You are on page 1of 63

PAMELA L. TUMA and ANN L.

HUBBARD
Physiol Rev 83:871-932, 2003. doi:10.1152/physrev.00001.2003 You might find this additional information useful... This article cites 629 articles, 295 of which you can access free at: http://physrev.physiology.org/cgi/content/full/83/3/871#BIBL This article has been cited by 36 other HighWire hosted articles, the first 5 are: Intersectin-2L Regulates Caveola Endocytosis Secondary to Cdc42-mediated Actin Polymerization I. K. Klein, D. N. Predescu, T. Sharma, I. Knezevic, A. B. Malik and S. Predescu J. Biol. Chem., September 18, 2009; 284 (38): 25953-25961. [Abstract] [Full Text] [PDF] Axonal Targeting of Trk Receptors via Transcytosis Regulates Sensitivity to Neurotrophin Responses M. Ascano, A. Richmond, P. Borden and R. Kuruvilla J. Neurosci., September 16, 2009; 29 (37): 11674-11685. [Abstract] [Full Text] [PDF]
Downloaded from physrev.physiology.org on November 4, 2009

Liver Sinusoidal Endothelial Cells Are a Site of Murine Cytomegalovirus Latency and Reactivation C. K. Seckert, A. Renzaho, H.-M. Tervo, C. Krause, P. Deegen, B. Kuhnapfel, M. J. Reddehase and N. K. A. Grzimek J. Virol., September 1, 2009; 83 (17): 8869-8884. [Abstract] [Full Text] [PDF] Basolateral Internalization of GPI-anchored Proteins Occurs via a Clathrin-independent Flotillin-dependent Pathway in Polarized Hepatic Cells T. Ait-Slimane, R. Galmes, G. Trugnan and M. Maurice Mol. Biol. Cell, September 1, 2009; 20 (17): 3792-3800. [Abstract] [Full Text] [PDF] Polarized epithelial cells secrete matriptase as a consequence of zymogen activation and HAI-1-mediated inhibition J.-K. Wang, M.-S. Lee, I-C. Tseng, F.-P. Chou, Y.-W. Chen, A. Fulton, H.-S. Lee, C.-J. Chen, M. D. Johnson and C.-Y. Lin Am J Physiol Cell Physiol, August 1, 2009; 297 (2): C459-C470. [Abstract] [Full Text] [PDF] Medline items on this article's topics can be found at http://highwire.stanford.edu/lists/artbytopic.dtl on the following topics: Physiology .. IgA Cell Biology .. Endothelial Cells Cell Biology .. Transcytosis Physiology .. Microvasculature Updated information and services including high-resolution figures, can be found at: http://physrev.physiology.org/cgi/content/full/83/3/871 Additional material and information about Physiological Reviews can be found at: http://www.the-aps.org/publications/prv

This information is current as of November 4, 2009 .

Physiological Reviews provides state of the art coverage of timely issues in the physiological and biomedical sciences. It is published quarterly in January, April, July, and October by the American Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright 2005 by the American Physiological Society. ISSN: 0031-9333, ESSN: 1522-1210. Visit our website at http://www.the-aps.org/.

Physiol Rev 83: 871932, 2003; 10.1152/physrev.00001.2003.

Transcytosis: Crossing Cellular Barriers


PAMELA L. TUMA AND ANN L. HUBBARD Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland

I. Introduction II. Documented Transcytosis In Vivo A. Transcytosis in the vasculature B. Transcytosis in the brain C. Immunological protection and transcytosis D. Role for transcytosis in the homeostasis of micronutrients E. Additional transcytosis systems F. Role of transcytosis in plasma membrane biogenesis in vivo III. In Vitro Cell Models of Transcytosis A. What constitutes a good transcytotic cell model? B. Microvascular endothelial cell models C. Epithelial cell models D. Transcytosis outside of the epithelial world IV. More About Two Different Transcytosis Systems A. Caveolae-mediated transcytosis B. Clathrin-mediated transcytosis V. Mechanisms and Molecules Regulating Transcytosis A. Targeting machinery B. Cytoskeleton C. Lipids and transcytosis D. Perturbations of transcytosis E. Transcytosis versus direct PM delivery VI. Conclusion

871 872 873 876 879 881 885 885 886 886 892 893 895 896 896 898 902 902 909 911 913 916 917

Downloaded from physrev.physiology.org on November 4, 2009

Tuma, Pamela L., and Ann L. Hubbard. Transcytosis: Crossing Cellular Barriers. Physiol Rev 83: 871932, 2003; 10.1152/physrev.00001.2003.Transcytosis, the vesicular transport of macromolecules from one side of a cell to the other, is a strategy used by multicellular organisms to selectively move material between two environments without altering the unique compositions of those environments. In this review, we summarize our knowledge of the different cell types using transcytosis in vivo, the variety of cargo moved, and the diverse pathways for delivering that cargo. We evaluate in vitro models that are currently being used to study transcytosis. Caveolae-mediated transcytosis by endothelial cells that line the microvasculature and carry circulating plasma proteins to the interstitium is explained in more detail, as is clathrin-mediated transcytosis of IgA by epithelial cells of the digestive tract. The molecular basis of vesicle trafc is discussed, with emphasis on the gaps and uncertainties in our understanding of the molecules and mechanisms that regulate transcytosis. In our view there is still much to be learned about this fundamental process.

I. INTRODUCTION At its simplest, transcytosis is the transport of macromolecular cargo from one side of a cell to the other within a membrane-bounded carrier(s). It is a strategy used by multicellular organisms to selectively move material between two different environments while maintaining the distinct compositions of those environments. Cells have other strategies not involving membrane vesiwww.prv.org

cles to selectively move smaller cargo (ions and small solutes) across cellular barriers. Paracellular transport, the movement between adjacent cells, is accomplished by regulation of tight junction permeability, and transcellular transport, the movement of ions and small molecules through a cell, is accomplished by the differential distribution of membrane transporters/carriers on opposite sides of a cell. Together, these three processes contribute to the success of multicellular organisms.
871

0031-9333/03 $15.00 Copyright 2003 the American Physiological Society

872

PAMELA L. TUMA AND ANN L. HUBBARD

Historically, the existence of transcytosis was rst postulated in the 1950s by Palade in his studies of capillary permeability (426). He described a prominent population of small vesicles, many of which were in continuity with the plasma membrane, and hypothesized that these vesicles were the morphological equivalent of the large pore predicted by the physiologist Pappenheimer to explain the high permeability of blood microvessels to macromolecules (428). N. Simionescu was the rst to coin the term transcytosis to describe the vectorial transfer of macromolecular cargo within the plasmalemmal vesicles from the circulation across capillary endothelial cells to the interstitium of tissues (538). During this same period, another type of transcytosis was being discovered. Immunologists comparing the different types of immunoglobulins found in various secretions (e.g., serum, milk, saliva, and the intestinal lumen) speculated that the form of IgA found in external secretions (called secretory IgA, due to the presence of an additional protein component) was selectively transported across the epithelial cell barrier (577, 578). The pathway and origin of the component acquired during transport were actively investigated, and in 1980 secretory component (SC) in secretory IgA was identied as the ectoplasmic domain of the intestinal epithelial cell membrane receptor that binds dimeric IgA and transports it through multiple intracellular compartments to the opposite side of the cell (391, 423). These two historic transcytotic systems are still actively investigated today. We now know that transcytosis is a widespread transport process; a variety of cell types use it, different carriers and mechanisms have evolved to carry it out, and the cargo moved by it is diverse. Cell types: we are most familiar with transcytosis as it is expressed in epithelial tissues, which form cellular barriers between two environments. In this polarized cell type, net movement of material can be in either direction, apical to basolateral or the reverse, depending on the cargo and particular cellular context of the process. However, transcytosis is not restricted to only epithelial cells. Reports of cultured osteoclasts (398, 490) and neurons (221) carrying vesicular cargo between two environments indicate that the strategy of vesicular transcytosis has been used elsewhere. Mechanisms: in intestinal cells transcytosis is a branch of the endocytic pathway, with cargo being internalized via receptor-mediated (i.e., clathrin-coated) mechanisms and progressively sorted away from internalized material destined for other cellular destinations. However, transendothelial transport in blood capillaries does not conform to this scenario, since different carriers and a more direct route are used to cross the cell. Such differences illustrate that multiple transcytotic mechanisms have evolved that depend on the particular cellular context. Furthermore, they illustrate that cargo in the transcytotic pathways seems able to avoid degradation in
Physiol Rev VOL

lysosomes. How? Cargo: the nature of the transcytotic cargo also varies. Although today we might think of transcytosis as a selective process, the originally dened system, endothelial cells of the microvasculature, moves macromolecular cargo rather nonselectively within the uid phase of the transport vesicle or by adsorption to the vesicle membrane. Furthermore, transcytotic cargo is not limited to macromolecules. Several vitamins and ions utilize endocytic mechanisms and vesicular carriers as part of their transcellular sojourn. This brings up another unsolved mystery, that of a cell transcytosing particular cargo for use by other cells but also using some of it for its own metabolism. How is such apportionment made? A major goal of this review is to summarize the widespread occurrence of transcytosis and focus on its many variations. First, we present documented examples of in vivo transcytosis in mammals, using the expanded denition given above. Next, we assess the status of in vitro cell models currently used to study the different types of transcytosis. We then review in more depth the two best-studied transcytosis systems, transendothelial transport of circulating macromolecules and transcytosis of IgA in polarized epithelial cells, focusing on the similarities and differences of their pathways and carriers. Finally, we present current information about the molecular mechanisms and regulation of transcytosis. Throughout, we identify gaps in our present understanding of this process, with the hope that interested researchers will ll in those gaps with insightful experiments and denitive answers. II. DOCUMENTED TRANSCYTOSIS IN VIVO Table 1 documents that transcytosis is widespread. As expected, epithelial cells forming barriers between the outside world and the interstitium or between the internal world (circulation) and the interstitium are the major cells participating in transcytosis. However, the question of whether transcytosis occurs in all adult epithelia (e.g., kidney and skin) is open. While proximal tubule cells are endocytically active, only micronutrients seem to be transcytosed by them in vivo. Other segments of the nephron, e.g., the collecting tubule, are more difcult to assess. Transcytosis certainly occurs in the most obvious fetal organs, the yolk sac and placenta, and it probably operates elsewhere in the developing fetus. Further examination of Table 1 reveals that the transport of iron, vitamin B12, and the immunoglobulins IgA and IgG occurs in several organs. However, the routes and fates of the molecules are not always the same. Curiously, the routes and mechanisms by which circulating hormones gain access to their target tissues have not been extensively explored (181). Finally, although not yet examined in all polarized cells, the biogenesis of apical plasma membrane
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS

873

proteins is an example of endogenous molecules using transcytosis to attain their destination. A. Transcytosis in the Vasculature The most extensive exchange in vivo is that of plasma constituents across the endothelium that lines the inner surface of the blood vasculature. Of the three types of endothelium, continuous, fenestrated, and discontinuous (sinusoidal), only the rst two form selective cellular barriers to the passage of macromolecules between the circulation and the underlying interstitium. All continuous and fenestrated endothelia throughout the vascular system are capable of the rapid and extensive bidirectional exchange of small and large molecules, but those of the capillaries and postcapillary venules are the major players in this activity (535, 536, 539). These two parts of the vascular tree constitute what is often called the microvascular exchange system, whose surface area is enormous (600 m2) (Fig. 1A). While fenestrated endothelia are more permeable to small solutes and water than are continuous endothelia, their relative permeability to macromolecules, and hence participation in transcytosis, is controversial (534). Although obvious, it is nonetheless important to state that transcytosis is but one of many important functions carried out by vascular endothelial cells, which are dynamic and capable of rapid responses to local changes in the environment. 1. Structural features of continuous endothelium The simple, squamous epithelial cells of continuous endothelium are quite distinctive morphologically (Fig. 1B). They are remarkably thin (0.2 0.5 m) in regions not including nuclei. A dening feature of these and all epithelial cells is a basement membrane that underlies their basal surface (Fig. 1C). It is made collaboratively by the endothelial and underlying interstitial cells. The most prominent intracellular feature is a population of smoothsurfaced vesicles of 50 70 nm diameter, some of which are in continuity with the plasma membrane facing the circulation (the apical or luminal surface), others in continuity with the opposite surface (the basolateral or abluminal surface), and still others apparently free in the cytoplasm (Fig. 1C). These vesicles, which have an 35nm-diameter opening with a thin diaphragm across it, were originally termed plasmalemma vesicles but are now called caveolae (small caves) because of their characteristic ask shape (Fig. 1D) (10). In continuous endothelial cells, the frequency of caveolae varies widely depending on the organ. For example, in endothelium of skeletal muscle (the diaphragm), the estimate is 1,200/m3, whereas in pulmonary capillaries it is only 130/m3 (539). This variation does not correlate with permeability, suggesting other functions for caveolae. Caveolae are also
Physiol Rev VOL

found in other cell types where their functions and compositions are actively being investigated (reviewed in Refs. 9, 546). An important feature of endothelial cells is their tight junctions, which represent a barrier to paracellular diffusion (219). While there is good experimental evidence that the permeability of endothelial cell tight junctions changes depending on local conditions (336), the molecular basis has yet to be elucidated. The discovery of a large family of tight junction membrane proteins, the claudins, and their capacity to form heteroligomeric complexes with distinct permeability properties (580), will undoubtedly provide insights into the dynamic regulation of tight junction permeability in capillaries. Important to an understanding of transcytosis in endothelial cells is the endocytic system, including clathrincoated vesicles, endosomes, and lysosomes. These organelles are present in all capillary endothelium but are not abundant, and they are usually located in the thicker, perinuclear regions of cells. The endocytic system is clearly functional, as attested by the delivery of modied albumins and oxidized low-density lipoproteins (LDLs) to lysosomes (296, 506). But how do these cells distinguish between cargo destined for transcytosis versus that for degradation? The simplest explanation is that different cargoes use different receptors that are localized to different entry sites in the plasma membrane (PM). However, how do endothelial cells themselves utilize the same cargo that they transport for use by other cells; that is, how is the apportionment of cargo for self versus others regulated? As we shall see in section III, at least one cargo molecule (e.g., native LDL) may use different entry ports (i.e., caveolae versus clathrin-coated vesicles), offering the interesting possibility that the point of entry determines the subsequent fate of a particular internalized cargo molecule. 2. Microvascular permeability and transcytosis Microvessels are approximately two orders of magnitude more permeable than other epithelia, making them leaky to the passage of circulating proteins into the interstitium. Most macromolecules move across capillary endothelium by bulk-phase not receptor-mediated mechanisms. Nonetheless, there is selectivity to the process, with the size and charge of cargo being important factors. Furthermore, although transport is bidirectional, the concentration gradient extending from the blood (apical side) to the interstitium (basal side) dictates that the bulk of transport is in an apical-to-basolateral direction. Finally, different continuous capillary beds have distinctive permselectivities, as evidenced by the varied compositions of the lymph draining from them. The basis for high capillary permeability has been a
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

874
TABLE

PAMELA L. TUMA AND ANN L. HUBBARD

1. Documented in vivo transcytosis


Cell Type Continuous capillary endothelium Cargo Molecules 1.7 nm Albumin Direction A-BL and BL-A A-BL and BL-A Receptor/Carrier Fluid phase in caveolae gp60 in caveolae See text Albumin plays multiple roles in caveolae-mediated transcytosis throughout the vascular tree Comments Reference No. See text 183, 378, 503, 602

Organ System Heart, lung, skeletal muscle, adipose tissue

Orosomucoid IgG

A-BL and BL-A A-BL and BL-A

Unidentied in caveolae FcRn in unidentied carrier Fluid-phase in caveolae Chorionic gonadotropin receptor in CCV Nonspecic in caveolae Insulin receptor via unidentied carrier LDL receptor via unidentied carrier Tf receptor via CCV Tf receptor via unidentied carrier FcRn ? Both phagocytic and clathrin-mediated uptake

456 Contributes to IgG homeostasis and prolonged half-life of IgG in body Not via LDL receptor Target is Leydig cell Not via Tf receptor Rabbit, rhesus monkey, binding to isolated human brain capillaries In vitro study, but LDL receptor present in vivo Debate over Fe Tf Apo-form preferred over holo Efux from brain Degradation in lysosomes is predominant fate Lack of thick glycocalyx thought to permit entry; 1-integrin on apical PM mediates entry; transcytosis thought to be through prelysosome then exocytosis Multiple intermediate compartments in pathway No intracellular intermediates identied 47

Testis

Arterial endothelium Continuous capillary endothelium (has markers of brain EC) Cerebral endothelium (tight continuous)

LDL cholesterol Gonadotrophin Transferrin (Tf)-iron Insulin LDL Iron Tf IgG Membrane associated Antigens, pathogens

A-BL A-BL A-BL A-BL A-BL A-BL BL-A BL-A very little ABL; no BL-A A-BL

600 181, 182 236 133, 431 112 104, 476 635 501, 634 590 259, 400, 402, 404, 492

Brain

Downloaded from physrev.physiology.org on November 4, 2009

Choroid plexus Adult intestine M cells in Peyers patches of ileum

Absorptive enterocytes

dIgA Newly synthesized apical PM proteins Vitamin B12 dIgA Newly synthesized apical PM proteins Ceruloplasmin Vitamin B12 Vitamin D

BL-A BL-A

pIgA receptor via CCV Unknown

64 148, 355

Liver

Absorptive enterocytes in terminal ileum Hepatocytes Hepatocytes

A-BL BL-A BL-A

Cubilin/megalin via CCV pIgA-receptor via CCV Unknown

See Table 2 for molecular players Multiple intermediate compartments in pathway Entry mechanisms unknown, intermediate compartments may be same as those of pIgA Postulated to be desialylated in transit to hepatocytes Degradation of TCII in lysosomes; unknown storage site for B12 Degradation of protein carrier in lysosomes; fate of vitamin D? Same fate as above

128, 129, 515, 519 238, 468 32, 499

Sinusoidal endothelium Kidney Proximal tubule cells

A-BL A-BL A-BL

Unknown TCII; megalin-mediated in CCV D binding protein; megalin-mediated in CCV Retinol binding protein; megalin-mediated in CCV FcRn via CCV

570 311 413

Vitamin A

A-BL

347

Neonatal intestine Yolk sac Placenta

Enterocytes

Maternal IgG

A-BL

Maternal B12 Syncytiotrophoblasts Maternal IgG Iron

A-BL A-BL A-BL

Cubilin?/megalin/TCII FcRn Maternal Tf receptor via unidentied carrier

Subsequent movement into apical endosomes then fusion with lateral membrane Identication of cubilin as target of teratogenic antibodies Apical location of receptor Apical location of receptor and HFE receptor modulator; ferreportin and endogenous copper oxidase believed to facilitate iron release at basolateral PM

1, 245

450, 516 543 107, 172, 432, 433

Fetal capillary endothelial cells

Iron, maternal IgG

BL-A

Tf receptor and FcRn via unidentied carriers

14

Physiol Rev VOL

83 JULY 2003

www.prv.org

TRANSCYTOSIS
TABLE

875

1Continued
Cell Type Trachea Upper airways Bronchial epithelium Cargo HRP, ferritin IgA Albumin Bioactive-Fc fusion protein Albumin dIgA Iron IgG Direction A-BL BL-A A-BL A-BL Receptor/Carrier Unknown IgA-receptor via CCV Unknown FcRn Comments Presence in large endosomes; degradation also? A-BL at low levels may be entry point for pathogens Degraded fragments released at BL (Ussing chamber) Transport of intact protein determined by bioassay Question of which alveolar cell type involved Presumed to be same as in intestine and liver Transferrin synthesized by lactating gland; pathway of iron unknown Neonatal rodent only Separate route for Thy-T3/T4 (lysosomes) Reference No. 469 273 269 553

Organ System Lung

Perfused rat lung Mammary gland Alveolar epithelium

A-BL BL-A BL-A BL-A A-BL

gp60 in caveolae pIgA-receptor Tf receptor via unidentied carrier Not identied; not FcRn Megalin-mediated endocytosis via CCV

267 497 310 245 352

Thyroid

Thyroid epithelial cells

Thyroglobulin

Downloaded from physrev.physiology.org on November 4, 2009

A, apical; BL, basolateral; HFE, hemachromatosis gene product; PM, plasma membrane; Tf, transferrin; TCII, transcobalamin II; CCV, clathrin-coated vesicle; Thy, thyroglobulin; T3/T4, thyroxines; pIgA, polymeric IgA; FcRn, IgG receptor; LDL, low-density lipoprotein.

