204
learning, 3, 18
lecithin, 69
leptin, 33, 36, 154
lesions, 152
leukocytes, 90
life cycle, 142
lifetime, 115
light, 75, 196, 197
linoleic acid, 21, 31, 34, 37, 43, 47, 49, 52, 55, 59,
66, 71, 73, 74, 88, 110, 112, 120, 121, 123, 147,
148, 167, 194, 197, 198, 214
lipases, 38, 48, 76, 97
lipemia, 101, 110
lipid dysregulation, 107
lipid metabolism, 41, 61, 83, 119, 128, 139
lipid peroxidation, 90, 102
lipoproteins, 6, 31, 36, 60, 61, 86, 100, 101, 109,
118, 119
liquid chromatography, 37
liquid phase, 71, 168, 178, 191, 194
liquids, 161
Listeria monocytogenes, 42
lithium, 30, 69
liver, viii, 3, 6, 28, 30, 36, 38, 39, 40, 41, 43, 52, 61,
63, 69, 78, 79, 83, 86, 89, 90, 91, 93, 99, 101,
102, 122, 123, 199
liver cells, 70, 79, 123
liver cirrhosis, 122
liver disease, viii, 39, 63, 89, 101, 102
localization, 10, 20, 36, 93, 134
locus, 157
low temperatures, 200, 213
low-density lipoprotein, ix, 13, 105, 109
lower lip, 35, 123
low-grade inflammation, 112, 122
lubricants, 69
lumen, 134
Luo, 43, 117
lymphatic system, 86
lymphoma, 113, 119
lysis, 26

M
machinery, 26, 27
macrophages, 6, 92, 107, 108, 117
magnesium, 98, 156

magnitude, 168, 170, 173, 191


majority, 90, 187, 194, 198
malate dehydrogenase, 79
Malaysia, 70, 71, 73, 94, 95
mammalian cells, 2, 98
mammals, 14, 66, 82, 99
man, 100, 101
management, 121
manipulation, 56
manufacturing, 70, 71, 146
marine fish, 51, 58
mass, 46, 49, 50, 51, 68, 96, 140, 146, 149, 160, 162,
164, 166, 168, 191, 199, 200, 203, 204
mass spectrometry, 96
materials, 26, 28, 165
matrix, ix, 6, 47, 70, 79, 83, 125, 130, 168
maturation process, 151, 215
MB, 118, 119
MCP, 65
measurements, 149, 156, 168, 175
meat, viii, ix, 22, 40, 45, 46, 48, 49, 50, 51, 52, 53,
57, 58, 59, 60, 62, 74, 105, 106, 121
media, 162
medication, 151
Mediterranean, x, 37, 51, 106, 118, 211, 212
Mediterranean countries, 212
mellitus, 111
melon, 204
melting, 24, 71, 73, 78, 96, 146, 147, 161, 164, 165,
168, 170, 178, 179
melting temperature, 78, 161, 165
melts, 68
membrane lipid, vii, 17, 126, 139, 140
membrane permeability, 26
membranes, vii, 1, 2, 20, 25, 26, 27, 30, 37, 42, 128,
132
memory, 3
meta-analysis, 37, 39, 109, 111, 119, 121
Metabolic, 36, 39, 102, 106, 116, 122, 123, 140
metabolic disease, ix, 90, 105, 106, 115
metabolic disorder(s), 34, 120
metabolic pathways, 152
metabolic syndrome, 4, 34, 37, 100, 106, 107, 110,
112, 114, 116, 119, 120, 122
metabolism, viii, ix, 9, 13, 14, 30, 31, 33, 34, 35, 38,
39, 40, 41, 42, 57, 61, 63, 80, 83, 87, 89, 93, 98,
99, 100, 105, 107, 110, 112, 118, 120, 135, 143
metabolites, 11, 18, 28, 84, 118
metabolized, 3, 30, 40
metabolome, 18
metals, 71
methanol, 7, 10, 40, 214
methodology, 98

227

Index
methyl group(s), 18, 68, 132
methyl palmitate, vii, 1, 2, 11, 161, 164, 168, 169,
173, 174, 175, 176, 177, 178, 179, 180, 181, 183,
184, 185, 186, 191, 194, 204
methylation, 41, 214
methylene chloride, 202
Mexico, 73
Miami, 1, 10
mice, 13, 22, 33, 92, 99, 102, 108, 117, 152, 153,
156, 157
microbiota, 78, 152, 154, 158
microorganism(s), 13, 18, 24, 26, 27, 41, 46, 146,
151, 152
Microsoft, 21
microsomes, 28, 61, 136
microstructures, 97
mineralization, 149, 150, 153, 156
Ministry of Education, 115
misuse, 59
mitochondria, vii, 1, 6, 8, 26, 28, 30, 36, 39, 41, 42,
43, 79, 102, 126
mitochondrial damage, 90
mitogen, 91, 92, 103, 104, 107, 118
models, vii, 1, 9, 10, 48, 89
modifications, vii, 17, 66, 130
molar ratios, 189
molecular mass, 24, 160, 161, 162, 163, 164, 168,
172, 173, 174, 177, 187, 191, 193, 194, 198, 199,
203
molecular structure, 48, 68
molecular weight, 164
molecules, 6, 8, 9, 10, 20, 68, 74, 75, 79, 81, 84, 85,
86, 91, 135, 138, 165, 174, 193, 199, 201
monolayer, 134
monomers, 131, 132, 141
monosodium glutamate, 70
monounsaturated fatty acids, 34, 37, 39, 73, 75, 82,
109, 115, 118
Moon, 14, 99
morbidity, 9, 153
morphology, 77
mortality, ix, 9, 59, 62, 105, 106, 110, 112, 120, 121,
122, 153
Moscow, 125
MR, 94, 139
mRNA, 20, 59, 65, 70, 87, 108
mucin, 156, 157
mucosa, 30, 152
mucus, 152, 155
muscles, 49, 50, 57, 107
mutagenesis, 155
mutation, 117
mycobacteria, 41

myocardial infarction, 4, 6, 112, 122


myristic acids, viii, 30, 42, 45, 54

N
Na+, 5
NAD, 8, 65, 79, 84
NADH, 8, 9, 79, 83, 84, 85
National Institutes of Health, 10
National Research Council, 45
natural selection, 151
necrosis, 14, 90
Netherlands, 120
neurons, 9
neuroprotection, 2, 9, 10, 11
neuroprotective agent, vii, 1
neurotransmission, vii, 1
neurotransmitter, 2, 20
neutral, 47, 52, 69, 88, 134, 140, 142
neutral lipids, 52, 134, 142
neutrophils, 107
New Zealand, 10, 73
NH2, 107
nicotinamide, 85
Nigeria, 70
nitric oxide, vii, 2, 11, 12, 17, 90, 111
nitric oxide synthase, 2, 11
nitrogen, 89, 214
NMR, 141
non-esterified PA, vii, 17
norepinephrine, 5, 11
normal development, 131
North Africa, 212
North America, 72
nutraceutical, 36
nutrient(s), 21, 26, 27, 37, 47, 49, 59, 146, 148, 151,
212
nutrition, vii, viii, x, 17, 45, 57, 61, 62, 63, 69, 74,
92, 94, 96, 115, 119, 145, 146, 149, 154, 155,
156, 157, 211
nutritional status, 75

O
obesity, 4, 33, 34, 103, 106, 107, 108, 117, 118, 119,
123
obstruction, 9
octane, 164, 188, 190
OH, 131, 138
oil production, 160, 212
oilseed, 46, 129

228

Index

oleic acid, viii, 7, 13, 21, 30, 34, 35, 36, 42, 45, 47,
48, 52, 53, 54, 55, 58, 61, 66, 71, 72, 73, 74, 75,
76, 78, 88, 96, 108, 110, 120, 123, 132, 138, 147,
148, 160, 194, 197, 198, 203, 212, 213
olive oil, x, xi, 73, 95, 101, 106, 200, 211, 212, 213,
214, 215, 216, 217
omega-3, 12, 13, 61
operating costs, 169
operating range, 162
opportunities, 20
organ(s), 83, 89, 131, 133, 134, 151, 157
organelle(s), 26, 28, 127, 128, 134
organic compounds, 164
organic matter, 138
organic solvents, x, 159, 187, 200, 202, 204
organism, 78, 79
osteoporosis, 149, 155
overweight, 112, 122
ox, 6
oxidation, vii, ix, 8, 9, 13, 14, 17, 20, 30, 31, 32, 33,
34, 38, 39, 42, 70, 71, 83, 84, 85, 86, 90, 99, 100,
105, 107, 108, 111, 117, 123, 126, 130, 132, 134
oxidative stress, 10, 15, 90, 91, 101, 102, 103, 111,
116
oxygen, 7, 9, 24, 90, 101, 177