FIG. 1. The ultrastructure of a capillary network, an endothelial cell, its membrane, and caveolae. A: the vascular casts of the forestomach are shown in this scanning electron micrograph. The submucosal vessels are seen under the two-dimensional mucosal capillary network. [From Imada et al. (254), copyright 1987 Springer-Verlag.] B: this transmission electron micrograph is representative of the ultrastructure of an endothelial cell if the capillaries indicated in A were viewed in cross section. [From Bolender (40) by copyright permission of The Rockefeller University Press.] C: a higher magnication of the indicated region of the endothelial cell in B. The caveolae attached to both the luminal and basal plasma membrane (PM) domains are indicated. [From Fawcett (147), with permission from Journal of Histochemistry & Cytochemistry.] D: a higher magnication of the indicated caveolae in C. Caveolae (vesicles; v) open to the blood or tissue fronts or that appear to be completely closed are indicated. [From Bruns and Palade (65) by copyright permission of The Rockefeller University Press.]

Physiol Rev VOL

83 JULY 2003

www.prv.org

876

PAMELA L. TUMA AND ANN L. HUBBARD

controversy between physiologists and morphologists for over 50 years. Numerous reviews documenting the history, experimental details, and different interpretations have appeared in this and other journals (377, 467, 539, 571, 608, 614). Because the controversy centers on whether tight junctions or caveolae serve as the major (only) conduit for transported cargo, we will briey recap this story. On the basis of experiments in which he compared the compositions of the blood and lymph in the hindleg muscle of cats injected with various size tracers, Pappenheimer et al. (428) postulated in 1951 that plasma components (ions, small solutes, and proteins) were transported through two types of rigid pores: a small, 3- to 5-nmdiameter pore present at a frequency of 100/m3 and a large, 20- to 40-nm-diameter pore present at 1% the frequency of the small ones (428). When the endothelium was seen at the ultrastructural level, no structures corresponding to the postulated pores were found. Instead, caveolae were observed, leading Bruns and Palade (65, 66) to suggest that they performed the function ascribed to the rigid pores. In the early years of this controversy, differences in the cell systems, approaches, and tracers used by researchers in the two camps often yielded conicting results with differences in interpretations. However, both sides progressively rened their experimental approaches and have arrived at an apparent consensus: caveolae do play a role in transport across endothelia, either as fused channels (physiologists) or bonade transport vesicles (cell biologists). Our position is that caveolae contribute to the high permeability of continuous endothelium. However, because their number exceeds the number of functional pores predicted by Pappenheimers results, there must be other functions for caveolae. In fact, they have been proposed to harbor signal transduction components in both active and inactive states (9, 546). The recent reports of mice genetically engineered to lack the protein caveolin-1, a major component of caveolae, are particularly relevant and are discussed in section IV. Although the controversy has focused on bulk-phase transcytosis, receptor-mediated transcytosis of specic macromolecules also takes place across the endothelia of the microvasculature (Table 1). Albumin and orosomucoid are both transcytosed in competable, saturable, and temperature-dependent fashions, and caveolae mediate their transport. A putative receptor for albumin of 60 kDa that is present only on continuous capillary endothelia has been identied by several groups (Table 1), but it has not yet been cloned and sequenced. The transcytosis of IgG across continuous endothelial cells is particularly interesting, in light of the expression of the neonatal Fc receptor (FcRn) by these cells (47) and the receptors role in maintaining high serum IgG in the adult (Table 1). Does the receptor work in both the apical-to-basal and reverse
Physiol Rev VOL

directions? Are clathrin-coated vesicles used, as they are to carry maternal IgG from the gut to the interstitium (apical to basolateral) in neonatal rodents (Table 1)? Is excess IgG degraded when the endothelial FcRn receptor is saturated with its ligand, absent, or dysfunctional? If so, how? This area of endothelial cell biology deserves further study, because it might reveal the mechanism(s) used to selectively deliver a ligand (IgG) from the interstitum back to the circulation (basolateral to apical). We will return to several of these issues below. Finally, one polypeptide hormone, human luteinizing hormone/chorionic gonadotrophin (hLH/CG), is reportedly transcytosed via clathrin-coated pits and vesicles across the continuous endothelium of the testis (Table 1). The transport is apparently mediated by the same receptor present on Leydig cells, the target of the hormone in the testis. This system deserves further study, because it is one of two documented examples in which clathrin-coated vesicles of an endothelium transcytose cargo; the other is brain endothelia and transferrin-Fe (see sect. IIB1C). B. Transcytosis in the Brain Since the 19th century dye experiments of Ehrlich, the brain has been known as a privileged organ where access is tightly regulated so that the environment remains chemically stable. The brains uid is different from either the blood or noncerebral tissue. The two principal gatekeepers of the brain are the cerebral capillary endothelium and the epithelial cells of the choroid plexus (Fig. 2A). These cellular barriers are specialized for the passage of different nutrients from the blood (132, 552). The capillaries move nutrients that are required rapidly and in large quantities, such as glucose and amino acids. These small molecules are transported by membrane carriers using facilitated diffusion. The choroid plexus supplies nutrients that are required less acutely and in lower quantities. These are folate and other vitamins, ascorbate, and deoxyribonucleotides. Their transport requires energy since the blood concentrations of these nutrients are extremely low. Of relevance to this review is experimental evidence that transcytosis of a limited set of macromolecules occurs across brain capillaries from blood to the interstitium (the blood-brain barrier) but does not occur across the epithelial cells of the choroid plexus into the cerebrospinal uid [the blood-cerebrospinal uid (CSF) barrier]. 1. Cerebral capillaries Compared with other organs, the abundance of capillary endothelium in brain is very high (35). At the same time, permeability is about two orders of magnitude lower than that of endothelia in peripheral organs, giving rise to the designation of this endothelium as tight continuous (201).
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS

877

Downloaded from physrev.physiology.org on November 4, 2009

FIG. 2. The barriers of the blood-brain (A) and placenta (B). A: in a, a diagram indicating the location of the choroid plexus in the human brain is shown. In b, the relationship between the blood, brain, and cerebrospinal uid is shown. The dashed line follows two typical open pathways connecting the ventricular cerebrospinal uid with the basement membrane of parenchymal blood vessels and with the basement membrane of the surface of the brain. A, astrocytic process; C, choroid plexus epithelium; Cs, choroid plexus stroma; E, endothelium of parenchymal vessel; EC, endothelium of choroid plexus vessel; Ep, ependyma, GJ, gap junction; N, neuron; SCSF, cerebrospinal uid of the subarachnoid space; TJ, tight junction; VCSF cerebrospinal uid of ventricles. [From Brightman and Reese (60) by copyright permission of The Rockefeller University Press.] B: a low magnication diagram of the chorionic villus is shown (a). The arterio-capillary-venous network (network) is indicated at the top. [From Moe (380).] In b, a cross section of a full-term villus is shown. The placental membrane separates the maternal blood from the fetal blood. At the end of pregnancy, this membrane becomes very thin. [From Moe (380).] In c, a more detailed version of the chorionic villus is shown. The microvillar surfaces (MV) and basal membranes (basal) are indicated. CT, cytotrophoblast; FV, fetal vessel; SK, syncytial knot; ST syncytiotrophoblast. [From Moore and Persaud (383) by copyright permission of Saunders.]

Two features of brain endothelia are different from endothelia in the periphery. Brain endothelial cells have the lowest frequency of caveolae (100/m3), and the character of their tight junctions is inuenced by underlying astrocytes through the actions of soluble factors, including cytokines (111, 263). Claudins 1 and 5 as well as occludin appear to be relevant players in providing a particularly tight junction
Physiol Rev VOL

(229, 289, 319, 385). Very little macromolecular cargo is transcytosed across the cerebral capillary endothelium. The three best-studied ligands are insulin, LDL-cholesterol, and iron; questions and controversy surround each. A) INSULIN. The nding that insulin-sensitive glucose transporters (GLUT 4) were present in the brain (312, 369) led to the search for insulin receptors on endothewww.prv.org

83 JULY 2003

878

PAMELA L. TUMA AND ANN L. HUBBARD

lium and hormonal effects on the cells. Although receptors were found (431, 592), the current status of insulins transport by brain capillaries is not resolved. B) LDL. Cells in the brain require cholesterol, which is synthesized endogenously (127), but can also be provided by the transcytosis of plasma LDL intact across brain endothelium (112). There is good evidence for the presence of LDL receptors on the luminal PM of cerebral endothelium (376). Such expression is unusual, since cells that are constantly exposed to the high LDL levels in plasma normally downregulate their LDL receptors. Experimental evidence from in vitro studies indicates that cholesterol levels in the underlying astrocytes play a role in regulating LDL receptor expression levels in the overlying endothelial cells (111). The puzzle here is how a cell distinguishes between LDL for its own needs and LDL for use by cells behind the permeability barrier it forms. In the periphery, this seems to have been solved by receptormediated endocytosis via clathrin-coated pits/vesicles for internal use versus uid-phase (non-receptor-mediated) transcytosis via caveolae for use by interstitial cells (600). However, in the brain, there is virtually no uid-phase (i.e., nonselective) transcytosis. Thus it will be important to localize LDL receptors in brain endothelium at the ultrastructural level; are they in caveolae or clathrincoated pits? Another important issue is how transcytosed cholesterol is presented at the abluminal surface of endothelial cells, since apoprotein B, which is the apoproteincarrying cholesterol in the circulation, is not present in CSF. Apoproteins A1 and E are the principal cholesterolcarrying molecules in the brain (127, 629). Underlying pericytes of the brain endothelium have been characterized as phagocytic; perhaps they participate in the degradation of apoprotein B and release of cholesterol into the CSF. Cholesterol dynamics in the brain have been reviewed recently (624). C) IRON. Iron is also transported across the blood-brain barrier, but there is conicting data as to whether it is delivered with or without transferrin (Tf), the principal iron-carrying protein of plasma (52). (Iron and Tf are discussed in more detail in sect. IID2.) While several in vivo studies have reported that injected 125I-Tf does not accumulate to the same extent as 59Fe administered similarly (104), others report equivalent accumulations (152, 635). Tf receptors are denitely present on brain endothelium (248), and Tf is internalized by brain endothelium in vivo via clathrin-coated vesicles (476), leading some to speculate that plasma Tf may be carrying Fe across and then recycling back unloaded (52, 384, 429, 430, 545, 591, 635). Such a scenario would require a milieu on the basal side of sufciently low pH to effect irons release. Because the pH of the underlying interstitium in brain is not known to be acidic, there must be novel dissociation mechanisms not yet discovered. Whatever the mechanism, iron is not free in the brain interstitium but is
Physiol Rev VOL

complexed to Tf. Again, there is conicting data about the source of this protein. Tf is synthesized and secreted by the epithelial cells of the choroid plexus (384). However, hypotransferrinemic mice have been shown to accumulate substantial amounts of intraperitoneally administered human transferrin intracranially, indicating that the endogenous source could also be derived from the serum (126). The brain endothelial insulin and Tf-Fe transport systems have received attention from researchers working on therapeutic drug delivery systems (96, 161, 165). An anti-Tf-receptor antibody, OX26, is transcytosed into the brain mass, but the amounts are extremely low, 1% of the antibody injected. Although this amount may be sufcient for drug delivery, it is not denitive evidence for quantitative transcytosis of the receptor along with its cargo. Nonetheless, this approach is being combined with toxins that bind to specic claudins and transiently open tight junctions, to deliver macromolecular drugs (100). D) IMMUNOGLOBULIN G. It turns out that brain endothelial cells express the FcRn and transport intracranially delivered IgG out of the brain in a receptor-mediated fashion (Table 1). The question is how circulating IgG initially crosses into the brain. As for the peripheral endothelium, the vesicular carrier and molecular mechanisms responsible for IgG transport are as yet unknown. 2. Choroid plexus The choroid plexus is composed of a highly convoluted sheet of cuboidal ependymal epithelium that sits on a closely apposed basal lamina. Both morphological and biochemical tracers have provided good experimental evidence that the apical tight junctions of these epithelial cells are the blood-CSF barrier in the choroid plexus (see Fig. 2A). To date, no obvious ultrastructural or molecular features distinguish these junctions from neighboring ependymal cells, but we would predict that specic claudins are responsible for this difference (323). Interestingly, the basal lamina may act as an inducer of the tight junction specializations that make this cell type highly impermeable to macromolecules (reviewed in Ref. 590). The epithelial cells make much of the CSF that nourishes and cushions the brain. The protein content of CSF (25 mg/100 ml) is low relative to that of plasma (6,500 mg/100 ml), and the composition is different. Transthyretin, which binds thyroxine, and Tf are made and secreted by the epithelial cells, while apoproteins E and A1, which are present in lipoprotein particles in the CSF, are made by astrocytes. The presence of these proteins in CSF raises the obvious questions of where and how their ligands, presumably from the blood, reach them? Furthermore, are the components of CSF uid functionally accessible to brain tissue or part of a drainage system much
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS

879

like the lymphatics? To our knowledge, there are no denitive answers. What about transcytosis in the choroid plexus? Interestingly, although endocytosis is robust at the basal surface of the epithelial cells, transcytosis across to the apical environment is minimal to nonexistent; rather, virtually all tracers internalized from the basal side end up in lysosomes (590). The endocytic activity may reect the high permeability of the fenestrated capillaries that supply the choroid plexus and hence the abundance of plasma proteins bathing the basal side of these cells. Van Deurs (589) examined transcytosis in the apical to basolateral direction as a possible route for elimination of waste from the CSF. Intraventricular injection of soluble horseradish peroxidase and cationized ferritin resulted in their overwhelming delivery to lysosomes; very small amounts appeared in coated pits along the lateral surface (589). The conclusion that apical-to-basolateral transcytosis was not an active pathway has been conrmed by others (24) using additional electron microscopic (EM) tracers. C. Immunological Protection and Transcytosis At several stages in the intricate choreography of the vertebrate immune response, transcytosis is used to move antigens and protective antibodies across epithelial barriers (Table 1). Antigen sampling is the rst step in the mucosal immune response and entails the apical-to-basolateral delivery of soluble and particulate antigens to underlying mucosal-associated lymphoid tissue. This transcytotic event is carried out principally by M cells that are located in lymphoid follicle-associated epithelium throughout the gastrointestinal and urogenital tracts (175, 401, 402, 579, 621). Later in the mucosal immune response, polymeric IgA, secreted by appropriately activated plasma cells, is transcytosed along the basolateralto-apical axis by epithelial cells in the digestive tract, liver, and mammary gland and is released as secretory IgA into the gut lumen, bile, and milk, respectively (245, 302, 393). The third use of transcytosis occurs in a form of systemic immune protection, termed passive immunity, which is the transport of maternal IgG to the developing fetus or neonate. Species differences dictate whether maternal blood or milk is the source of the IgG and whether the placenta or the intestine is the site of this transfer (178, 245). Certainly, the last two transcytotic processes start with uptake of their cargo through clathrin-coated pits/vesicles and may transit through parts of the endosomal system. Thus the mechanisms regulating these itineraries most likely differ from those used by endothelial cells, where a caveolar pathway predominates.
Physiol Rev VOL

1. Structural features of intestinal epithelial cells Figure 3 shows the tissue organization and ultrastructural appearances of M cells and enterocytes (adsorptive columnar cells), the two epithelial cells participating in transcytosis in the intestine. These cells are very different from one another and the capillary endothelial cell. Depending on the species, M cells comprise a variable but small percentage of the epithelia overlying organized mucosal-associated lymphoid tissue, making them a very minor cell population in the gastrointestinal tract. Being epithelial cells, their basal extensions sit on a basal lamina, but much of their basal membrane lines an extracellular pocket in which migrating monocytes and lymphocytes accumulate. As can be seen in Figure 3B, the pocket is a short distance from the apical surface. Thus these cells have evolved a short transcellular pathway much like the endothelial cells, but in contrast they have few to no caveolae; rather, coated pits are present on the apical PM. Figure 3B also shows that M cells do not have the luxuriant brush border that is present on adjacent absorptive enterocytes. They have short microvilli, or microfolds, hence the name M cells. In contrast, absorptive enterocytes are simple columnar cells with several apical features in addition to their brush borders (Fig. 3C). Clathrin-coated pits are present at the base of microvilli, and a thick glycocalyx composed of integral membrane proteins with glycosaminoglycan side chains emanates from the microvillar membrane. This latter structural feature as well as the rigidity of the microvilli are thought to prohibit microorganisms from attaching and invading enterocytes. The intracellular organization of these columnar epithelial cells is also polarized, with basally located nuclei, supranuclear Golgi, and an abundance of pleiomorphic membrane compartments underlying the terminal web of the brush border (Fig. 3C). The basolateral-to-apical length of this cell is 20 versus 0.2 m for a capillary endothelial cell, making the transcytotic route across enterocytes potentially much longer. Furthermore, microtubules are an important structural element of the transcytotic pathway in enterocytes, but not in M or endothelial cells. 2. M cells, transcytosis, and antigen sampling Quite early, researchers studying the routes of pathogen invasion discerned that specic regions of the intestine collected adherent particulate material present in the gut lumen; when the regions were visualized at the EM level, M cells were identied as the invasion route (72, 579, 621). The transcytotic route across M cells is thought to be part of the mechanism by which antigens are routinely sampled along the entire mucosal surface. Not surprisingly, numerous pathogens have evolved mechanisms to exploit the transcytotic process as a means to invade and disseminate before a strong enough immune rewww.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

880

PAMELA L. TUMA AND ANN L. HUBBARD

Downloaded from physrev.physiology.org on November 4, 2009

FIG. 3. The small intestine (ileum) contains Peyers patches. A: a schematic drawing of a Peyers patch is shown that illustrates the general arrangement of gut-associated lymphoid tissue. Lymphoid follicles in the submucosa are associated with dome areas that extend into the gut lumen. The domes are covered with specialized epithelium that contains M cells. B lymphocytes mainly populate the lymphoid follicles, while T cells predominate the interfollicular areas. High endothelial venules (HEV) in the interfollicular areas are the route through which lymphocytes enter the Peyers patch. [From Gebert et al. (175) by copyright permission of Academic Press.] B: a transmission electron micrograph of an M cell and adjacent columnar cells (CC) from the region indicated in A. The typical M cell has short, irregular microvilli and a basolateral pocket (P) into which the lymphoid cells (here resembling plasma cells) and macrophages migrate. Luminal antigens are endocytosed, transported across the apical cytoplasm (bracket), and delivered to the basolateral pocket. [From Weltzin et al. (616) by copyright permission of The Rockefeller University Press.] C: a low-magnication transmission electron micrograph of several absorptive cells and part of a goblet cell from the region indicated in A is shown from a fasted rat. The lumen of the intestine is at the top and a small portion of the lamina propria (LP) is shown at the bottom. A thin basement membrane separates the basal surfaces (BL) of the cells from the lamina propria. An elongated nucleus is located in the basal region of the cell under which is a dense cluster of mitochondria and few free ribosomes and rough endoplasmic reticulum (RER). The apical cytoplasm contains long mitochondria, a prominent Golgi component (G), RER, and smooth endoplasmic reticulum (SER) concentrated at the terminal web (TW). The free surface is covered by microvilli (Mv). L, lipid droplet. [From Cardell et al. (77) by copyright permission of The Rockefeller University Press.]