P
Pacific, 59, 61
palm oil, viii, 34, 46, 47, 48, 52, 55, 56, 57, 58, 59,
60, 61, 63, 68, 70, 71, 72, 77, 78, 87, 88, 92, 94,
95, 97, 100, 101, 106, 110, 120, 133, 146, 160,
197, 200, 202
palmate, 18
palmitic acid methyl ester, vii, 1, 2
Pamirs, 140
pancreas, 14, 111
parallel, 68, 90, 102
parasite, 26
parenchyma, 89, 90
parents, 150
participants, 111
partition, 194, 202
pasta, 97
pathogenesis, 11, 20, 103
pathogens, viii, 17, 25, 26, 27, 28, 30, 36, 40, 151,
152
pathology, 101
pathophysiology, 100
pathways, vii, 17, 20, 26, 30, 35, 74, 91, 92, 101,
103, 104, 108, 128, 132, 134, 135, 137, 150, 153
pattern recognition, 91
perfusion, 6, 13

permeability, 26, 27, 42, 43


permit, 26
peroxidation, 26, 28, 90
peroxynitrite, 11
pH, 21, 26, 68, 147
pharmaceutical, 18, 20, 70, 160
pharmacology, vii, 1
phase transitions, 93
Philadelphia, 98
Philippines, 76
phosphate, 14, 79, 134, 135
phosphatidylcholine, 56, 138
phosphatidylethanolamine, 136
phospholipids, ix, 18, 26, 28, 30, 32, 35, 37, 52, 74,
83, 87, 106, 112, 114, 116, 119, 122, 123, 124,
134, 136, 142, 146
phosphorylation, ix, 28, 30, 36, 41, 104, 106, 108,
110, 117
photosynthetic systems, 215
physical activity, 156
physical interaction, 191
physical properties, viii, 57, 58, 63, 76, 161
physicians, 39
physicochemical properties, 97
Physiological, 39
physiological mechanisms, 150
physiology, 14, 93, 99, 100
physiopathology, 100
pigs, 49, 52, 58, 60, 62
pilot study, 122
pith, 68, 146
plants, vii, 1, 17, 18, 21, 24, 34, 46, 68, 126, 127,
128, 129, 130, 132, 133, 134, 135, 136, 137, 138,
139, 140, 141, 142, 143, 146, 197
plasma membrane, 8, 83, 131
plastid, 126, 127, 128, 129, 130, 135, 136
platelet aggregation, ix, 105, 109, 119
PLP, 65, 70
PM, 119, 120
polar, ix, 75, 125, 129, 133, 141, 142, 198
polarity, 133
pollen, 134, 142
polyesters, 132, 141
polymer(s), ix, 125, 130, 131, 132
polymorphism, 77
polypeptide, 99
polyunsaturated fat, ix, 6, 41, 51, 58, 66, 67, 71, 73,
74, 76, 86, 93, 101, 106, 107, 110, 115, 116, 117,
119, 121, 137, 198
polyunsaturated fatty acids, 73, 74, 76, 93, 101, 107,
115, 116, 137, 198
pools, 127
population, 37, 109, 122, 124