sponse can be mounted (403, 464). In recent years, this route of entry has been studied intensively in the hopes of understanding the basic mechanisms of antigen sampling and developing effective vaccine delivery systems against stealthy invaders. Because adherence is an essential rst step in invasion, researchers have focused on identifying the molecular basis for the selective adherence of antigens and pathogens to M cells and not adjacent enterocytes. Lectin staining in situ has been used in attempts to identify particular glycosidic moieties that might be differentially expressed by M cells (160, 186, 187). Recently,
Physiol Rev VOL

1-integrin was localized to the M cell apical surface and proposed as the receptor for several pathogens (257259). This membrane protein, which is expressed on the basolateral surface of neighboring absorptive cells, has a cytoplasmic tail that could mediate the endocytosis of particles bound to its extracellular domain. In support of this notion, M cells are avidly endocytic and their apical membrane is much more dynamic than the rigid and stable brush border of the enterocyte. In fact, both phagocytic and pinocytic mechanisms appear to operate at the apical surface of these cells. Adsorbed macromolecules are enwww.prv.org

83 JULY 2003

TRANSCYTOSIS

881

docytosed via clathrin-coated vesicles (404) and delivered to a prelysosomal/lysosomal compartment from which they are released into the underlying pocket for subsequent uptake by lymphocytes and macrophages (404, 425, 616). Thus, unlike endothelial cell transcytosis, lysosomes appear to play a role in M cell transcytosis. Whether the cargo in this compartment is modied by acid hydrolases present in it (6) is not currently known. 3. Transcytosis of IgA The large amount of mucosa-associated lymphoid tissue and its specialization for the production of IgA make IgA the major immunoglobulin in humans (301, 389). Given that it is synthesized and secreted by plasma cells located in the lamina propria of the digestive, respiratory, and urogenital tracts yet functions in external secretions, IgA must be delivered across an epithelial barrier. This requirement is accomplished by the polymeric IgA-receptor (pIgA-R), a single transmembrane protein synthesized by the epithelial cells. As discussed in detail in section IV, this receptor has a long (100 amino acid) cytoplasmic tail that contains most of the signals necessary to direct it through its cellular itinerary. However, unlike most other endocytic receptors that perform repeated rounds of cargo uptake, delivery and recycling, the extracellular domain of pIgA-R is cleaved upon delivery to the apical surface and released into the lumen with its ligand. The presence of the added secretory component stabilizes IgA in the gut lumen. This unique transcytotic system is expressed in many epithelia throughout the body, including kidney, trachea, and the digestive tract, including the liver (Table 1). Interestingly, some pathogens appear to have exploited the small percentage of uncleaved pIgA-R present in the apical membrane of nasopharyngeal epithelial cells to gain entry into the underlying interstitium (Table 1). This result suggests that the receptor is able or can be coerced to transcytose in an apical-to-basolateral direction. The mechanism, whether normal or pathogen induced, may have therapeutic potential. 4. Transcytosis of IgG The transfer of maternal immunoglobulins to fetal or neonatal offspring provides the latter with systemic immunity until their immune system matures. Several organs transport IgG-type immunoglobulins (245). As with IgA, maternal IgG must be transcytosed across an epithelial barrier. In all mammalian species, it is transcytosed in an apical-to-basal direction. Thus in humans, IgG in the maternal blood is transported across the placenta (Fig. 2B), while in rodents, maternal IgG is rst delivered into milk, a basal-to-apical route, and secondarily across the absorptive epithelial cells of the small intestine, an apicalto-basolateral route.
Physiol Rev VOL

The receptor mediating the apical-to-basal transport of IgG is FcRn, a distant member of the major histocompatibility complex (MHC) I family (542). As for other MHC I proteins, the FcRn is a heterodimer, with a transmembrane heavy chain and 2-microglobulin light chain. The heavy chain has a cytoplasmic tail containing an internalization motif that mediates endocytosis of maternal IgG via clathrin-coated pits and vesicles present at the base of the apical brush border of neonatal rodent enterocytes. The receptor and ligand are transported through an endosomal compartment to the lateral surface of these cells (1). Thus this transport system shares the property with M cells of using a prelysosomal compartment to deliver its cargo. However, the unique pH dependence of binding allows the ligand to remain associated with the receptor at low pH (in the gut lumen and through slightly acidic endosomes) and be released at neutral pH (in the interstitial space) without apparent modication. The FcRn recycles back to the apical PM in neonatal intestine. Although the nding that 2-microglobulin (2-M) knock-out mice lacked the apical-to-basal IgG transport system in the neonatal intestine was expected, it was initially surprising that circulating IgG in 2-M-null adults exhibited a much shorter half-life than in wild-type mice (177). This result suggested that FcRn played a role in IgG homeostasis (541), conrming a long-standing hypothesis by Brambell (54) that the prolonged circulation of IgG in plasma was due to a receptor capable of protecting IgG from degradation. Given the pH dependence of IgG binding to FcRn, the current view is that intracellular FcRn binds nonspecically endocytosed IgG within an endosome-like compartment and returns it to the circulation (177). The tissues and cells performing the protective function may be hepatocytes or endothelial cells, since both express FcRn at the PM. Quantitative studies are needed. An important nding in the 2-M knock-out mouse studies was that the levels of maternal IgG in colostrum and milk were normal, indicating that FcRn does not play a role in the transcytosis of IgG in the rodent mammary gland. This is not so surprising, considering that the direction of transport is opposite to that in the placenta or intestine, although in endothelial cells, the FcRn presumably carries IgG in the basal-to-apical direction (Table 1). The identity of the mammary gland receptor system will be important to determine. D. Role for Transcytosis in the Homeostasis of Micronutrients Most vitamins, essential minerals, and trace elements, collectively called micronutrients, come from the diet. Thus they must cross an epithelial barrier somewww.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

882

PAMELA L. TUMA AND ANN L. HUBBARD

where along the digestive tract; this occurs primarily at the level of the intestine. However, transcytosis is the least used route for micronutrient absorption. Lipid-soluble vitamins associate with bile acid micelles in the gut lumen and are thought to then partition progressively and passively across absorptive cells, associating with chylomicra somewhere before or at the basal side of the cells. Dietary vitamin B12 (cobalamin) is an exception, because it uses vesicle-mediated steps, in part, to cross intestinal cells. Many minerals are assumed to be absorbed paracellularly (61). This assumption is based on calculations of transit times and absorption rates. However, dietary iron is transported across the intestinal epithelium via multiple membrane transporters; once in the circulation, its delivery to the brain and fetus requires transcytosis. Additionally, Cu and Zn, as well as other heavy metals, appear to be transported into intestinal absorptive cells via membrane transporters at the apical plasma membrane. Finally, the kidney proximal tubule cells provide an important function in vitamin homeostasis by avidly scavenging several vitamins (Table 1) from the urine using a modied type of transcytosis. 1. Vitamin B12 All cells require vitamin B12 as a coenzyme in onecarbon transfers. Methyl malonyl CoA mutase uses it in the adenosyl-B12 form to convert methyl malonyl CoA to succincyl CoA in the mitochondria; methionine synthetase uses it in the methyl-B12 form to convert homocysteine to methionine in the cytoplasm. B12s journey to the cytoplasm of all cells is a fascinating and curiously

convoluted process. The players so far identied are listed in Table 2 and placed in cellular context in Figure 4A. Several reviews cover this topic in more detail (276, 488, 514). A) UPTAKE FROM THE INTESTINAL LUMEN. In carnivores, B12 is present in ingested meat as the cofactors mentioned above. Upon digestion by pancreatic enzymes in the small intestine, free B12 is bound by intrinsic factor (IF), a 27-kDa glycoprotein secreted by parietal cells of the stomach (317). In the terminal ileum of the small intestine, the luminal B12-IF complex binds to its receptor, cubilin, a large membrane-associated glycoprotein that is located in the microvillar brush border of absorptive enterocytes (515). Cubilins association with megalin, a member of the LDL receptor-related (LRP) family of endocytic receptors (reviewed in Refs. 192, 619), leads to the internalization via clathrin-coated vesicles of the entire cubilin-IF-B12 complex. After delivery of the IF-B12 complex to endosomes, cubilin and megalin recycle for further rounds of endocytosis; since cubulins association with megalin is stable at pH 5, it is thought to stay associated throughout. Meanwhile, dissociated B12-IF is delivered to lysosomes, where the protein is degraded by leupeptin-inhibitable acid hydrolases (196) and B12 is transported out of lysosomes. This last step is mediated by a yet-to-be-discovered transporter(s). Interestingly, in this pathway there is no avoidance of lysosomes; to the contrary, lysosomal function is essential, since failure to degrade IF within the intestine results in a B12 deciency in all subsequent tissues and cells. Some investigators are exploring the B12 entry pathway as a means to deliver drugs orally (487).

Downloaded from physrev.physiology.org on November 4, 2009

TABLE

2. Vitamin B12 (cobalamin)


Molecule Site of Synthesis/Expression Biochemical Characteristics Functions Reference No.

B12 R binders (haptocorrins) Intrinsic factor (IF) Cubilin

Bacteria

Salivary gland, stomach, placenta, circulation, and granulocytes Parietal cells/stomach Enterocyte in terminal ileum Enterocyte in terminal ileum; also present in kidney proximal tubule, placenta, thyroid Liver and enterocyte

Megalin

Small secreted glycoproteins (includes TCII, not a glycoprotein) 27-kDa secreted glycoprotein 400-kDa peripheral membrane glycoprotein secreted apically by enterocyte 600-kDa transmembrane glycoprotein, clathrin internalization motif in cytoplasmic tail 45.5-kDa secreted protein

Coenzymes (adenosylcobalamin, methylcobalamin) Bind cobalamin

69

Binds B12 in small intestine Binds B12-IF complex at apical PM Apical PM receptor that binds cubilin-IF-B12 and TCII-B12 complexes in Ca-dependent manner and mediates endocytosis; recycles Binds B12 in circulation Bind B12; unknown functions Basolateral PM receptor for TCII-B12

317 294, 515

381

Transcobalamin binding protein (TCII) TCI and III TCII receptor

448, 460, 463

Granulocytes Kidney, liver, placenta, intestine

Secreted glycoproteins 62-kDa transmembrane glycoprotein

49, 518

Denitions are as in Table 1. Physiol Rev VOL


83 JULY 2003

www.prv.org

TRANSCYTOSIS

883

Downloaded from physrev.physiology.org on November 4, 2009

FIG. 4. The intracellular itineraries of vitamin B12 and iron in polarized enterocytes and nonpolarized peripheral cells. A: the current model for the trafcking of vitamin B12 is shown. In the polarized enterocyte, B12 complexed with intrinsic factor binds the cubilin-megalin complex and is internalized via a clathrin-mediated pathway. In acidic endosomal vesicles (EV), the ligand dissociates and is sorted to the degradative pathway, whereas the cubilin-megalin complex is sorted to dense apical tubules (DAT) and recycled back to the PM. In lysosomes, intrinsic factor is degraded and vitamin B12 is released and then secreted coupled to transcobalamin (TC) II that is shown in a secretory vesicle. In the peripheral cell, the TCII-B12 complex that is bound to the TCII receptor is internalized via coated pits (CP). TCII ligand is degraded in lysosomes and B12 is released into the cytoplasm by mechanisms that are not completely understood and delivered to other cellular destinations. B: the proposed trafcking pathways for iron (Fe) are shown. Iron is transported into the polarized enterocyte via DMT1 present at the apical cell surface. Ferroportin present at the basolateral PM transports iron from the cell where it binds transferrin in the circulation. The iron-transferrin complexes bind peripheral cells via associations with the transferrin receptor, and the ligand-receptor complexes are internalized in coated pits (CP). Iron dissociates from transferrin in an early endocytic compartment (EE) and apotransferrin, still bound to the receptor, is delivered back to the PM via a recycling compartment (RC). It is unclear how the free iron is released into the cytoplasm. Here we have suggested that DMT1, present on early endosomes, transports the iron. CV, coated vesicle; lys, lysosome; mito, mitochondria; PLC, prelysosomal compartment.

B) TRANSFER TO CIRCULATING TRANSCOBALAMIN II AND TRANSCYTOSIS.

A puzzle is the mechanism by which cytoplasmic B12 is subsequently transported into the basal milieu surrounding the enterocyte. Extracellular transcobalamin (TC) II, a 40-kDa protein in the interstitium/circulation, serves as the major functional carrier of B12 (Table 2). (TCI and TCIII are also B12 carriers, but their functions remain unknown.) TCII is synthesized and secreted principally by the liver (206, 495). Evidence that enterocytes express a TCII transcript (460) suggests that B12 may be loaded onto newly synthesized protein as it transits the secretory pathway (463). The B12-TCII complex would then be released
Physiol Rev VOL

at the basolateral surface of the enterocyte. Of course, this scenario requires the presence of a B12 membrane transporter in the secretory pathway. Once in the circulation, how does TCII-B12 reach cells? All cells express a TCII receptor, which mediates the endocytosis of the complex via a clathrin-mediated mechanism. The TCII receptor is a single transmembrane glycoprotein of 62 kDa (49) that functions as a homodimer at the plasma membrane (Table 2). After internalization, the TCII-B12 complex is delivered to lysosomes, where TCII is degraded and B12 is again transported into the cytoplasm for subsequent use as a
www.prv.org

83 JULY 2003

884

PAMELA L. TUMA AND ANN L. HUBBARD

cofactor (249). But how does B12 reach cells behind a selective barrier, for example, the brain or testis? Although a B12-TCII receptor on brain or testicular endothelial cells has not been reported, we predict that it must be there. Could it be the same receptor as that found on the basolateral PM of most epithelial cells, even though it would be on the apical PM of these endothelia? What is the mode of transcytosis and the subsequent fate of the TCII protein and B12 in brain endothelial cells? Are the fates different from those in other cells? Clearly, this system would be interesting to explore further.
C) INVOLVEMENT OF ADDITIONAL EPITHELIA IN B12 HOMEOSTASIS.

There are many excellent reviews covering different aspects of iron metabolism (5, 11, 203, 277, 278, 485).
A) TRANSPORT ACROSS THE INTESTINE APPEARS NOT TO REQUIRE

There are additional aspects of B12 homeostasis that deserve comment. The kidney, yolk sac, and placenta express the protein components involved in intestinal B12 absorption (294). For example, cubilin is very abundant in the kidney proximal tubules, where it can bind and internalize B12-IF, again via megalin (91, 517). This is strange, since IF is not normally found in the circulation. Not unexpectedly, given the small size of TCII and thus its ltration by the glomerulus, there is a scavenger of B12TCII in the kidney, and it is megalin itself on the apical PM of proximal tubule cells. Similar to its behavior in the intestine, megalin internalizes the protein-B12 complex and delivers it to lysosomes, where TCII is degraded and B12 is stored in a form that is retrievable into the circulation upon demand. This last step, storage of B12, points to possible differences between B12 dynamics in intestinal and kidney epithelial cells and is worth following up. Finally, antibodies to cubilin cause severe defects in developing fetuses, possibly due to a failure to deliver B12 and/or other essential components to the developing central nervous system via the yolk sac (516). As can be seen in Table 1, the cubilin-megalin system has the capacity to bind and move many different cargo molecules. 2. Iron An essential cofactor in the homeostasis of every cell, iron in mammals exists in three predominant forms: bound to the circulating plasma protein transferrin; as heme in intracellular proteins such as mitochondrial cytochromes, hemoglobin, and myoglobin; and in ferritin, the storage form of iron (5). Iron is avidly reutilized by mammals, and thus the daily requirement for it is small. Of 3 4 g total body iron, only 12 mg are lost per day through desquamation. However, because there is no mechanism for its disposal, excess iron leads to disease (525, 623). Furthermore, the insolubility of both valence states at neutral pH and the toxicity of Fe2 in the presence of oxygen makes control of this essential micronutrient especially vital. Studies of both iron deciency and overload in human disease and animal models have helped to elucidate the mechanisms of iron homeostasis.
Physiol Rev VOL

Dietary iron is initially absorbed in the duodenum in either the heme and nonheme (free or chelated) form; much more is known about uptake of the latter. The current consensus is that free Fe2 is transported directly across the apical membrane of absorptive epithelial cells via a multistep process (203) involving the divalent metal transporter DMT-1 (Fig. 4B) (202). The protein accepts a broad range of divalent ions. Older reports in the literature implicated a vesicular process for irons uptake, consisting of a mucin at the apical surface binding free iron, followed by its import via an integrin and transfer to a molecule termed mobilferrin, which was later identied as calreticulin, an endoplasmic reticulum lumen chaperone. With the identication of DMT-1, the mobilferrin hypothesis is now open to question (12). After transport into the epithelial cell, cytoplasmic Fe2 is subsequently directed across the intestinal basolateral membrane via another newly identied transporter, called ferroportin (372). In intestine, a membrane form of the ferro-oxidase ceruloplasmin, called hephaestin (607), is thought to aid both in the oxidation of Fe2 and the loading of Fe3 onto Tf. How this occurs is still unknown, although Caco-2 cells are reportedly capable of transferring apically derived iron onto apo-Tf in the basal medium (342). Interestingly, the livers of atransferrinemic mice become iron overloaded, implying that a nonTf-bound transport mechanism must operate to deliver iron from the intestine into the hepatocyte. Possible carriers have recently been reported (reviewed in Ref. 277). B) UPTAKE OF TF-BOUND IRON. Once in the circulation, iron is carried principally by hepatocyte-derived plasma Tf. Cells directly accessible to the circulation take up iron via the well-studied process of Tf receptor-mediated endocytosis (Fig. 4B); that is, iron bound to Tf is internalized in clathrin-coated vesicles, dissociated from Tf by the low pH in endosomes, and transported across the membrane, most likely by a DMT-like transporter. Apo-Tf bound to its receptor recycles to the same cell surface for subsequent rounds of uptake, delivery, and recycling. How do cells located behind a selective barrier obtain this essential mineral? And how do cells responsible for transcytosing it obtain sufcient iron for their own metabolic needs? In the adult, iron reaches cells in peripheral tissues by nonselective caveolar-mediated transcytosis of Tf-bound iron across the endothelium; there is no dissociation in transit. C) TRANSCYTOSIS OF IRON IN THE PLACENTA AND BRAIN. As described in section IIB1 for brain, transcytosis of iron across cerebral capillaries is receptor mediated. Also, as described earlier, the extent to which plasma-derived Tf moves across brain endothelial cells is controversial and presently unresolved (52). There is a similar uncertainty
VESICULAR CARRIERS.

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

www.prv.org

TRANSCYTOSIS

885

in the transcytosis of iron across the human placenta. As for brain endothelium, the direction across the syncytiotrophoblasts is apical to basolateral (Fig. 2A). Is maternal or fetal Tf the carrier? Finally, the fetal endothelial capillary is an additional cellular barrier that must be crossed and the mechanism is currently unresolved. E. Additional Transcytosis Systems 1. Lung Transcytosis occurs in the upper regions of the respiratory tract and involves two receptor systems already described, pIgA-R and FcRn (Table 1). Secretory IgA is a known constituent of the lungs immune defense system, with bronchial epithelial cells carrying out basolateral-toapical transport of dIgA, which is secreted by local plasma cells in underlying lymphoid tissue (reviewed in Refs. 444, 491). A recent study using a clever biological read-out has documented the efcient apical-to-basolateral transcytosis of intact IgG across bronchial epithelium via the FcRn (553). This latter pathway could possibly be exploited to deliver genes systemically. Finally, albumin, which is found in lung uid, is endocytosed specically at the apical surface of airway epithelia but is then subsequently degraded. At the alveolar level, the question of whether albumin is transcytosed intact is uncertain (see Ref. 437 for review). Malik and colleagues (267) have recently reported the presence and function in type II epithelial cells of a gp60 membrane protein related to that found in endothelial cells. 2. Mammary gland Milk is composed largely of locally synthesized nutrients that are secreted by alveolar epithelial cells in the lactating gland (523). However, a substantial fraction of milk proteins is thought to be derived from the serum, meaning that transcytosis must be used to deliver these exogenous molecules. To date, distinguishing between the two sources has not been systematically done. Local plasma cells secrete IgA, which is transcytosed by luminal epithelial cells using the pIgA-R (Table 1). As already described, rodents but not humans transcytose maternal IgG into milk for several days after pups are born. The receptor and pathway are unknown. Micronutrients in milk are supplied from the maternal circulation, but neither their transcellular path nor the source of binding protein (e.g., for iron, B12, or vitamin D) is clear. Iron is transcytosed, but milk transferrin is synthesized in the gland, potentially necessitating an intracellular transfer. Ca2 is also derived from the maternal circulation, but its concentration in milk is 100-fold higher than that in serum. The Golgi Ca2-ATPase has been proposed as the pump that sequesters Ca2 in vesicles, which are subsePhysiol Rev VOL

quently delivered to the apical membrane with release of Ca2 via exocytosis (523). 3. Thyroid The thyroid hormones triiodothyronine (T3) and thyroxine (T4) are produced from their iodinated precursor protein, thyroglobulin, which is stored in the lumen of a thyroid follicle. Upon stimulation by thyroid stimulating hormone at the basolateral surface, apical endocytosis increases and thyroglobulin is internalized. The mechanism of uptake is not yet known and may be nonspecic (143, 226, 483). Endocytic vesicles fuse with lysosomes, and the cathepsins act on thyroglobulin to release 20-kDa fragments containing the hormonogenic regions, which are further cleaved to T3 and T4 by endo- and exopeptidases. It is a mystery how the hormones reach the circulation, since T3 receptors are present in the thyroid cell, yet net movement of the hormones is toward the basolateral surface. Once in the circulation, T3 and T4 are bound to their protein carrier, transthyretin, which is synthesized in the liver. The complex is transcytosed across continuous endothelium via caveolae. It is presently unclear how these hormones reach the brain. Perhaps the choroid plexus, which synthesizes transthyretin, plays a role. Approximately 10% of the thyroglobulin protein internalized from the thyroid follicular lumen is not processed in lysosomes, but is transcytosed intact into the circulation. In the 1980s, Herzog (227) found that this amount did not represent spillover from saturation of a putative lysosomal delivery system, since lysosomes continued to ll as increasing amounts of thyroglobulin were internalized. More recently, researchers have identied megalin as the apical membrane receptor mediating thyroglobulins apical-to-basolateral transcytosis across the follicular cell (348, 350, 351). This nding raises several questions. Does megalin also cross the cell and reach the basolateral PM? How does megalins cargo avoid lysosomes in the thyroid, since in the intestine, its cargo, vitamin B12 bound to intrinsic factor, is delivered to lysosomes for degradation? The in vitro cell system models described in section III could be useful for answering these questions. F. Role of Transcytosis in Plasma Membrane Biogenesis In Vivo Of the in vivo transcytosis systems described so far, a common feature is that the cargo is exogenous. However, in two epithelial cell types, the hepatocyte and the absorptive enterocyte of the intestinal villus, endogenous membrane proteins are themselves the cargo! These cells use basolateral-to-apical transcytosis exclusively (hepatocytes) or in part (enterocytes) as a pathway for the delivwww.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

886

PAMELA L. TUMA AND ANN L. HUBBARD

ery of specic classes of newly synthesized apical plasma membrane proteins. In hepatocytes we determined some years ago that three single transmembrane domain (TMD) apical proteins and one glycosyl-phosphatidylinositol (GPI)-anchored apical protein followed this route (32, 499). Similar observations were made by Maroux and colleagues (148, 355) in the rabbit intestine for aminopeptidase N. Thus the question is whether such an indirect pathway of apical PM biogenesis exists in other epithelia in vivo. Unfortunately, we may never have a denitive answer, largely because other epithelial tissues are not sufciently homogeneous nor as amenable as the liver and intestine to the biochemical approaches that were used. In these studies, animals were rst administered radiolabeled amino acids in a pulse-chase fashion, then enriched populations of apical and basolateral membranes were isolated, the specic membrane proteins immunoprecipitated and their radioactive content determined. Because hepatocytes constitute 70% of the total cells in the liver, subcellular fractionation methods could be used that yielded preparations of highly enriched hepatocyte organelles. Similarly, intestinal mucosa could be scraped from the surface of everted intestines, thus enriching for epithelial cells and allowing subsequent subcellular fractionation of membranes into apical- and basolateral-enriched fractions. Why are these in vivo studies important? First, they represent reality, that is, the physiological situation. Second, the results were different from those reported earlier using in vitro polarized cell models, specically MDCK cells derived from dog kidney (360, 479). In the latter cells, newly synthesized apical PM proteins were shown to be delivered directly from the trans-Golgi network (TGN) to the apical surface. Gradually, as different epithelial cells and more membrane proteins have been studied, the plasticity in the routes and mechanisms for the delivery and retention of PM proteins in epithelial cells has become apparent. Such a realization reinforces the importance of studying a variety of epithelial cells to learn the full repertoire of mechanisms. It also points to the possibility that transcytosis is an ancient route, since all epithelial cells express this transport system. III. IN VITRO CELL MODELS OF TRANSCYTOSIS The use of in vitro cell models to study transcytosis has many advantages over in vivo systems. First, variation among animals is eliminated, as is the confounding issue of cargo possibly being modied or endocytosed by cell types other than the one under study. Moreover, in vitro systems can be manipulated in ways not possible in vivo, allowing investigators to measure the effects of different variables (e.g., temperatures, pharmacological agents,
Physiol Rev VOL

etc.) with greater precision and to explore the molecular mechanisms of transcytosis. However, these advantages are offset by the loss of the in vivo context (e.g., cues from extracellular matrix, other cell types), which undoubtedly provides levels of regulation that are missing in vitro. For this reason, it is important to be cautious in extrapolating in vitro results to the in vivo situation and to compare results obtained in the two systems whenever possible. A. What Constitutes a Good Transcytotic Cell Model? An ideal cell model would faithfully recapitulate the in vivo transcytotic system in the types, amounts, and kinetics of cargo transported across the cellular barrier. If these criteria are met (not a simple feat), we can assume that other parameters, such as cellular organization, relevant machinery, tight junction permeabilities, etc., are in place. What experimental factors should be considered and assessed in establishing a good (i.e., less than ideal) in vitro cell model? Based on a review of the literature and our own experience, we have come up with six issues that pertain to simple (bipolar) epithelia. An important factor is the choice of substratum. Quite early, physiologists recognized that simple epithelial cells achieved a higher degree of polarity and manifested a more differentiated phenotype when they were grown on porous surfaces with media bathing them on both sides. This three-dimensional arrangement simulates the in vivo condition. Filters of various chemistries and coatings are now commercially available (Table 3). To date, cell growth, morphology, and polarity on different lters have been compared systematically in only a few cases (71, 601). The issue of pore size is relevant for two reasons. First, cells can migrate through larger pores and grow on the underside of lters (581). Obviously, this situation would make interpretation of transcytosis measurements difcult. Not surprisingly, different cell types have varying tendencies for this behavior. However, pore diameters of 1 m or less seem to prohibit migration of even the most aggressive cells. Second, the pore size can affect access of transcytotic tracers in the basal medium to the basal surface of cells. Of course, the larger the transcytotic cargo (e.g., bacteria), the more relevant this issue. However, the extent to which even small reagents such as the surface-labeling reagent NHS-biotin (molecular mass 557 Da) have access to the basolateral surface of lter-grown cells has been debated in interpretation of experimental results (241). A third factor to consider in the quest for a good in vitro cell system is the cell seeding density. The goal is to obtain a conuent monolayer of homogeneous cells. For
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS
TABLE

887

3. Commercially available lters


Name Material Pore Size, m Comments

Company

Costar

Transwell Transwell-Clear Transwell-COL Falcon Biocoat

Polycarbonate Polyester Polytetrauorethylene (PTFE) Polyethylene-terepthalate (PET) or uoropolymer (FP) PET or FP

0.112 0.43 0.43 0.48 0.48

Becton-Dickinson

Millipore

Millicell-CM (Biopore-CM) Millicell-HA Millicell-PCF

Polystyrene Mixed cellulose esters Polycarbonate

0.4 0.45 0.412

Opaque Transparent Coated with bovine collagen (I and III) Transparent, translucent, high and low pore density Coated with bovine collagen (I or IV), laminin, bronectin, or Matrigel Transparent, needs coating Coating not needed Coating not needed

[Adapted from Hughson and Hirt (241).]

Downloaded from physrev.physiology.org on November 4, 2009

example, sparse seeding of some cell types can lead to conuence being achieved at different times across the lter. This situation could have a profound inuence on the differentiation state of the cells. Caco-2 cells, a wellstudied model of intestinal absorptive enterocytes, progressively differentiate over a period of 1520 days after achieving conuence. Therefore, they are seeded at high density so that conuence is achieved across the entire lter synchronously. The disadvantage of seeding at high density is the possibility of selecting for more rapidly attaching cells. Likewise, some cell lines alter their phenotype depending on the seeding density. This appears to be the case for placental BeWo cells, which form multiple layers if seeded too high (327). Such a situation could lead to erroneous results and interpretations regarding transcytosis. BeWo cells seeded at low density (subconuence) will grow into a conuent monolayer (Table 4). The integrity of the monolayer is obviously vital to every study of transcytosis, and there are different methods for assessing it. Transepithelial electrical resistance (TER) measurements are commonly used as an indication of tight junction integrity in a monolayer, and commercial instruments are available for these measurements (241). For meaningful interpretation of results, especially when studying brain endothelium permeability, it is important to compare the in vivo and in vitro TERs (201). Radiolabeled inulin (molecular mass 5,200 Da), an inert, uncharged molecule that reports paracellular leak, is often used, because it can be added to the apical medium and measured in the opposite bath after various times of incubation. Here, a positive control consisting of parallel measurements made to a lter with no cells is important, since there is not immediate equilibration of a tracer between the two chambers in the absence of cells. A simple method for assessing monolayer integrity is to ll the upper chamber to the top, then leave the cells overnight and measure leak by a fall in the uid level in the top chamber. The ideal would be to measure the integrity of
Physiol Rev VOL

each lter before using it experimentally, but this is rarely done. Knowing the extent to which a cells surface is polarized at a molecular level is crucial to a meaningful interpretation of the results of transcytotic studies. We have termed this factor the polarity index, which is the relative distribution of a membrane component in the two PM domains, apical and basolateral. Since the polarity index of most PM proteins is lower in vitro than in vivo, a larger fraction of a particular transcytotic receptor (and its associated intracellular machinery) may be in the incorrect PM domain in vitro. This condition would lead to an apparent higher transcytotic activity in the wrong direction. A possible solution would be to determine the in vivo and in vitro polarity indices for the receptor under study and then correct back to the in vivo index. Finally, we assert that complete balance sheets are essential when studying transcytosis in vitro. By this we mean accounting for all of the tracer over the time course of the experiment. Unfortunately, such bookkeeping is not usually done. Of course, two measurements are routinely made: 1) tracer uptake from one side of the monolayer and 2) its appearance on the opposite side. But these two measurements alone are not sufcient for a denitive demonstration of transcytosis. It is also important to determine 3) the intracellular accumulation of the cargo, 4) any possible degradation/metabolism, and 5) the integrity of the cargo that crossed the monolayer. These additional measurements give clues as to whether protein cargo avoided lysosomal proteolysis, a feature of some transcytotic pathways. It is important here to raise a cautionary note about the nature of tracers used to detect and measure transcytosis. Radiolabeled tracers have often been used in biochemical studies, while macromolecules adsorbed to colloidal gold or coupled to the cytochemical agent, horseradish peroxidase (HRP), have been used in microscopic studies. Because chemical or physical modications to a
www.prv.org

83 JULY 2003

888

TABLE
Transcytosis Studies Characteristics Peripheral endothelial cells 118, 530, 531 11- to 150-kDa proteins 0.8-m pore gelatin-coated polycarbonate lter Lung microvessel endothelium less permeable than endothelium of larger vessels in comparative study; 2- to 5fold higher density of plasmalemmal vesicles in microvessel endothelium 511 Reference No. Culture conditions Cargo Comments Reference No.

4. Useful in vitro transcytotic systems

Cell

Origin

Primary endothelial

Bovine pulmonary microvessels

Bovine pulmonary artery Bovine pulmonary vein Bovine pulmonary microvessels 118 0.4-m gelatin-coated polycarbonate lter
125

I-albumin and HRP

268, 576, 602

Primary endothelial

Bovine aorta microvessels

97

3-m pore gelatin and bronectin-coated PET lters; cells used 56 days after seeding Lipoprotein lipase Brain endothelial cells

Anti-gp60 antibody stimulated A-BL transcytosis with no change in monolayer integrity Intact and active lipase moved BL-A via VLDL receptor/heparin sulfate proteoglycan-specic process

PAMELA L. TUMA AND ANN L. HUBBARD

Downloaded from physrev.physiology.org on November 4, 2009

Physiol Rev VOL


113116, 376 LDL Astrocytes from newborn rat cerebral cortex cocultured with endothelial cells on opposite sides of 0.4-m pore rat tail collagen-coated Millicell CM lter Transferrin and iron Lactoferrin 271, 407 Collagen-coated Transwell lter Strep pneumococci 560 Collagen-bronectin-coated Transwell lter Alzheimers amyloid beta 140 peptide Intestine 637 Transwell lters; cells used 15 20 days after conuency BL population of apical PM proteins Endogenous p137 membrane protein Endogenous TCII receptor and 125I-TCIIB12

414

From A-125I-LDL and LDL-Au into multivesicular bodies, but 125I-LDL not degraded; from A-acetylated LDL taken up by another receptor and degraded; concludes that lysosomal pathway is functional in conuent cells. A-BL transcytosis of 125I-transferrin59Fe 7-fold greater than BL-A A-BL of intact cargo is mediated by lipoprotein-related receptor A-BL of only transparent forms mediated in part by PAF receptor; slow (46 h)

112

83 JULY 2003

Coculture of Bovine brain primary endothelial cells (stable up to 50 generations) with rat astrocytes

122 151 473

TER of 660 after coculture for 7 days; TER maintained 10 days; expression of -glutamyltranspeptidase, P-glycoprotein and alkaline phosphatase; permeability to sucrose, inulin, and propranolol showed good correlation with in vivo; [3H]dextran (70 kDa) used to test integrity of monolayer

www.prv.org

EC219 and human endothelial cells

TER of 125 (rat) and 200 (human)

Endothelial cells

Rat and human brain microvessel endothelial cells immortalized with retrovirus Collagenasedissociated human brain cerebral cortex microvessels

Positive for factor VIII, acetylLDL uptake; used after 5 days

A-BL preferred; RAGE-mediated in part; no degradation

343

Caco-2

Human primary colon carcinoma

307, 308, 375 141 49, 451

Similar to fetal colon and adult small intestine absorptive enterocytes; brush-border ectoenzymes high but glycocalyx low to absent

Single TMD apical proteins transcytosed, but GPI did not Bidirectional transcytosis without degradation A-BL transcytosis preferred

TABLE
Transcytosis Studies Characteristics HIV Millicell-HA lter Facilitated A-BL movement of toxin B and TSST-1, not toxin A Reference No. Culture conditions Cargo Comments Reference No. 232, but see 8, 400 207

4Continued

Cell

Origin

Caco-2

TER 150

TER 200 1415 days after seeding cells Type I collagen coated Millicell HA lter

S. aureus toxins, enterotoxins A, B and toxic shock syndrome toxin 1 (TSST-1) Oligopeptides, bradykinin (9 aa)

526

Insulin-transferrin conjugate

520, 628

284

Vibrio cholerae and latex beads

A-BL 4- to 10-fold over BL-A, hydrophobicity index a factor, transport concluded to be adsorptive, not receptor mediated 3-Fold greater A-BL than BL-A of conjugate; ligand not degraded after movement (binding to anti-insulin); transcytosis of free insulin 1/15 of conjugate in A-BL direction; possible oral route for insulin; caution: polarity index of Tfn-R not measured A-BL transport

285

Downloaded from physrev.physiology.org on November 4, 2009

Physiol Rev VOL


124 0.4-m pore, type I collagencoated lter; [3H]inulin 3-m polycarbonate lter; Caco-2 also studied; day 11 best for Caco-2 and day 14 for T84 E. coli K1 strains Transwell lter endogenous FcRn Botulinum neurotoxin, types A, B, and tetanus Cholera toxin (84 kDa) 484 Not used for transcytosis studies Liver 135 pIgA 51 Collagenase-dissociated cells cultured in collagen gel sandwich; no lter needed No lter needed HDL cholesterol No lter needed 3 No lter needed; cells used 3 days after seeding Horseradish peroxidase Short-chain uorescent lipid analogs 83, 521 No lter needed; cells used 10 12 days after seeding NBD-phosphatidylcholine and ergosterol pIgA Endogenous and exogenous apical PM proteins

T84

Human colon carcinoma

In the presence of murine lymphocytes of Peyers patch Caco-2 cells differentiate into M-like cells Crypt cell features (ion secretion)

125 344

TRANSCYTOSIS

BL-A 3.6-fold over A-BL, balomycin sensitive (vacuolar ATPase inhibitor) Intact A toxin A-BL 1.5-fold over BL-A; toxin B not bound; Caco-2 also studied; MDCK totally inactive Active bacteria transcytosed A-BL; good controls for cytopathic effect and paracellular movement 13% of apically applied toxin appeared on BL side after 2 h

83 JULY 2003

70

313

www.prv.org

HT29

Human colon carcinoma

Pluripotent intestinal cell line; useful for studies of intestinal development (/glucose)

Primary hepatocytes

Rat liver

Perinuclear accumulation before delivery to apical Scavenger receptor-B1-mediated uptake at B surface and selective A transcytosis of lipid

222

Primary hepatocyte couplets

Rat liver

Consistently polarized cells after 34 days; canaliculi formed Short-term (024 h) culture of groups of cells with common bile canaliculus; visual assays used due to small fraction (30%) of couplets

532

Same

Same

489 593, 594, 596

HEP-G2

Human hepatocellular carcinoma

Variable fraction of polarized cells

213, 626, 627

WIF-B

Rat hepatoma (Fao)-human broblast (WI38) hybrid

Up to 90% polarized cells, apical cysts form, not canaliculi

BL-A transcytosis via tubulovesicular intermediates BL-A and A-BL transcytosis of C6-NBDsphingomyelin; BL-A of C6-NBDglucosyl-ceramide Vesicular and nonvesicular BL-A transport MAL-2 required for BL-A transcytosis Use of antibodies to measure BL-A transcytosis

120 34, 250

889

890

TABLE
Transcytosis Studies Characteristics Liver 209, 470 0.4-m pore Transwell lters; cells used 34 days after seeding; 2,000 TER 0.4-m pore Transwell lters; cells used 35 days after seeding 3-m pore collagen-coated PET and 0.4-m pore polycarbonate lters Human pIgA-R Human FcRn Various uid-phase markers [e.g., Lucifer yellow, HRP, Texas red-dextran (10 kDa)] Exogenous rabbit pIgA-R and ligands (anti-pIgAR IgG; dIgA) 44,605 Reference No. Culture conditions Cargo Comments

4Continued

Cell

Origin

Reference No.