229

Index
Portugal, 37
positive feedback, 19
positive relationship, 111
potassium, 3, 5, 12, 30, 69
potato, 132
poultry, 22, 48, 49, 52, 53, 58
precipitation, 71
premature infant, 148, 154
preparation, 77
preterm infants, 148, 150, 153, 156
prevention, 27, 35, 62, 106, 110, 120, 121, 155
probiotics, 155
producers, 70, 73
progenitor cells, 92, 104
pro-inflammatory, 90
project, 160
prokaryotes, ix, 28, 35, 125
prokaryotic cell, vii, 17, 28
proliferation, 33, 91, 151, 152
propane, x, 159, 161, 164, 177, 179, 180, 181, 182,
183, 184, 185, 186, 187, 191, 199, 200, 201, 204,
205
prostacyclins, 18
prostaglandins, 18
prostate cancer, 123
proteasome, 104
protection, 34, 90, 148, 151, 152, 155, 157
protective role, 153
protein kinase C, 6, 13, 91, 92, 103, 104, 107, 111,
118
protein kinases, 91, 92, 107
protein synthesis, 14, 26, 89
protein-protein interactions, 18
proteins, 18, 20, 26, 27, 28, 30, 40, 41, 66, 82, 83,
86, 88, 93, 99, 118, 134, 149, 150
proteome, 18
pulp, 72, 73, 75, 76, 212
purification, 164
purity, 167, 170, 199, 202

reactive oxygen, 89, 90, 107


receptors, ix, 20, 28, 88, 91, 92, 103, 105, 109, 110
recognition, 91, 103, 155
recommendations, viii, 105, 115
recovery, 101, 199
recruiting, 108
recrystallization, 71
recycling, 191
red blood cells, 83
red wine, 60
redistribution, 76, 92, 150
relaxation, 12, 13
replication, 28
requirements, 70, 86, 115, 119, 146
researchers, 31, 55
residues, 18, 20, 78, 137, 138, 200
resistance, ix, 5, 33, 36, 71, 91, 92, 106, 108, 123
resources, 39
respiration, 30
response, vii, ix, 13, 17, 34, 70, 87, 90, 98, 100, 105,
108, 112, 120, 131, 150, 152, 153, 156
responsiveness, 100
retail, 58, 200
reticulum, 6, 33, 82, 83, 90, 91, 102, 107, 111, 113,
117, 123, 126, 128, 130
retina, 3, 11
retinol, 69, 70, 93
rings, 12
risk(s), viii, ix, 2, 4, 6, 12, 13, 33, 34, 37, 39, 42, 45,
46, 47, 63, 66, 91, 105, 106, 107, 109, 110, 111,
112, 113, 114, 115, 116, 119, 120, 121, 122, 124
risk factors, 4, 37, 110, 112, 115, 116, 122
RNA, vii, 17, 65, 137, 155
rodents, 12, 36
room temperature, 24, 71, 72, 161
root(s), 131, 132, 133
Russia, 125

salmon, 55, 56, 57, 60


salts, 55, 57, 59, 68, 69, 72, 86, 88
saturated fatty acids, viii, 6, 24, 34, 35, 39, 45, 61,
63, 68, 70, 73, 74, 75, 77, 85, 100, 115, 117, 118,
120, 123, 147, 148, 191, 194
saturation, 66, 74, 190, 194, 195, 198
scatter, 168, 187
science, 59, 66
sebum, 36
secretion, 33, 34, 61, 74, 88, 90, 91, 108, 121, 140,
148, 157
sedentary behavior, 116

quality of life, 60

R
rainfall, 213
rape, 77, 187
rape seed, 77, 187
raw materials, 205
reactions, 6, 7, 68, 69, 77, 79, 80, 81, 82, 85, 127,
130, 131, 132, 134, 135