Madin-Darby canine kidney (MDCK)

Normal dog

MDCK I, high resistance TER 510,000; collecting duct features

MDCK II, low resistance (TER of 150), some proximal tubule features, but no megalin, limited brush border

Comprehensive biochemical and morphological analysis of endocytosis, recycling and transcytosis from both A and BL Multiple studies; BL-A transcytosis of dIgA rapid (30 min)

390

397

454, 556

PAMELA L. TUMA AND ANN L. HUBBARD

Downloaded from physrev.physiology.org on November 4, 2009

Physiol Rev VOL


569 3-m pore high density PET lter Retinol-binding proteinvitamin A 243 0.4-m pore Transwell lter Vitamin B12-TCII 293 Millicell-HA lter; cells used 14 days after seeding Lung 110 Albumin, 70-kDa dextran
57

Immortalized rat proximal tubule cells (IRPT) LLC-PK

Rat

IgM and IgA bound, translocated and released equally; therefore, preference for IgA in vivo thought to be ECM barrier to IgM pentamers diffusion Synthesis and tracking of endogenous FcRn; nonvectorial targeting of newly synthesized with repeated rounds of A-BL and reverse; cytoplasmic dileucine important for transcytosis Megalin-mediated; 20-fold greater A-BL than BL-A transcytosis of vitamin A

347

83 JULY 2003

Male pig

ts-SV40 transformed; proximal tubule proteins expressed (glut2, megalin); TER of 140; [3H]mannitol permeability of 1.4/40% (/cells) Proximal tubule cells; lack 1B adapter subunit

405

www.prv.org
76 Transwell-COL lter; cells used 4 days after seeding; airliquid interface Tissue culture-treated polycarbonate lter Used after 48 h; not lter grown 267 188 153, 522 441 0.4-m pore Transwell-Clear lter; 7002500 TER 0.4-m pore Transwell lter

OK

Oppossum

TER of 50

Co-B12-IF

TCII degraded 60% B12 stays inside; 20 25% B12 complexed to newly synthesized TCII or haptocorrin goes A; 812% B12 goes BL 100% A-BL of intact cargo via cubilin; LLC-PK and MDCK have much less to no cubilin

462

Guinea pig

Criteria for monolayer integrity: lack of leak into A over 24 h

Albumin transported A-BL 10-fold greater than BL-A; dextran not transported Tf-HRP

110

Primary tracheal cells Primary alveolar type II cells

Rat

123

Substrate and cell density important for maintaining type II phenotype (microvilli, lecithin synthesis, lamellar bodies); transdifferentiation into type I cells (with caveolin 1)

Albumin

267

A549

Human type IIlike cells

Calu-3

Serous cell phenotype

Peptides, proteins and dextrans 1,000150,000 mol wt IgA Strep pneumococci

50% of apically internalized conjugate moved A-BL; conjugate transported HRP alone Saturable endocytosis; overlap with caveolin not lysosome markers; which alveolar cell type studying? in vivo clearance also Permeability same at 4 and 37C, indicating paracellular transport BL-A upregulated by IFN- A-BL via hpIgA-receptor; in vivo correlation and MDCK cell also studied

291

330 549

Detroit 562 cells

Human bronchial cell Human pharyngeal carcinoma

TER of 160

TABLE
Transcytosis Studies Characteristics Placenta 39, 283, 288 Millicell lter IgG 10% of apically applied transcytosed in 60 min; A-BL BL-A 10-fold; HRP transcytosis in both directions LDL-Au to lysosomes and IgG-Au to basal tubular elements 550 Reference No. Culture conditions Cargo Comments

4Continued

Cell

Origin

Reference No.

Normal human term placenta 0.45-m pore Millicell-HA lter LDL and IgG FITC-IgG and FITCdextran (70 kDa) IgG 436 0.4-m pore low-density lter; cells used 7 days after seeding at subconuence

Primary placental syncytiotrophoblasts

550 139, 140

BeWo

Human chorionic carcinoma

Cytotrophoblasts isolated and cultured; keratinocyte growth medium induces synctia formation; alkaline phosphatase expressed on apical surface Formation of synctial cells that produce placental hormones and have apical brush border; TER of 60

557

Downloaded from physrev.physiology.org on November 4, 2009

Physiol Rev VOL


Tf-iron; endogenous Tf-R 266 IgG 0.4-m pore Transwell lter; cells used 4 days after seeding; TER of 600 0.4-m pore Transwell COL lter; TER of 2050 Thyroid 363 Grown on oating collagen gels Thyroglobulin 405 406 Cationized ferritin None Apical PM proteins Transwell lter TSH; inulin permeability low Transwell lter; cells used 17 days after conuency Transwell Additional systems 174 pIgA-R-transfected cells seeded on 0.45-m pore Transwell lters, coated with types I, III, or IV collagen 0.4-m pore Matrigel-coated polycarbonate lters; plus retinoic acid for 7 days at permissive then 36 h at nonpermissive 396 IgA 38, 395 Thyroglobulin Newly synthesized exogenous apical PM protein

A-BL of hIgG mediated by FcRn and 3.5fold greater than BL-A; dextran both directions, much less compared A-BL of IgG in MDCK / placental alkaline phosphatase expression to BeWo; A-BL IgG in MDCK low / PLAP; A-BL IgG much higher in BeWo; conclusion, not PLAPmediated Bidirectional transcytosis of receptor and Tf BL-A via FcRn

587, 588

TRANSCYTOSIS

83 JULY 2003

Primary endothelial cells

Human placenta

14

Primary culture

Adult pig

Expression of thyroid-specic genes

A-BL movement of intact protein via megalin-mediated endocytosis and avoidance of lysosome Injected into colloidal space

351

www.prv.org

Same

Human

TER of 1,500

227, 228 405 Biotinylation 636

FRT

Fischer rat thyroid

FRTL-5

Clone of FRT

Polarized but no thyroid-specic gene expression; TER of 10,000 Question about polarity

Megalin-mediated transcytosis

348, 349, 352

Mammary alveolar epithelial cells

Lost expression of tissuespecic genes (e.g., whey protein)

Two cell layers with phenotypes similar to alveolar and myoepithelial cells; TER of 20010,000; 6-fold BL-A over A-BL transcytosis Indirect route from TGN-BL-A of hemagglutinin in infected RPE-J cells

497

RPE-J

Rabbit midpregnant gland immortalized with SV40 Retinal pigment epithelial cells immortalized with ts-SV40

TER of 300

45

891

892
Reference No.

PAMELA L. TUMA AND ANN L. HUBBARD A, apical; BL, basolateral; TSH, thyroid stimulating hormone; RAGE, receptor for advanced glycation end products; SR, scavenger receptor; TER, transepithelial electrical resistance; TMD, transmembrane domain; PAF, platelet activating factor.

Transcytosis Studies

Late endosome to functional secretory domain, which was identied as apical based on delivery of hemagglutinin to it Quantitatively little transported from dendrite to axon

Must have been apical to basolateral need to look in Ohkura, 1990; avoidance of lysosomes

molecule can change its biological properties, we assert that one must determine if those changes have altered the molecules qualitative or quantitative behavior. Unfortunately, many researchers assume that the modied molecule is normal, which can lead to erroneous or, at the least, uncertain results. In the remainder of this section we highlight in vitro cell systems currently used for studies of transcytosis. Table 4 presents a partial list. We particularly focus on the extent to which a particular cell line being used and the cargo being followed represent good physiological models. In several cases, there is the need for development and/or improvement of cell models. B. Microvascular Endothelial Cell Models
Downloaded from physrev.physiology.org on November 4, 2009

398, 490

Comments

418

Degraded bone products; Ca2

221

Cultured endothelial cells originating from various tissues and vessels abound (155), but few have been used to study transcytosis in vitro. Why? There seem to be two reasons. First, primary endothelial cells show multiple changes after isolation, including a decreased number of caveolar proles, increased permeability (101), and even transformation from a continuous to fenestrated phenotype (477). These phenomena point to the plasticity of the cells and reinforce the notion that local context dictates the physiology of the vascular bed (538). A second reason is that physical forces (osmotic pressure, ow) in vitro are totally different from those in vivo. Nonetheless, selected aspects of endothelial cell transcytosis have been successfully studied in vitro (Table 4). For example, mechanistic aspects of the albumin receptor, which stimulates uid-phase transcytosis, have been studied in vitro by Malik and colleagues (13, 89, 379, 576, 602 and discussed below). Interestingly, although the binding/uptake of modied LDLs is commonly used to characterize endothelial cells, and even has been used to purify them (606), to our knowledge there has not been a comprehensive in vitro analysis of the dynamics of the different scavenger receptors (558) or their cargoes. Are the diverse modied LDLs degraded via the lysosomal system and/or transcytosed? Are there regional differences among endothelial cell responses to modied LDLs in vivo? A fruitful area for future research might be to map the surface distributions of the endothelial scavenger receptors, LOX and CD36 (caveolae or clathrin-coated pits?), and determine the intracellular/transcellular fates of the cargoes. Considerable work has been done (2, 36, 108, 296, 326, 399, 461, 496, 606), but we still lack an understanding of the role played by the endothelium in handling atherogenic particles. Several reasonable in vitro models of brain endothelial cells have been reported (105, 113, 201). Kim (286) has used primary cells and secondary lines from rat and human cerebral cortex to study bacterial adherence and
83 JULY 2003

Cargo LDL

Reference No.

417

0.4-m pore type IV bovine collagen-coated Millicell-CM lter; cells used 20 h after seeding

Culture conditions

Characteristics

Newborn rat skin

4Continued

Human, rat, and rabbit

Origin

Primary epidermal cells

TABLE

Osteoclasts

Hippocampal cells

Cell

28

Tf

Physiol Rev VOL

www.prv.org

TRANSCYTOSIS

893

penetration. One example is in Reference 407. Dehouck and colleagues (85, 114 116) developed a coculture system that appears to represent a good in vitro model of brain capillary endothelium. In brief, freshly isolated astrocytes from newborn rat cerebral cortex are plated on the bottom side of commercial lters (or the bottom of a dish) coated with rat tail collagen, cultured for 3 wk, at which time a subconuent monolayer of adult bovine brain endothelial cells is plated on the upper side of the lters. After 8 days, conuent endothelial monolayers have formed, and only those exhibiting 500 cm2 are used. In a series of articles, these researchers have characterized the coculture system and validated its use for in vitro studies of brain capillary permeability. The endothelial cells express all of the standard markers of brain endothelial cells, such as -glutamyl transpeptidase, Pglycoprotein, and glucose transporters. Furthermore, a comparison of drug transfer in vivo to that in this in vitro system comes out favorably. Several studies from this group deserve further comment. Earlier in this review, we referred to the conicting evidence for and against the quantitative transcytosis of plasma Tf when iron is delivered to the brain interstitium. Results from the in vitro brain endothelial system showed that Tf is transcytosed with iron to the abluminal (basal) side of the monolayer (122). However, the subsequent fates of both components still remain a mystery. The story of the brain endothelial cell LDL receptor is equally puzzling. In conuent endothelial cells (i.e., with underlying astrocytes), LDL is transcytosed in an apical to basolateral direction via LDL receptor residing in caveolae. In contrast, in growing cells, LDL is endocytosed via receptors in clathrin-coated pits, the protein components are degraded, and cholesterol is subsequently used by the endothelial cells themselves (111, 112). The molecular basis for this switch, from receptor-mediated endocytosis of LDL for intracellular utilization to receptor-mediated transcytosis of LDL for use by interstitial cells, is currently unknown. Its elucidation should yield important insights into the machinery required for each type of vesicle trafc. C. Epithelial Cell Models 1. Intestine Three cell models of the intestinal epithelium have been used to study the transcytosis of macromolecules ranging from endogenous apical PM proteins and toxins to bacteria (Table 4). Caco-2 cells are the most commonly used because they differentiate furthest along the cryptto-villus axis and are the easiest to transfect. As mentioned above, the intestinal differentiation program takes up to 20 days in vitro. However, even the most differentiated Caco-2 cells lack the thick glycocalyx expressed by
Physiol Rev VOL

mature enterocytes in vivo; this apical surface feature is considered by some to represent a functional barrier to the adherence and invasion of the intestinal mucosa by many pathogens (160). Therefore, its absence in Caco-2 cells needs to be considered when extrapolating from in vitro results to in vivo possibilities (8). Furthermore, the apparent bidirectional transcytosis of some ligands (e.g., of a transferrin-insulin conjugate, Table 4) may represent reduced polarity in vitro. Interestingly, by coculturing lymphocytes in the basal medium with Caco-2 cells on a lter, the latter have been induced to express M cell-like morphology and activity (284). This achievement has exciting implications for the study of pathogen invasion in vitro (513). Lets hope that the model is further developed. HT29 cells can also differentiate along the enterocyte pathway to a mid-mature state, and they additionally have the capacity to differentiate into goblet cells (637). However, HT29 cells are not as easily transfected as Caco-2 cells, nor do they form as tight a monolayer; thus the latter cells are preferred for studies of transcytosis in vitro. The third intestinal cell model, T84, is arrested at an immature enterocyte stage, similar to a crypt cell in vivo. They have been used recently to study IgG transcytosis (Table 4). Additional in vitro models are under development (e.g., Ref. 425a). 2. Liver The dissociation of liver tissue into isolated cells that were suitable for culture was rst described over 30 years ago (37). However, the loss of structural polarity and mixing of membrane domains made them poor models for studies of polarity. The development of isolated couplets was a signicant advance for short-term studies of polarized hepatocyte functions, such as transcytosis and bile formation (50). As the importance of the extracellular matrix in maintaining gene expression and promoting cell repolarization became apparent, investigators began culturing hepatocytes on different matrix components in various geometries and physical states. The most successful reconstitution of polarity has come from the use of collagen gels or Matrigel in a sandwich conguration (194, 328). Despite these advances, the limited lifespan of primary hepatocytes and the difculties in reproducibly obtaining such polarized cultures prompted the use of secondary hepatocyte cell lines. A) WIF-B CELLS. Cassio and colleagues (83, 84) have generated many somatic cell hybrid lines in their studies of the genetic basis of liver-specic gene expression. The WIF-B cells that we currently use were generated by fusion of the differentiated (i.e., expressing liver-specic genes) but nonpolarized rat hepatoma cells (Fao) with human skin broblasts (WI-38 cells). Clonal selections ultimately yielded a polarized hepatic phenotype, the WIF121 cells, which exhibited a maximum apical polarwww.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

894

PAMELA L. TUMA AND ANN L. HUBBARD

ity of 40%; that is, a maximum of 40% of cells in a mature culture formed apical poles (or cysts) with adjacent cells. In collaboration with Cassio, we performed further selections and generated the WIF-B cells, whose apical polarity index is 80% (521). Before using these cells for membrane trafc studies, we characterized their mature phenotype, especially with regard to cell polarity (251). Most proteins show the same distribution as in vivo, as does microtubule organization, at least near the apical PM (374). Microtubules radiate from the apical membrane, with actin and foci of -tubulin also concentrated in this region. The presence of -tubulin leads us to conclude that the minus ends of polarized microtubules are closest to the underlying apical PM. We also explored the structural and functional properties of the tight junction boundary (251). ZO-1, the tight junction-associated protein, marks the boundary between the two PM domains. Using short chain lipid analogs in living cells, we established that the tight junctions were an effective fence prohibiting the lateral mixing of outer leaet PM lipids. Surface-labeling of living cells with sNHS-LC-biotin (557 Da) indicated that small molecules had access to the entire PM, while streptavidin (60 kDa) was restricted to only the basolateral domain, establishing that the tight junctions also provided an effective barrier to the diffusion of large molecules into the apical space. B) HEP-G2 CELLS. The human hepatoma Hep-G2 is currently being used for studies of polarized membrane trafc (Table 4). This line, which expresses many liver-specic genes, was generated in the late 1970s from liver tumor biopsies in which the histology presented as a well-differentiated hepatocellular carcinoma with a trabecular pattern (3). The existence of bile canalicular-like cysts within and between cultured HepG2 cells was reported (90); EM observations and one apical PM marker were used in this study. Unfortunately, neither a systematic characterization of these cells nor the culture conditions giving maximum polarity have been performed. In addition to membrane protein trafcking, lipid trafcking has been actively studied in polarized HepG2 and to limited extent in WIF-B cells and hepatocyte couplets (102, 198, 213, 233235, 251, 364, 626, 627). This focus stems from the fact that the major exocrine function of hepatic cells is secretion of bile, whose principal nonprotein components are phosphatidylcholine (PC), cholesterol, and bile salts. To date, lipid analogs have been used in these studies. Given the recent identication of transporters and carriers implicated in cholesterol and phospholipid transport (reviewed in Refs. 46, 138, 150a, 256) and the interest in sphingolipids as possible organizers of lipid microdomains (reviewed in Ref. 237), our understanding of the molecular bases of the vesicular and nonvesicular transcytotic pathways between the basolateral and apical domains of polarized hepatocytes is cerPhysiol Rev VOL

tain to increase in the next few years. However, it is important to remember that results from studies using in vitro cell models and articial lipids as surrogates need to be conrmed by in vivo studies. One obvious feature of polarized hepatocytes is that they sequester their apical surface away from the substrate or bathing medium. This structural organization prohibits direct access to the apical PM, which is conned between adjacent cells and bounded by tight junctions. Hence, quantitative determinations of the polarity index, TER, or rates and extents of PM protein transport to or between the two PM domains using accepted methods (e.g., surface biotinylation as in simple epithelial cells) are not feasible. However, Hoekstra and colleagues (596) have reported methods for differentially measuring uorescent lipid analogs present at the two PM domains of Hep-G2 cells. We have developed methods in WIF-B cells for detection of trafcked antibodies at the apical PM (250) and proteins secreted into the apical lumen (34). 3. Kidney As discussed in section II, there is little evidence for in vivo transcytosis of macromolecular cargo in kidney. Nonetheless, MDCK cells, which are derived from dog kidney, are the most-studied epithelial cell model and have been used extensively to study transcytosis (Table 4). These cells were originally developed by nephrologists for permeability and electrical studies. Their subsequent use by cell biologists (86) for studies of the formation of tight junctions, establishment of polarity, and vesicle trafc have popularized MDCK cells to the point that they are now the NIH-3T3 broblast of the epithelial eld. An advantage is that MDCK cells are easily cultured, easily transfected, and become polarized 35 days after seeding. They were used in the now classical studies showing that enveloped viruses bud in a polarized fashion and that the newly synthesized viral membrane glycoproteins are targeted directly from the TGN to the appropriate PM domain (480). Furthermore, much of our current understanding of the IgA transcytotic pathway and the sorting signals in the pIgA-R comes from the elegant studies performed in MDCK cells by Mostov and colleagues (393, 394). We expand on this latter topic in section IV. Two MDCK strains with very different features were identied some time ago (Table 4). The MDCK I cell has a high TER and characteristics reminiscent of the renal collecting duct, whereas the more commonly used MDCK II strain, whose TER is one order of magnitude lower than that of MDCK I cells, has phenotypic features closer to those of the renal proximal tubule. Our own anecdotal experience with MDCK II cells indicates to us that there are many cell variants, some well-documented (367) and others not. Tsukita and colleagues (166) recently reported that stable expression of exogenous claudin 2 in MDCK I
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS

895

cells lowers its TER to that of MDCK II cells, which express endogenous claudin 2. Does the expression of this tight junction family member in MDCK II cells induce morphological and biochemical changes beyond those contributing to the change in TER? The rst comprehensive and quantitative analysis of apical and basal endocytosis in polarized epithelial cells was performed by Simons and colleagues on MDCK I cells (44, 605). In that study, uid-phase endocytosis from the apical PM was much less robust than from the basolateral PM. However, apically endocytosed material was transcytosed basally to a relatively greater extent than the converse (material taken up from the basal surface transcytosed to apically). Without knowing the equivalent in vivo endocytic and transcytotic extents at the two poles, this information is hard to interpret. The apical-to-basolateral transcytosis of micronutrients by the proximal tubules of the kidney in vivo makes this topic appropriate for study in kidney cell lines. An important requirement is that the cells express the apical scavenger receptors cubilin and megalin, which have been implicated in the retrieval mechanism (Table 4). Unfortunately, MDCK cells appear not to express these receptors while OK and LLC-PK cells do. The latter cells have been used for studies of vesicle trafc (109, 334). We favor the rat proximal tubule line (IRPT, Table 4), because it expresses functional levels of megalin, and useful reagents are available to rat proteins. 4. Additional epithelial cell systems Both primary cells and cell lines, alone and in coculture with endothelial cells, are being used to study transcytosis (Table 4). Calu-3 cells, derived from the upper airways, form tight monolayers and have been found to express and secrete IgA. It would be interesting to know if they also express FcRn and transport IgG in a polarized fashion. Could they be useful for therapeutic drug delivery studies? Likewise, the fact that alveolar type II cells are capable of trans-differentiating into type I cells under dened conditions may make it possible to sort out the confusion over which cell type, if either, is able to transcytose albumin from the thin epithelial uid layer in the alveolar lumen to the interstitium. Although type I cells contain a huge number of caveolae, Malik and colleagues (267) have recently reported that it is the type II cell that transcytoses albumin via the gp60 membrane protein and caveolae. B) PLACENTA. Both primary cells originating from protease dissociation of human term placenta and a human placental cell line, BeWo, have been used in in vitro studies of placental transcytosis (Table 4). Under appropriate culture conditions, these preparations differentiate from single cell cytotrophoblasts to multicellular syncyA) LUNG.

tiotrophoblasts. However, the variable TER values reported suggest that different culture conditions are being used that could affect the monolayer integrity of the BeWo cells (Table 4); furthermore, the cell lines delity to syncytiotrophoblasts in vivo is still not totally characterized. Nonetheless, two predominant in vivo cargoes, IgG and iron, are being successfully studied and the molecules involved in their transcytosis identied. B12 and cholesterol are also supplied maternally, yet have not been studied in the isolated placenta. Clearly, there is fertile ground for future work in this system. C) THYROID. Culture of primary thyroid follicular cells has been reasonably straightforward (Table 4). Additionally, a rat thyroid cell line, FRT, was generated and several sublines exist, with varying degrees of thyroid-specic expression. Both endogenous PM protein and thyroglobulin pathways have been studied with interesting results; that is, Zurzolo et al. (636) reported that immature FRT cells use transcytosis to deliver apical PM proteins, while mature cells use the direct TGN-to-apical route. The mechanistic differences in the two pathways could be identied using such model. D) MAMMARY. Although it is important to explore the mechanism by which plasma constituents reach the milk, the culture of primary mammary epithelium from lactating glands for such studies has not been reported (382). Because these cells contain and secrete fat globules, they are apparently very difcult to establish in culture and maintain in a differentiated state. D. Transcytosis Outside of the Epithelial World 1. Bone-resorbing osteoclasts In the nal part of this section, we present a fascinating in vitro transcytotic system that probably operates in vivo but has not yet been conrmed. Bone is a dynamic tissue that it is constantly being laid down and resorbed. Bone degrading cells, osteoclasts, become polarized under appropriate hormonal stimulation and set up an elaborate, sealed structure, called a lacuna, against a segment of bone (572, 585). The cells then secrete hydrolytic enzymes and acid into this extracellular lacuna, which is lined by a rufed border with membrane markers of lysosomes/late endosomes. Inside the lacuna, the bone mineral content (principally Ca and phosphate) is solubilized and collagen I is partially degraded. These components are then endocytosed at the rufed membrane front, transcytosed across the cell to a secretory zone, an apical PM domain in the middle of an otherwise basolateral PM (398, 490), and secreted into the marrow cavity. Subsequently, the products are carried into the circulation. Since these cells lack tight junctions, a morphological hallmark in epithelial cells, the system is ideal for studying the establishment of a dynamic polarity in the absence of juncwww.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

Physiol Rev VOL

83 JULY 2003

896

PAMELA L. TUMA AND ANN L. HUBBARD

tional elements. Furthermore, the transcytotic pathway in these cells differs from that of intestinal epithelial cells, because the cargo is carried from a late endosome (the rufed border) to the apical PM. 2. Neurons It is relevant in this section to discuss the possible existence of transcytosis in nerve. Early studies in cultured neurons suggested that the dendritic and axonal PM domains in this polarized cell are functionally equivalent to the respective basolateral and apical PM domains of epithelial cells (544). Carrying this analogy a step further, Dotti and co-workers (221) asked if transcytosis also occurred. Using hippocampal cultures, the researchers found evidence for limited transcytosis of Tf and its receptor from dendrites to axons (221). Given the subsequent work on targeting of native and foreign PM proteins (68, 264, 533), it seems that transcytosis is a minor activity in vitro; its occurrence in vivo is questionable. IV. MORE ABOUT TWO DIFFERENT TRANSCYTOSIS SYSTEMS A. Caveolae-Mediated Transcytosis Transendothelial transport differs in several ways from other types of transcytosis. The transport is rapid (30 s), the cargo is predominantly uid not receptorbound, and a unique vesicle acts as the shuttle between apical and basal surfaces. Despite these differences, the targeting and fusion machinery are reported to be similar to those used elsewhere (see sect. V). In this section, we briey summarize early work then focus on recent developments that raise new questions about caveolae-mediated transcytosis in endothelium. 1. The endothelial cell surface The blood-endothelial cell interface is complex in composition and dynamic in its functions (Fig. 5). The apical PM outside of caveolae is negatively charged and thus capable of repelling negatively charged blood cells. Early in vivo ultrastructural studies of endothelium throughout the vascular tree established that sialo-glycoconjugates and glycosaminoglycans (principally heparan sulfate proteoglycans) provided the negative charge barrier. Combinations of enzymatic treatments, lectins, and tracers of different pI values were used (537, 540). Clathrin-coated membranes and pits along the apical and basal surfaces of endothelial cells were also negatively charged. Thus cationic molecules were found to preferentially bind at these sites with subsequent delivery to and degradation in lysosomes (536). In contrast, anionic molecules, which
Physiol Rev VOL

include most plasma proteins, were excluded from this pathway. Instead, they were included in the uid internalized by apical PM caveolae, shuttled to the basal side, and released into the interstitium. 2. Caveolae and caveolin As stated earlier and shown in Figure 1, caveolae are ask-shaped pits present on and continuous with the apical and basal PMs of all endothelial cells. Although nearly ubiquitous, they are most abundant in terminally differentiated cells, such as adipocytes, smooth muscle cells, type I pneumocytes and, of course, endothelial cells throughout the vascular tree. Even in the early 1980s, endothelial cell caveolae were recognized as chemically distinct microdomains of the PM (Fig. 5). Numerous excellent reviews cover caveolar structure and function in greater detail than we can (9, 546, 553a). Our focus is on their role in transcytosis. As stated in section II, there is still considerable debate over whether caveolae mediate transcytosis in endothelial cells. Perhaps much of this conict can be put to rest now that caveolin-1-decient mice have been generated (130, 465). In animals lacking caveolin-1, the major structural component of caveolae in many tissues including endothelia, no identiable PM-associated caveolae were observed. In cultured broblasts from transgenic embryos, internalization of albumin was inhibited whereas transferrin uptake by clathrin-coated vesicles was not changed (465). Furthermore, and perhaps most signicantly, albumin delivery from blood vessel lumens to the underlying interstitium was completely inhibited in perfused transgenic mice (512). Morphologically, goldlabeled BSA was found only in the lumen of caveolin-1decient blood vessels, whereas in control endothelial cells, the marker was concentrated at caveolar PM invaginations and in internalized caveolae that were detached and closed from the PM. In aortic ring segments isolated from control or caveolin-1-decient mice, iodinated albumin transcytosis was measured. Robust transport was seen in control mice, whereas transcytosis was virtually eliminated in the knock-out mice. Interestingly, no accumulations of the tracer during longer incubations were observed in the transgenic mice, suggesting that albumin was not being internalized via clathrin-coated vesicles. However, altered extravascular oncotic pressure normally associated with albumin transcytosis defects was not observed in these animals (130), nor was there any change in cerebrospinal uid albumin concentrations, a transport pathway thought to be caveolae mediated. These results along with the surprising result that these animals are viable and fertile suggest other compensatory transport processes must be present. Nonetheless, the data strongly indicate that caveolae do indeed mediate transcytosis in endothelial cells.
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS

897

Downloaded from physrev.physiology.org on November 4, 2009

FIG. 5. Transendothelial transport pathways. A: the four transport pathways present in endothelial cells are depicted. Also shown is the preferential distribution of anionic sites on the endothelial cell surface and basal lamina. B: the ask-shaped plasma membrane invagination, the caveola, contains the oligomeric coat protein caveolin. C: an enlarged version of the indicated region in B is shown. Caveolin forms a hairpin structure in the bilayer with both its carboxy- and amino-terminal ends facing the cytoplasm. Three palmitoyl chains attached to the caveolin carboxyterminal tail insert into the bilayer. Caveolin is present in membranes enriched for glycosphingolipids and cholesterol. The glycosphingolipids are asymmetrically distributed to the outer leaet while cholesterol is present in both leaets where it is thought to ll spaces around the glycosphingolipid headgroups. Although the phospholipids present on the cytoplasmic face have not yet been identied, their fatty acid chains may be saturated to optimize packing. cp, Coated pit; CV, clathrin-coated vesicle; cav, caveolae; ch, channel.

Despite these major advances, there are still many unresolved issues. Although caveolae are the likely transcytotic carriers, it is not clear how they transport molecules across the endothelial cell layer. Do caveolae, like clathrin-coated vesicles, pinch off from the PM, travel the short distance across the cell, and fuse with the opposite surface? N-ethylmaleimide (NEM) sensitivity of transport and the identication of caveolae-associated dynamin and specic members of the vesicle docking and fusion machinery (see sect. V) are consistent with this idea. In particular, Schnitzer and colleagues (416) have reported that dynamin activity leads to the scission of caveolae from endothelial PM. Also, EM of albumin-containing free
Physiol Rev VOL

caveolae and caveolae associated with both the basolateral and apical surfaces of the cell are consistent with caveolae as vesicular shuttles (183185, 378). Alternatively, caveolae on opposing membrane surfaces may transiently (?) fuse with each other producing channels through which ligands pass (Fig. 4). Such structures that contain transcytosing albumin have been occasionally observed at the ultrastructural level (183). One possibility is that both mechanisms operate but mediate the transport of specic ligands or ligands differentially modied. Since more caveolae are observed than the predicted number of pores, the question as to whether all caveolae are transcytosis-competent is also raised. Are some caveolae or
www.prv.org

83 JULY 2003

898

PAMELA L. TUMA AND ANN L. HUBBARD

channels reserved for transport while others serve as signaling centers? If so, how are they different? 3. Albumin and orosomucoid transcytosis Although most transcytosed cargo in endothelial cells is uid, two of the best-studied molecules, albumin and orosomucoid, are transported predominantly via receptor-mediated mechanisms. The rst clues that albumin was internalized via receptors came from morphological studies where albumin association with endothelial cells was concentrated in caveolae open to blood vessel lumens (183, 184, 378). Three major albumin binding proteins were subsequently identied (p18, p31, and p58 60) that were present on endothelial luminal cell surfaces (179, 180, 507, 508). Further work revealed that p18 and p31 were likely scavenger receptors and that gp60 was the bona de albumin receptor (506, 575). Albumin binding to gp60 and gp60 antibody cross-linking activates the transendothelial transport of albumin in vitro and in vivo (602). In both cases, the tyrosine phosphorylation of gp60 itself, caveolin-1, and the tyrosine kinases pp60 src and Fyn were induced upon activation. Addition of the specic tyrosine kinase inhibitors herbimycin and genistein blocked albumin internalization, indicating that transcytosis is a tyrosine kinase-dependent process (576). More recently, this activation has been further correlated with a Gi-coupled src kinase signaling pathway that is discussed below (see sect. VD1). Like albumin, when examined ultrastructurally, transport of orosomucoid is rapid and occurs via caveolae (456). Previous studies showed that orosomucoid binding to cultured endothelial cells was saturable, specic, and high afnity, implying the presence of specic receptors (504). Ligand blotting of endothelial cell lysates with 125Iorosomucoid identied three receptor candidates of 14, 20, and 7 kDa that have not been examined further (456). Like albumin, orosomucoid transcytosis is also NEM sensitive, consistent with the identication of caveolae as vesicle shuttles between the two domains during transcytosis (456). Because none of these possible receptors or gp60 has been cloned, the structural signals that direct transcytosis of these molecules and their associated ligands have not yet been identied. Will there be any similarities to those identied in pIgA-R (see below)? What dictates their placement in caveolae over clathrincoated pits? B. Clathrin-Mediated Transcytosis As discussed in section IIC3, one of the best-studied examples of transcytosis emerged from efforts aimed at understanding the molecular basis of mucosal immunity. Here we will discuss in detail the transcellular itinerary of the pIgA-R and its ligand, pIgA. Also,
Physiol Rev VOL

we will describe what is understood about the signals encoded in the cytoplasmic tail of the receptor that direct the complexs circuitous journey. Finally, we will compare the pIgA-R transcytotic pathway with that of newly synthesized hepatic and intestinal apical PM residents. 1. The pIgA-R transcytotic pathway The intracellular itinerary that pIgA-R follows during its life cycle is complex and may vary among epithelial cell types examined and among species (388, 439). Nonetheless, from studies performed in hepatocytes, enterocytes, and their respective in vitro model systems as well as from MDCK cells that stably express exogenous pIgA-R, a general picture is emerging that is summarized in Figure 6. From work performed in rat hepatocytes (565), it was determined that pIgA-R is synthesized in the ER as a 105-kDa polypeptide and travels to the Golgi where it is terminally glycosylated giving rise to an 116kDa glycoprotein. This form of the receptor is delivered from the TGN to the basolateral surface where it is further modied (probably phosphorylated, see below) reaching its mature form of 120 kDa. Early in vivo experiments, also performed in hepatocytes, further determined that the basolaterally located receptor is subsequently internalized (in the presence or absence of bound pIgA) by clathrin-coated pits and delivered to early endosomes (176, 238). Other receptor-mediated ligand systems (e.g., ASGP-R) were also present in these early endosomes, but only the pIgA-R and its ligand were delivered to a subapical compartment before their release into the bile. The other internalized receptor proteins moved along one of two other arms of the endocytic pathway, to lysosomes or back to the basolateral PM. The transcytotic pathway taken by pIgA-R in MDCK cells has been studied extensively with the goal of identifying and characterizing all of the intracellular intermediates. Debate over nomenclature and different experimental approaches led to conicting views that now appear to be resolved. Recent reports (316, 609) have established that at least three compartments comprise the basolateral-to-apical transcytotic pathway of pIgA-R in MDCK cells: basolateral early endosomes, a common endosome, and an apical recycling endosome (Fig. 6). In hepatocytes and enterocytes, there are only two identied transcytotic intermediates: basolateral early endosomes found in both cell types and a common endosome described in enterocytes or the subapical compartment (SAC) in hepatocytes (29, 250, 290) (Fig. 6). The basolateral early endosomes in the different cell types appear to be rather analogous structures. They are the rst entry points for molecules internalized from the basolateral surface, they are biochemically distinct from apical early endosomes (that
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS

899

Downloaded from physrev.physiology.org on November 4, 2009

FIG. 6. The intracellular itinerary of polymeric IgA receptor (pIgA-R) in hepatocytes, MDCK cells, and enterocytes. In general, pIgA-R is synthesized in the endoplasmic reticulum and transported to the trans-Golgi network, where it is sorted for delivery to the basolateral surface. At the basolateral surface, the receptor binds its ligand and is transcytosed through several intracellular intermediates to the apical surface. The receptor is cleaved by a cell surface protease releasing the pIgA-R ectodomain and bound ligand into the lumen.

are rst to receive apically internalized molecules), and morphologically, they are located at the basolateral pole of the cell. The MDCK apical recycling endosome is most similar to the hepatic SAC. Both have a neutral pH, only apically destined membrane components traverse them, and they are located closest to the apical PM. The major difference between them is the absence of recycling apical PM proteins in hepatic SAC and their presence in the MDCK apical recycling endosome. Why such a similar structure is not present (or not yet identied) in enterocytes is a puzzle. Also, why do hepatocytes lack a common endosome (that receives cargo from both cell surfaces)? These differences may be pointing to unique and important differences in membrane transport among epithelial cell types. Once the receptor has reached the apical cell surface, it is cleaved and its ectodomain is released into the luminal spaces. The receptor is constitutively synthesized and transcytosed whether ligand is present or not such that both the unoccupied SC and secretory IgA are recovered from apical secretions. In hepatocytes, the pIgA-R is
Physiol Rev VOL

cleaved efciently at the apical surface such that the pathway is one-way; all receptor molecules that reach the apical cell are clipped. However, in MDCK cells, pIgA-R proteolysis is less efcient such that a small proportion of intact pIgA-R at the apical cell surface can be recycled, and even a smaller portion can be transcytosed back to the basolateral surface (Fig. 6). Whether this is a physiologically meaningful process is not clear. These MDCK cells are overexpressing the rabbit form of pIgA-R, and it is possible that in the canine context, this receptor has altered dynamics. Examination of the dynamics of the very low levels of the endogenous MDCK pIgA-R may help clarify this point. Work performed in vitro in nasopharyngeal cells has shown that uncleaved apical pIgA-R mediates endocytic entry of Streptococcus pneumoniae, suggesting that apical internalization of pIgA-R may be part of its life cycle in certain cell types (273). Identifying the cell surface protease and understanding its proteolytic activity, specicity, and tissue distribution will help us clarify the posttranscytotic fate of the receptor in these other cell types.
www.prv.org

83 JULY 2003

900

PAMELA L. TUMA AND ANN L. HUBBARD

2. Signals and regulation of pIgA-R transcytosis The 103-amino acid cytoplasmic domain of pIgA-R contains multiple signals that mediate its circuitous journey. From work performed mainly in MDCK cells, the signals were found to fall into three categories: targeting to the basolateral PM, promoting rapid endocytosis, or preventing degradation (Fig. 7). With the use of deletion analysis, the pIgA-R basolateral targeting signal was identied in a sequence located just adjacent to the TMD, and this amino acid stretch was sufcient to direct chimeric reporter proteins to the basolateral PM (81). Site-directed mutagenesis of the receptor tail revealed that internalization from the basolateral surface required two cytoplasmic tyrosine residues, both of which are found in sequences similar to previously identied internalization motifs (421). Phosphorylation of serine-726 has also been shown to mediate rapid pIgA-R basolateral internaliza-

tion, and it has been suggested this modication is reected in the increased molecular mass (120 kDa) of the basolaterally located receptor (231, 422). Once internalized, the receptor is rapidly transcytosed across epithelial cells, and phosphorylation of serine-664 may facilitate this process. Also, a region of the tail has been shown to prevent the receptor from being degraded further, ensuring its efcient transcytosis once internalized (55). From these studies, it has been proposed that newly synthesized pIgA-R leaves the TGN in a nonphosphorylated form. Upon delivery to the basolateral surface, the receptor is phosphorylated (serine-726), which exposes the tyrosine-based internalization motifs allowing rapid internalization. Subsequent phosphorylation at serine-664 promotes transcytosis, thus sorting the receptor away from the degradative or recycling pathways (388). Transcytosis of pIgA-R is also regulated upon ligand

Downloaded from physrev.physiology.org on November 4, 2009

FIG. 7. The targeting signals encoded by the cytoplasmic carboxy-terminal tail of pIgA-R. A schematic drawing is shown summarizing the mutations made in pIgA-Rs cytoplasmic domain and their effects on receptor trafcking. From these studies, the indicated trafcking signals were identied. The more detailed versions of the basolateral targeting and internalization signals were identied in Refs. 19, 20, 421, 422. aa, Amino acid; BL, basolateral; TMD, transmembrane domain.