230

Index

seed, 46, 70, 76, 77, 126, 129, 131, 132, 133, 134,
137, 138, 139, 140, 141, 142, 143, 160, 197, 204
seedlings, 142
selectivity, 27, 109, 164, 191, 198, 204
selenium, 75
sensitivity, 4, 31, 34, 114, 122, 124
sequencing, 198
Serbia, 105, 115, 116
serine, 91, 92, 110
serum, viii, 28, 31, 37, 39, 41, 45, 47, 52, 56, 61, 86,
87, 88, 89, 100, 106, 107, 109, 110, 111, 112,
113, 114, 118, 119, 120, 121, 122, 123, 124
serum albumin, 28
serum cholesterol, viii, 31, 41, 45, 47, 52, 61, 86, 88,
100, 112, 114, 121, 122, 124
shape, 51
sheep, 50
shelf life, 71
shellfish, 51, 58
shock, 26
showing, 148, 167, 183, 202
side chain, 20
signal peptide, 155
signal transduction, ix, 40, 105, 153
signaling pathway, 40, 91
signalling, 35
signals, 79
silica, 141
silver, 141
skeletal muscle, ix, 14, 30, 35, 79, 83, 103, 106, 108,
110, 111, 114, 116, 118, 122, 123, 124
skin, 25, 36, 41, 49, 50, 70, 94, 95
small intestine, 83
smooth muscle, 3
smooth muscle cells, 3
sodium, 3, 30, 68, 69, 76, 214
sodium hydroxide, 69
solid matrix, 162, 163, 166, 202, 204
solid phase, 165, 168
solubility, x, 83, 159, 162, 163, 164, 165, 166, 168,
169, 170, 172, 173, 174, 175, 178, 179, 180, 181,
182, 184, 186, 187, 190, 191, 193, 194, 200, 203,
204, 205
solution, 214
solvents, 161, 162, 164, 184, 187, 190, 195, 198,
201, 202, 205
South Africa, 159
South America, 73, 75
soybeans, 57, 203
specialization, 143
species, vii, ix, 1, 3, 21, 22, 27, 38, 41, 48, 50, 51,
55, 58, 60, 72, 74, 75, 76, 89, 90, 97, 101, 107,
125, 126, 128, 129, 134, 138, 139, 140, 142, 152

specifications, 95
Spring, 117
SS, 99, 102, 121, 123, 141
SSS, 66, 70
stability, 35, 58, 60, 70, 76, 92, 151, 215
starvation, 33
state(s), vii, ix, 1, 39, 51, 83, 89, 91, 97, 125, 148,
178, 202
statistics, 14, 212
Stearic acid, viii, 15, 45, 109, 205
sterile, 151
sterols, 73, 200, 201, 202
stimulation, 2, 30, 39, 89
stock, 195
storage, ix, 18, 21, 33, 34, 66, 69, 70, 71, 78, 83, 89,
93, 125, 126, 128, 129, 131, 133, 134, 135, 139,
141, 142, 211
stress, 33, 43, 86, 89, 90, 91, 102, 107, 111, 113,
117, 123, 132
stroke, ix, 2, 5, 6, 9, 10, 14, 15, 105, 111, 112, 116,
122, 123
stroma, 135
structure, vii, viii, x, 1, 3, 9, 24, 34, 35, 48, 61, 63,
75, 77, 97, 100, 120, 129, 134, 145, 148, 151,
153, 154, 155, 178, 205, 217
subarachnoid hemorrhage, 11
suberin, 131, 132, 141
substitutes, 74, 77, 96
substitution, 88, 95
substrate(s), ix, 39, 69, 82, 84, 86, 92, 104, 106, 108,
117, 127, 128, 129, 130, 131, 134, 135, 139
sulfur, 20
Sun, 12, 41, 87, 101, 117, 121, 156
superoxide, vii, 17, 89
supplementation, viii, 12, 31, 45, 54, 55, 58, 122
suppression, 90, 137, 143
surfactant, 160, 161
surplus, 86
survival, 9, 28, 90, 91
susceptibility, 152
swelling, 28, 29, 30, 39, 42, 43
Switzerland, 62, 115
syndrome, 106, 112
synthesis, vii, x, 1, 2, 6, 7, 10, 14, 18, 19, 20, 27, 28,
30, 34, 35, 54, 55, 66, 78, 79, 84, 86, 88, 90, 92,
98, 111, 113, 126, 127, 128, 129, 130, 131, 132,
134, 138, 139, 140, 142, 145, 146, 149

T
T cell(s), 154, 157
target, vii, 1, 26, 70, 86, 153
target organs, 86

231

Index
techniques, 70, 162, 212
technological advances, 40
technology, 66, 73, 205
temperament, 154
temperature, xi, 26, 72, 138, 139, 146, 147, 160, 161,
162, 163, 164, 165, 166, 168, 169, 170, 172, 173,
175, 178, 179, 180, 181, 182, 183, 184, 185, 187,
191, 194, 198, 203, 211, 214
temperature dependence, 173
texture, 74, 77
TGA, 88
Thalassiosira, 23
therapeutic interventions, 9
therapy, 9, 10, 122
thermal analysis, 96
thermal degradation, 161, 162, 200, 202
thermograms, 96
threonine, 91
thrombin, 9
thrombosis, 106
thromboxanes, 18
thrombus, 9
tissue, ix, 22, 33, 39, 48, 52, 53, 56, 61, 78, 83, 90,
99, 105, 107, 112, 114, 123, 131, 133, 134, 138,
142, 150
TLR, 66, 91, 103, 107, 117
TLR4, 92, 108, 117
TNF, 31, 34, 66, 90, 91, 107, 117
TNF-alpha, 117
TNF-, 31, 34, 66, 90, 91
tobacco, 132, 140
tocopherols, 71, 200, 201, 202
total cholesterol, viii, ix, 34, 45, 47, 62, 87, 88, 92,
105, 109
total energy, 87
toxicity, viii, 9, 41, 63, 70, 101, 163
trade, 212
traditions, 74
trafficking, 18, 35
traits, 60
transcription, 14, 88, 90, 91, 117
transcription factors, 14, 90
transesterification, 40, 132
transformation, 37, 97, 137
transforming growth factor, 103
transient ischemic attack, 122
translocation, 34
transmission, 11
transport, 18, 26, 27, 32, 40, 42, 79, 83, 90, 99, 100,
118, 162
transportation, 131
treatment, 9, 10, 51, 92, 100, 101, 116

triacylglycerols, viii, ix, 41, 48, 55, 59, 63, 69, 74,
92, 111, 125, 126, 129, 139, 141, 142, 143, 146,
148, 156
trial, 110, 112, 148, 155, 156
triglycerides, viii, x, 4, 12, 18, 41, 48, 62, 63, 71, 73,
74, 75, 77, 98, 109, 110, 120, 123, 141, 142, 145,
155, 159, 160, 161, 163, 191, 193, 197, 198, 200,
201, 202, 203, 204
tumor, 14, 107, 117, 118
tumor necrosis factor, 107, 117, 118
Turkey, 22, 51
turnover, 33, 39, 149
Tuskegee University, 17
type 1 diabetes, 91
type 2 diabetes, viii, ix, 63, 91, 102, 105, 112, 114,
121, 124

U
U.S. Department of Agriculture, 115
UK, 58
ultrasound, 156
uniform, 56
United States (USA), 9, 67, 73, 93, 94, 98, 140, 143
updating, 217
USDA, 58, 93, 119

V
vacuum, 71, 161, 162
vagina, 39
variables, 123
variant angina, 13
variations, 73, 128, 194, 215
varieties, 73, 95, 212, 213, 214, 215, 216
vascular tonicity, vii, 1
vasculature, 9
vasoconstriction, 5, 6, 13
vasodilation, vii, 1, 2, 3, 4, 5, 6, 9, 10, 11, 12, 13
vasodilator, 2, 3, 10, 11, 12
vasomotor, 6
vasopressin, 5
vasospasm, 6, 13
VAT, 3
vegetable oil, 46, 55, 70, 73, 74, 75, 77, 87, 97, 116,
146, 148, 152, 212
vegetables, 35, 38, 133, 142
Vereshchagin, 133, 140, 141, 142, 143
victims, 9
virus infection, 37
viruses, 21
viscosity, 73

232

Index

vitamin A, 69, 70, 92, 93, 94


vitamin E, 57, 71, 73, 94
vitamins, 18, 75, 86
VLDL, 66, 86, 88, 109, 118, 120
volatile organic compounds, 187
vulnerability, 41

well-being, x, 78, 145, 150


workers, 175, 179, 191, 201, 203
World Health Organization (WHO), 47, 62, 93, 109,
115
World War I, 68
worldwide, 70, 212
WWW, 97

W
Y
Washington, 94, 115, 141
waste, 212, 213, 215
waste water, 213, 215
wastewater, 216
water, 21, 55, 67, 68, 69, 72, 81, 84, 86, 123, 131,
188, 190, 213

yeast, 21, 35, 41, 134, 136, 140, 143


yield, x, 28, 54, 57, 73, 82, 83, 85, 86, 100, 145, 197,
198, 202, 203, 204, 214
yolk, 51, 56, 59

You might also like