Physiol Rev VOL

83 JULY 2003

www.prv.org

TRANSCYTOSIS

901

binding, which signicantly enhances its transport from the apical recycling endosome to the apical PM in MDCK cells (548). Ligand binding rapidly causes the tyrosine phosphorylation of many proteins in MDCK cells including phospholipase C-1, from which a signal transduction cascade has been proposed (388). Similarly, pIgA-R transcytosis in intact hepatocytes is stimulated by intravenous injection of pIgA into rats (189). Although the accelerated transport step was not identied, clues to the activated tyrosine kinase came from studies performed in mouse hepatocytes (341). Coimmunoprecipitations showed that p62yes interacts with the pIgA-R tail in rat hepatocytes, and in p62yes knock-out mice, both basolateral to apical constitutive and activated transcytosis of pIgA were impaired (341). However, this mechanism may not be universal, since dIgA does not activate transcytosis in FRT cells that express exogenous pIgA-R (493). Also, the human forms of pIgA-R do not respond to ligand binding in polarized MDCK cells (all other studies were performed using the rabbit form), further suggesting that mechanisms regulating transcytosis differ among organisms. Careful comparison of cytoplasmic tail sequences may help pinpoint the reasons for the differential regulation. 3. Transcytosis of pIgA-R versus newly synthesized apical PM residents As described in section IIF, hepatocytes and enterocytes use the transcytotic pathway to deliver newly synthesized resident proteins to the apical surface. This indirect pathway includes transport of the newly synthesized apical proteins from the TGN to the basolateral PM where they are then selectively endocytosed and transcytosed to the apical PM (33, 499). Are the transcytotic pathways of the pIgA-R and apical residents shared? We tested this hypothesis by immunolocalizing newly synthesized apical PM proteins and pIgA-R in hepatocytes from bile-duct ligated rats, a condition reported to perturb hepatic IgA transport (29, 306). We found that both the residents and pIgA-R accumulated in the SAC. We next applied pulse-chase metabolic labeling combined with subcellular fractionation, vesicle immunoisolation, and immunoprecipitation of apical PM proteins to obtain more quantitative information about this intracellular compartment (30). We found newly synthesized dipeptidyl peptidase IV (DPPIV), a single TMD apical PM protein, rst in immunoisolated early endosomes and subsequently in isolated SAC. The high specic radioactivity of DPPIV (i.e., 35S-DPPIV/immunoreactive DPPIV) in this latter vesicle fraction indicated that very little preexisting (unlabeled) DPPIV was present. This meant that recycling of resident apical PM proteins was either very limited or did not involve SAC. The transcytosing molecules, although ultimately directed to the apical cell surface, must rst traverse the
Physiol Rev VOL

basolateral cell surface. Besides the basolateral targeting signal identied for pIgA-R, at least two other such signals have been identied. They either contain a tyrosine in the context of a short degenerate sequence or a dileucine motif (21), and in some cases, they overlap with those used at the basolateral PM in receptor-mediated endocytosis. In general, the tyrosine-based motifs are thought to be recognized by the -subunit of the Golgi tetrameric adaptor protein-1 (AP-1), whereas the leucine-based signals are thought to interact with the -subunit (361, 392). Identication of a new epithelial-specic 1-subunit, 1B, (157) has added a new twist to basolateral sorting. Its role in TGN to basolateral delivery was proposed when it was observed that introduction of 1B into LLC-PK1 kidney cells, which lack endogenous 1B, redirected its missorted LDL receptor (LDL-R) and Tf-R with high delity to the basolateral PM (157). Since these cells express the original 1-isoform, 1A, the 1B targeting appeared to be dominant (157). However, the specic transport step at which the 1B-subunit functions was not identied in this study. More recently, Gan et al. (173) reported that in the cells lacking the 1B-subunit, TGN to basolateral targeting of LDL-R and Tf-R occurred, but basolateral recycling was impaired, thus 1B sorts basolateral proteins postendocytically. This is consistent with the observation that basolateral targeting signals function both to deliver molecules to the basolateral domain, but also regulate basolateral internalization and recycling (for example, see Ref. 20). Despite where 1B functions, it is still a paradox that in liver that lacks 1B expression, the same basolateral receptors as in LLC-PK1 cells are delivered directly and with high delity to the basolateral PM. How do hepatocytes, without functional 1B/AP-1 complexes, sort proteins to the basolateral surface and maintain them there? What interprets the nontyrosine, nonleucine-based pIgA-R basolateral targeting signal? Also, how are the apical residents targeted to the basolateral surface in hepatocytes? Unlike pIgA-R, the cytoplasmic tails on several apical ectoenzymes that we have studied are very short (6 8 amino acids), with no identied or nonexistent (the GPIanchored proteins) signals suggesting that the mechanisms regulating transport must be quite different. A related puzzle is the mechanism(s) regulating internalization of the apical residents from the basolateral domain. Both newly synthesized pIgA-R and apical residents rapidly traverse the endoplasmic reticulum and Golgi and reach the basolateral PM within 45 min (32, 499). However, the rates of their redistribution to the apical PM vary greatly, which might reect differences in their modes of basolateral internalization. The different cytoplasmic tails may also reect different internalization routes, and in the case of the apical residents, these may likely be non-clathrin-mediated mechanisms. Alternatively, the proteins are internalized at the same rates and to the same extents. The variable trafcking kinetics inwww.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

902

PAMELA L. TUMA AND ANN L. HUBBARD

stead may reect differences in sorting or transport mechanisms elsewhere in the transcytotic pathway (e.g., basolateral early endosomes). V. MECHANISMS AND MOLECULES REGULATING TRANSCYTOSIS In this section, we discuss what is known about the mechanisms and molecules that regulate transcytosis in polarized epithelial cells. Are there unique mechanisms/ molecules required? Do different cell types require specic molecules or combinations of molecules to perform their specialized transcytotic functions? How does the cell discriminate between transcytotic delivery and self use? Surprisingly, there is little direct evidence for the involvement of specic molecules in transcytosis, but many promising candidates are emerging. We will describe some of these candidates, emphasizing those involved in vesicle transport, targeting, and consumption along the transcytotic route. However, a cautionary ag must be raised. Much of what is discussed is based on studies performed only in tissue culture models of polarized cells (especially MDCK cells) and has not yet been tested in a physiological context. Furthermore, many candidates have been named solely upon their subcellular location; the functional studies have not been performed. Nonetheless, there are exciting possibilities and the groundwork is clearly set to direct future investigations. A. Targeting Machinery 1. The SNARE hypothesis How do cargo-bearing vesicles deliver their contents to the correct target domain? Morphological, biochemical, and genetic approaches have successfully identied many molecules involved in vesicle targeting, a complex series of molecular events that minimally includes docking and fusion. These approaches indicate that the basic mechanisms are conserved among the membrane compartments throughout the biosynthetic and endocytic pathways and in organisms ranging from yeast to humans. The players so far identied fall into two broad categories: some are used repeatedly throughout the pathways, others belong to discrete protein families, where one or a few members act at one or a few transport sites. Members of three protein families are central players in vesicle targeting and fusion (216, 262, 335, 443). They are as follows: 1) the SNAREs which, in general, are a group of cytoplasmically oriented integral membrane proteins that are present on vesicles (v-SNAREs) or target membranes (t-SNAREs) (also referred to as Q- and R-SNAREs; see Refs. 613 and 144); 2) the Sec1/Munc18 proteins; and 3) small-molecular-weight GTP-binding proteins, the rabs.
Physiol Rev VOL

The SNARE hypothesis emerged to describe the mechanism by which the SNAREs promote membrane docking and fusion through interactions with an ATPase, N-ethylmaleimide sensitive factor (NSF), and its receptor, -SNAP (547), but has undergone considerable revision as more is learned about these and other essential proteins and the cyclical nature of the process is better appreciated (262). For vesicle trafcking to continue past one round, some of the machinery must continually cycle between vesicle and target membranes, making the order of events difcult to dene. Also, the t- and v-SNAREs, whose pairwise coupling was originally thought to confer targeting specicity, show promiscuity in their binding in vitro, suggesting that other factors are required for the high delity of membrane targeting observed in vivo. Furthermore, the ATPase, NSF, which was originally postulated to function in the fusion reaction itself, most likely functions as a chaperone to disassemble the SNARE pairs (204, 216). Whether this occurs when the pairs are in trans (on the cognate membranes) or in cis (on the same membrane after vesicle consumption) may differ depending on the cell type or transport step examined. It is also important to note that the SNARE hypothesis developed mainly from studies examining vesicle docking and fusion at the PM where SNAP-25 family members (t-SNARE isoforms) are required. Because these molecules are predominantly PM associated, the mechanisms regulating intracellular vesicle transport are likely different. Consistent with this is the nding that two t-SNAREs (both syntaxins) and two v-SNAREs are required during late endosome fusion, whereas at the PM, two different t-SNAREs (a syntaxin and SNAP-25 protein) and one v-SNARE participate (15). Interestingly, in both cases the molecules form core complexes that are structurally similar, indicating that the mechanisms, albeit different, are conserved. Newer models have integrated the enormous and confusing body of information and have identied common steps in membrane targeting which minimally include 1) SNARE activation (which likely includes NSF chaperone activity) and rab recruitment to proper organelle sites; 2) cognate membrane attachment; and 3) membrane fusion and bilayer mixing (262). The order of events and the mechanistic details are not clear, yet there is considerable evidence that the SNAREs, rabs, and Sec1/Munc18 proteins are key players in the process. In the following sections, we discuss the possible roles of selected members of the transport machinery in regulating transcytosis in polarized epithelial cells. Table 5 summarizes what we currently know. 2. NSF and -SNAP NSF was initially implicated as a regulator of polarized PM targeting from studies that examined the effects of NEM on protein transport. Addition of NEM reduced
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS

903

IgA transcytosis by 70% and its basolateral targeting by 90% in permeabilized MDCK cells (17, 332). It also inhibited fusion (as assayed by pIgA-R processing) of transcytotic carrier vesicles with the apical PM in a hepatic cell-free system reconstituting the nal step of transcytosis (564). Because NSF ATPase activity is inhibited by alkylating (617), it was the likely target for NEM. In support of that, addition of recombinant NSF restored much of the activity lost in MDCK cells or hepatic cell-free systems treated with NEM or anti-NSF antibodies (17, 253, 332, 564). Interestingly, direct apical PM targeting was insensitive to NEM (253, 332), indicating that it requires an unidentied NSF homolog or does not require NSF-like activity at all. Other key factors, the SNAP family of NSF receptors (-, -, and -isoforms), recruit NSF to organelles and activate its ATPase activity (617). -SNAP has been identied in polarized epithelial cells and its role in TGN to PM targeting examined in MDCK cells. Addition of -SNAP antibodies and treatment of cells with botulinum E (an -SNAP-specic toxin) inhibited direct transport to both domains (17, 253, 300). Unfortunately, its role in transcytosis was not examined. NSF has also been implicated as an important regulator of transcytosis in endothelial cells. Treatment of myocardial or rat lung endothelium with NEM signicantly inhibited transcytosis in situ (80 and 50%, respectively) (455, 505). NSF and its receptor -SNAP were also found associated with caveolae, supporting the role of this vesicle type as the transcytotic carrier in endothelial cells (509). Furthermore, immunoprecipitation of exogenously added recombinant myc-tagged NSF from rat lung endothelial cell extracts revealed the presence of large, ATP-dependent, NEM-sensitive complexes that contained many members of the transport machinery including - and -SNAP, cellubrevin, syntaxin, and rab5 as well as caveolin and dynamin (457). The complexes were also found to contain cholesterol, the ganglioside GM1, and other unidentied lipids. As predicted, these complexes were immunoprecipitated from membrane extracts, but they were also surprisingly recovered from cytosol preparations. How these supramolecular proteinlipid complexes function in membrane transport is not yet known but supports a role for both caveolae (the complexes contained caveolin) and the molecules of the SNARE hypothesis in endothelial transcytosis. Do the complexes present in the cytosol or on membranes serve the same function? Are similar complexes present in other polarized epithelial cell types? 3. t-SNARES and v-SNAREs There are two families of t-SNARE proteins: the syntaxins and the SNAP-25 family. To date, at least 18 syntaxin family members have been identied in mammalian cells of which 5 (syntaxins 1A, 1B, 2 4) are PM-specic
Physiol Rev VOL

isoforms (262). The SNAP-25 family is much smaller, with only three identied members: SNAP-25, SNAP-23, and SNAP-29 (262). Like the syntaxins, these proteins are relatively small (2329 kDa), cytoplasmically oriented molecules. Although not integral membrane proteins as are the PM syntaxins, SNAP-25 proteins associate with the bilayer through cysteine-linked palmitoyl chains located near the middle of the protein. Biochemically, SNAP-25 proteins bind PM-associated syntaxins and vSNAREs in vitro to form the four-stranded -helical ternary complexes required for vesicle docking and fusion (262). The distributions of syntaxins 2, 3, and 4 at the PM have been examined in different epithelial cells. In particular, we found that rat hepatocytes express three endogenous PM-associated syntaxin isoforms (syntaxins 2, 3, and 4) and SNAP-23 (163). Quantitative immunoblotting revealed that all four t-SNAREs are relatively abundant in liver (11 668 nM corresponding to 0.528 105 molecules/cell). Biochemically, each of the t-SNAREs was observed predominantly in hepatocyte PM sheets with overlapping but distinct expression patterns among the PM domains. Both syntaxin 4 and SNAP-23 are restricted to the basolateral PM while syntaxins 2 and 3 are more apically distributed, with greater enrichment of syntaxin 3 in this domain. Despite the biochemical abundance of the molecules, we were able to detect only syntaxins 2 and 4 in rat liver sections in situ. However, the distributions did not t with our biochemical data; we found syntaxin 4 in both domains. Similar discrepancies were observed in WIF-B cells. Like in liver hepatocytes, syntaxin 3 was at the apical domain, but unlike liver, syntaxin 2 was restricted to the apical domain. Also, syntaxin 4 and SNAP-23 were found in both domains. These varied distributions likely reect important differences in regulation of PM dynamics between the in vivo and in vitro systems and may point to interesting features of the cellular itineraries and functions of the t-SNAREs. Little is known how these putative targeting molecules are themselves targeted to the correct PM domain, and once delivered, how and if they are retained there. Interestingly, the PM syntaxins display remarkable variability in their domain distributions in other polarized cells (see Table 5). Only syntaxin 3 appears to be consistent with apical distributions observed in all polarized cells examined (117, 163, 170, 331, 438). SNAP-23 distributes to both PM domains in all but two epithelial cell types (see Table 5). Since it is thought to be required for vesicle docking and fusion, SNAP-23s uniform PM distribution ts with the current model of its ubiquitous involvement in t- and v-SNARE ternary complex formation. However, its absence at the apical PM in hepatocytes and pancreatic cells is somewhat paradoxical. Either these cells have unique mechanisms for regulating transport at
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

904

TABLE

5. Putative regulators of transcytosis in polarized epithelial cells


Epithelial Cells Examined t-SNAREs Location; syntaxin 3 overexpression in MDCK and Caco-2 cells and antibody sensitivity in MDCK cells Location Location; toxin sensitivity in MDCK and IMCD cells and anti-SNAP23 sensitivity in MDCK cells 59, 117, 163, 168, 331, 345, 438 Evidence for Role in Transcytosis Reference No.

Protein

Subcellular Location in Epithelial Cells [Morphology (M)/Fractionation (F)]

Syntaxin 2, 3, 4

Apical and/or basolateral PM (M, F)

Syntaxin 8, 13 Syntaxin 6, 10 SNAP 23 v-SNAREs Location; toxin sensitivity in MDCK and IMCD cells Location Location Location

BL early endosomes (F, M) TGN Apical and/or basolateral PM (M, F), endosomes (M, F)

Hepatocytes, pancreatic acinar and intestinal epithelial cells, kidney collecting duct, Caco-2, MDCK and WIF-B cells Hepatocytes, WIF-B cells N/D Hepatocytes, MDCK, WIF-B, and IMCD cells

562, 583 262 26, 88, 169, 255, 314, 333

VAMP 2

Endosomes (F)

27, 74, 253, 371 75 171 554, 622

VAMP 3/cellubrevin VAMP 7/Ti-VAMP VAMP 8/endobrevin Others Placenta, pinealocytes, MDCK cells

PAMELA L. TUMA AND ANN L. HUBBARD

Downloaded from physrev.physiology.org on November 4, 2009

Physiol Rev VOL


Murine intestinal, lung, kidney, testis and spleen epithelia, MDCK cells MDCK cells Rab proteins Anti--SNAP sensitivity in MDCK Location Location; overexpression in MDCK Location Hepatocytes Intestinal and kidney epithelia, hepatocytes, HT-29, and T84 cells Hepatocytes Kidney epithelia, hepatocytes, MDCK and WIF-B cells Hepatocytes MDCK Gastric parietal, pancreatic acinar and prostate epithelial cells, enterocytes, hepatocytes, and MDCK cells Small intestinal and kidney epithelia, hepatocytes, Caco-2 and LLC-PK1 cells Hepatocytes, enterocytes, kidney epithelia, MDCK, and Eph4 cells Kidney tubule epithelia Kidney tubule epithelia Gastric parietal cells, MDCK cells

Basolateral endosomes (M, F) Subapical structures (M), apical PM (M) Basolateral early endosomes (F, M), apical PM (M), subapical endosomes (M)

Gastric parietal, MDCK and IMCD cells Hepatocytes Caco-2 cells Hepatocytes, MDCK cells, and kidney epithelium

NSF

Munc 18-2

Golgi (M, F), endosomes (F), coated vesicles (F) Apical PM (M)

NEM and anti-NSF sensitivity and mutational analysis in MDCK Location; epithelial-specic expression

17, 253, 332, 359, 457, 509 472

83 JULY 2003

-SNAP

N/D

253, 509

www.prv.org

Rab1 and 2 Rab3B

Golgi (F), transcytotic vesicles (F) Tight junctions (M)

Copurication with transcytotic vesicles Location

265 595, 611

Rab3D Rab4 and 5

305 67, 272, 583 Location; TGN budding assay Location; sensitivity to rab8 inhibitory peptides Location; change in distribution upon parietal cell activation; mutational analysis in gastric parietal and MDCK cells 354 240 82, 134

Rab6 Rab8 Rab11

Apical PM (M) Apical and basolateral early endosomes, apical and basolateral PM (M, F) Golgi Golgi, basolateral PM (M, F) Subapical structures (M, F), apical recycling endosome (M)

Rab13

Tight junctions (M)

632

Rab17

165, 210, 247, 337, 631

Rab18 Rab20 Rab25

Basolateral PM (M), apical vesicles and tubules (M), transcytotic vesicles (F), apical recycling endosome (M) Basolateral PM (M), subapical tubules (M) Subapical tubules (M) Subapical structures (F, M), apical recycling endosome (M)

Location, epithelial-specic expression; mutational analysis and overexpression in MDCK and Eph4 cells Location Location Location; mutational analysis in MDCK cells

338 338 74, 82

TABLE

5Continued
Epithelial Cells Examined The exocyst MDCK cells Annexins Location 200 Evidence for Role in Transcytosis Reference No.

Protein

Subcellular Location in Epithelial Cells [Morphology (M)/Fractionation (F)]

rSec6 and rSec8

Tight junction (M)

Annexin II

Annexin IV

Apical and/or basolateral PM (M), early endosomes (F), subapical structures (M, F) Apical and/or basolateral PM (M, F), subapical structures (M) Location

Location; change in distribution upon activation of transcytosis with bile acids in hepatocytes Location

142, 149, 357, 509, 615, 620 274, 292, 356, 357, 365

Downloaded from physrev.physiology.org on November 4, 2009

Physiol Rev VOL


Intestinal and mammary epithelia, hepatocytes, MDCK cells Kidney, intestinal, uterine, tracheal and fallopian tube epithelia, T84 cells Hepatocytes Intestinal epithelia Enterocytes and MDCK cells MDCK cells Dynamins Endothelial and MDCK cells MDCK cells Motor proteins Intestinal epithelia, hepatocytes Pancreatic acinar cells, intestinal epithelia, hepatocytes, Caco-2 cells Intestinal epithelia, MDCK and WIF-B cells Intestinal epithelia MDCK cells MDCK cells

Annexin VI

260, 424, 509, 566, 615 357 618 150

Annexin VIII Annexin XIIIa Annexin XIIIb

Apical and/or basolateral PM (M, F), subapical structures (M, F) Apical PM (M) Apical PM basolateral PM (M) Apical PM, subapical structures (M, F)

Intestinal-specic expression; location Location; intestinal-specic expression Location; kidney-specic expression

TRANSCYTOSIS

83 JULY 2003

Dynamin-1

Caveolae, apical PM (M, F)

7, 416

www.prv.org

Dynamin-2

Apical and BL PM (M, F)

Location; GTP dependence, overexpression and mutational analysis in MDCK cells, anti-dynamin sensitivity in endothelial cells Location, GTP dependence, overexpression and mutational analysis in MDCK cells

Cytoplasmic dynein Myosin I

Golgi (F), endosomes (F) Zymogen granules (M, F), Golgi (F), apical PM (M, F)

MT dependence and orientation; NEM sensitivity Location, actin dependence, mutational analysis in Caco-2 cells

146, 415 25, 94, 136, 145, 440, 452

Myosin V

218, 304, 321 215, 218 315 270

Myosin VI RhoA Rac1

Apical PM (M, F), apical recycling endosome (M) Apical PM (M, F) Basolateral endosomes (M) Apical recycling endosomes (?)

Location, actin dependence, mutational analysis in MDCK cells Location, actin dependence Location, mutational analysis Location, mutational analysis

IMCD, inner medullary collecting duct; MT, microtubule; N/D, not determined; TGN, trans-Golgi network; PM, plasma membrane.

905

906

PAMELA L. TUMA AND ANN L. HUBBARD

their apical surfaces, or other SNAP-25 isoforms are yet to be identied. To date, the roles of the t-SNAREs in polarized PM targeting have been tested directly mainly in MDCK cells that were stably expressing pIgA-R and overexpressing wild-type syntaxins 2, 3, or 4 (332). Neither transcytosis nor basolateral transport of pIgA-R was affected in these cells. Similarly, in Caco-2 cells overexpressing syntaxin 3, no changes in basolateral protein targeting were observed (59). Together, these results suggest that the PM-associated syntaxins do not regulate these transport pathways or that other yet-to-be isoforms are involved. Alternatively, overexpression was not high enough to be inhibitory or does not negatively regulate these processes. However, overexpression of syntaxin 3 in MDCK and Caco-2 cells did lead to alterations in apical PM dynamics. In MDCK cells, a slight impairment (20%) of direct TGN to apical PM delivery of a chimeric pIgA-R molecule and IgA was observed as well as apical recycling (also 20%). Likewise in Caco-2 syntaxin 3 overexpressors, the direct apical targeting of sucrase-isomaltase and the apical secretion of -glucosidase was signicantly impaired. Furthermore, anti-syntaxin 3 antibodies inhibited direct targeting of hemagglutinin (HA) in MDCK cells, conrming a role for this syntaxin in apical delivery (300). Unfortunately, the effects on transcytosis of anti-syntaxin 3 or syntaxin 3 overexpression in Caco-2 cells were not examined. However, the role of SNAP-23 in transcytosis has been examined in MDCK cells by treating with SNAP-23/ 25-specic neurotoxins. In these cells, basolateral to apical transport of pIgA was inhibited by 30% (17), as was basolateral targeting of the receptor, but the TGN to apical targeting of HA was not affected (17, 253). More recent studies conrmed this result and found that toxin activity also impairs transferrin recycling (314). The direct involvement of v-SNARES in transcytosis has not been explored, but the obvious prediction is that they are required. VAMP 1, 2 and VAMP 3/cellubrevin are ubiquitously expressed, and the presence of VAMP 3/cellubrevin on endosomal structures in hepatocytes and VAMP 2 on endothelial caveolae has been reported (75, 371). VAMP 8/endobrevin is enriched in epithelial tissues and has been localized to the apical pole in kidney epithelium (622). Interestingly, in hepatocytes, this VAMP species was found to be enriched in basolateral early endosomal fractions, whereas in MDCK cells, it was localized to both apical endosomes and the apical PM (554). Whether this protein functions in basolateral or apical PM targeting (or both) is not yet known. In Caco-2 cells, another VAMP isoform, VAMP 7/TI-VAMP, is also localized to both the apical PM and in subapically located structures where it has been proposed to function in the later steps of apical PM delivery (171). Interestingly, this VAMP species has a long NH2-terminal extension that resembles a region in annexin XIIIb, another protein imPhysiol Rev VOL

plicated in regulating apical vesicle delivery (150). This sequence in annexin XIIIb encodes a lipid-binding domain, but whether VAMP 7/TI-VAMP shares this biochemical property is not yet known. A possible role for VAMP 2 in transcytosis has been suggested from studies in SLO-permeabilized rat lung endothelial cells using VAMP-specic neurotoxins (371). In cells treated with botulinum D and F, VAMP 2 cleavage occurred concomitant with the impairment of caveolaemediated cholera toxin B endocytosis. At the ultrastructural level, large, aberrant subplasmalemmal organelles were observed in treated cells, indicating that delivery of cholera toxin to intracellular intermediates (endosomes?) was impaired. Unfortunately, transcytosis was not examined in toxin-treated cells to determine whether VAMP 2 is a general regulator of caveolae-mediated internalization in endothelial cells. These ndings also expand the function of VAMP 2 to include regulation of endocytic transport, whereas previously, this v-SNARE was thought to function in exocytic membrane docking and fusion. Whether VAMP 2 functions in PM vesicle docking and fusion in endothelial cells has not yet been tested. Likewise, a possible role for VAMP 2 in endocytosis in other cell types may warrant further investigation. 4. Munc18 Munc18 homologs have been identied in systems from yeast to neurons and are thought to participate in multiple vesicle transport steps (262, 442). The 68-kDa mammalian Munc18 proteins peripherally associate with the PM through interactions with syntaxins; in vitro, they bind syntaxins 1, 2, and 3 with nanomolar afnity. Interestingly, Munc18 binding to syntaxins cannot occur when the syntaxins are bound to SNAP-25 proteins, suggesting that the different complexes form reciprocally. However, it is presently not known whether Munc18 isoforms play a positive or negative regulatory role in PM targeting. Mutational analysis of related proteins in yeast, Drosophila, and Caenorhabditis elegans all implicate Munc18 species as positive regulators whereas in vitro assays suggest the opposite (262, 442). As for most of the SNARE molecules, no direct evidence for the involvement of Munc18 proteins in polarized PM targeting exists. However, the Munc18 2 isoform is primarily limited to polarized epithelial cells (472). Furthermore, its expression seems restricted to the apical PM where it forms complexes with syntaxin 3 (471), a characteristic that suggests a unique function for Munc18 2 in vesicle delivery to the apical PM. 5. The rab proteins The rab proteins belong to the largest family of small molecular mass (20 30 kDa) GTP binding proteins. There are 11 known yeast isoforms and at least 60 rabs in
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS

907

mammalian cells (195, 354, 500). Examination of transfected cells either overexpressing wild-type or dominant negative mutant (usually the GDP-bound conformer) forms of various rabs either stimulate or inhibit protein transport and in some cases alter organelle morphology. Although their precise roles are not known, they have been proposed to function in one of three ways: 1) facilitating vectorial trafc via associations with the cytoskeleton; 2) regulating vesicle docking by recruiting effector molecules, thereby promoting the formation of molecular tethers; and 3) priming docking and fusion by activating SNARE molecules (195, 500). Given the large number of mammalian rabs and their varied distributions, it is likely that transcytosis in epithelial cells is regulated by multiple isoforms, but which ones? Rabs 3B, 13, 17, 18, 20, and 25 are preferentially expressed in epithelial cells (see Table 5), suggesting a unique function in polarized membrane transport. Although also expressed in nonpolarized cells, rabs 1, 2, 3D, 4, 5, 6, and 11 have also been implicated in regulating polarized PM transport. Of these 13 rabs, 9 have been localized to the apical pole: at the apical PM (rab3D), the tight junction (rabs 3B and 13), or in subapical structures (rabs 5, 11, 17, 18, 20, and 25). The multiple rab proteins in the apical region may point to the complexity of membrane transport events at this PM domain both in terms of specic transport steps as well as organellar intermediates. Rab5 is the most extensively studied isoform, and much is known about the relationship between its catalytic activity and function in membrane transport (478). In all nonpolarized cells examined to date, rab5 is localized to the PM, clathrin-coated vesicles, and/or early endosomes. Overexpression of rab5 increased endocytic transport and stimulated early endosome fusion in vitro, whereas inhibition of rab5 led to the opposite effects. Recently, it was proposed that rab5 recruits EEA1 (one of its many effectors) to sites of endosome fusion along with NSF and syntaxin 13 that together drive formation of a large multimeric complex which then coordinates fusion pore assembly (368). This general role of rab5 in endocytosis strongly suggests an important role in the early steps of the transcytotic pathway in most polarized cells. Consistent with this is the presence of rab5 on early apical and basolateral endosomes in hepatocytes, hepatic WIF-B cells, in mouse kidney epithelia and MDCK cells (67, 272, 583). Furthermore, when overexpressed in MDCK cells, increased rates of uid-phase internalization from the apical and basolateral PM were observed, suggesting a role for rab5a in endocytosis from both domains (67). In endothelial cells, rab5 is also localized to the PM and early endosomes, suggesting a role in endocytosis. However, it is not found associated with caveolae, suggesting it is not required for endothelial transcytosis (509). Rab17 has been strongly implicated in regulating
Physiol Rev VOL

transcytosis in a number of different epithelial cells, rst because of its restricted expression pattern and subcellular location, and more recently from functional studies. In kidney, rab17 expression was induced only upon mesenchymal differentiation to polarized cells (337). In intestinal tissue sections, it was detected only in polarized cells and not in surrounding, nonpolarized cells. In enterocytes, rab17 was detected in the basolateral domain, whereas in kidney cells it was found both at the basolateral PM and in apical tubules underlying the brush border (337). In MDCK and polarized Eph 4 cells, it was detected in subapically located vesicular structures (247, 631), and in hepatocytes, rab17 copuried with transcytotic vesicles (210). Functionally, the overexpression of wild-type rab17 in MDCK cells impaired the basolateral to apical transcytosis of dIgA (247). Conversely, in Eph 4 cells expressing GTPase-decient mutants, the basolateral to apical transcytosis of Tf-R and a chimeric receptor was enhanced as was apical recycling of the chimeric receptor (631). Rab17 has also been copuried with a population of transcytotic vesicles from rat liver, suggesting it is an important regulator of hepatic basolateral to apical transcytosis, too. Surprisingly, both rab1 and -2 also copuried with the vesicles, implicating them as additional regulators of transcytosis (265). The next steps will be pinpointing the site of function in the transcytotic pathway, examining whether the role of rab17 is universal among epithelial cells and to identify the cellular effectors that rab17 activity regulates. Rabs 11 and 25 have also been identied as important regulators of basolateral to apical transcytosis from functional studies performed in gastric parietal and MDCK cells (82, 134, 610). Both of these rabs have been localized to the apical recycling endosome in MDCK cells (82), and when the respective GTP-binding mutants were overexpressed, basolateral to apical transcytosing IgA accumulated in these structures (610). Interestingly, the activated form of rab25 inhibited transport more than the rab11 mutant. Also interesting is the observation that unlike for nonpolarized cells, rab11 is not required for Tf recycling (610). The puzzle is why these rabs apparently regulate the same transport steps at the apical PM. Do the differential responses suggest separable roles in transport? Also, why is rab11 required for Tf recycling in nonpolarized cells, but not polarized cells? Although rab25 expression is enriched in epithelial tissues, it is surprisingly absent in liver (193), pointing out yet another important difference in apical PM targeting in polarized hepatocytes. The recent identication of two rab11 effectors, rip11 and myosin Vb, has further conrmed a role for this rab in regulating late steps in basolateral to apical transcytosis (304, 458). In both cases, dominant negative mutations signicantly impaired IgA apical PM delivery, and corresponding accumulations of IgA in the apical recycling endosome were observed in MDCK cells. Like rab11, mywww.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

908

PAMELA L. TUMA AND ANN L. HUBBARD

osin Vb was found to regulate Tf recycling only in nonpolarized cells (304) and rip11 mutants did not impair basolateral transferrin recycling (458). Are different rab11 effectors required for transferrin recycling in nonpolarized cells? How are these two effectors both regulated by rab11 at the same transport step? Careful dissection of the molecular events required for vesicle budding from the apical recycling endosome, transport to the apical PM, and subsequent docking and fusion are required to specically identify how these molecules function. The expression patterns of rabs13 and 3B are also highly dependent on the polarized state of a cell (611, 632). In nonpolarized cells, they were found in cytoplasmic vesicles, whereas in polarized cells, they were recruited to tight junctions. Rab13 has so far been detected at tight junctions of Caco-2 cells, mouse intestinal cells, kidney, and liver (632) while rab3B has been observed in tight junctions of colonic epithelia, kidney, and liver (611). In both cases, their localizations were dependent on the integrity of the tight junction. When junctions were disassembled by Ca2 withdrawal, staining of rabs13 and 3B at the cell surface was lost, suggesting these rabs function in vesicle delivery to the tight junction and, in particular, regulate vesicular delivery of junctional components. At present whether any transcytotic vesicles are also specically delivered to the cell surface at sites of cell-cell contact, and by extension, under the control of these rab isoforms, is not yet known. Immunoadsorption and examination of the vesicles with which these rab isoforms are associating will provide important clues to their function. Interestingly, in a recent report (595) overexpressed, myc-tagged rab3B was localized not to tight junctions in MDCK cells, but to apically located structures that also contained unoccupied pIgA-R. At present, there are no explanations for this different staining pattern. Nonetheless, in the presence of dIgA, rab3B dissociated from pIgA-R and the GTP-bound mutant rab3B impaired dIgAactivated transcytosis to approximately control (-dIgA) levels. Together these results suggest that rab3B is a negative regulator of ligand-stimulated transcytosis in MDCK cells. It will be interesting to see if rab3B also regulates other forms of ligand-activated transcytosis in other cell types and whether the sites of delivery are at or near the tight junction. Because of the congestion of different rab isoforms in the apical and subapical region of the polarized epithelial cell, it is important to identify the specic intermediates participating in basolateral-to-apical transcytosis. How many different subapical compartments exist, and of those, which receive transcytosing molecules? How do the rabs distribute among them? Furthermore, much of the morphological and functional analysis on the different rab isoforms has been performed in transfected cells overexpressing either wild-type or mutated proteins. AlPhysiol Rev VOL

though these studies provided insight into rab function, careful enumeration and examination of the endogenous rab isoforms in a single cell type is required to clearly understand their role in vivo. Nonetheless, the concentration of rab proteins in the subapical regions of epithelial cells is striking and may point to the complexity of intracellular compartments and membrane transport events at this PM domain. It remains to be determined whether other putative transport machinery molecules or rab effectors are also concentrated at the apical regions of cells. 6. The exocyst Over a dozen genes have been identied in yeast that are required for TGN to PM transport (275, 408, 409), and of these, more than two-thirds of the gene products form a multimeric complex referred to as the exocyst. The exocyst subunits were localized to the yeast PM, but only to sites of rapid cell growth at small bud tips (573). Interestingly, this expression pattern differs from the yeast t-SNARE molecules, which are evenly distributed at the PM. From these data, it was suggested that the exocyst mediates vesicle delivery to restricted regions of the cell surface such that it additionally discriminates (beyond SNARE function) whether and/or where a vesicle docks (573). Mammalian homologs to the exocyst subunits are ubiquitously expressed and also form a multimeric complex that is mainly peripherally associated with the PM (239, 282, 574). The mammalian counterparts of the yeast Sec6, Sec8, and Sec10 exocyst have been localized to tight junctions in MDCK cells, and their discrete staining patterns were dependent on intact junctional complex formation (200, 324). Anti-rSec6 antibodies blocked transport of LDL-R from the TGN to the basolateral PM in permeabilized cells, whereas direct transport of an apical PM antigen, p75, was not changed (200). Conversely, overexpression of mammalian Sec10 enhanced PM transport of E-cadherin and basolateral secretion, whereas apical delivery of the integral PM protein gp135 was unchanged (324). Surprisingly, Sec10 overexpression also enhanced apical secretion. Taken together, these results implicate the exocyst as an important regulator of vesicle targeting to both PM domains, but delivery to the apical domain may be restricted to vesicles carrying secreted cargo. The placement of the complex further implicates the tight junction as an important site for vesicle delivery to either domain. Whether transcytotic vesicles are specically recruited to these or other sites on the PM inhabited by the exocyst has not been rigorously examined, but remains an interesting possibility. 7. Annexins Annexins are a large family of proteins grouped together due to shared amino acid sequence similarity and
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS

909

their biochemical properties (103, 387, 563). One distinguishing feature of most annexins is their ability to aggregate membrane vesicles in the presence of Ca2 in vitro (103). This activity led to the idea that annexins initiate membrane-membrane contact that results in fusion. Of the numerous annexin isoforms, a few have been identied as playing roles in polarized membrane targeting, but many of those were implicated based only on their subcellular location (see Table 5). However, more direct evidence has been demonstrated for annexins XIIIa and XIIIb, the former being an intestinal-specic isoform and the latter, a kidney-specic isoform (150, 618). In SLOpermeabilized MDCK cells, addition of either of these annexin isoforms enhanced direct apical PM delivery of HA (150, 309). Only the addition of recombinant XIIIa inhibited basolateral delivery of VSV-G. Accordingly, the addition of anti-annexin XIIIb antibodies specically blocked transport of TGN-derived vesicles to the apical cell surface while transport to the basolateral surface was not changed (299). Unfortunately, the roles of annexin XIIIa and XIIIb in transcytosis were not tested in these studies. Some evidence for a role for annexin II in transcytosis comes from studies performed in isolated hepatocyte couplets (620). Upon induction of transcytosis by the addition of bile salts, annexin II immunouorescence increased dramatically and was redistributed sequentially from beneath the basolateral PM to perinuclear structures and nally to the apical pole (620). Whether the annexin was simply a passenger on these transcytotic intermediates or was mediating transport activity is not known. 8. Dynamin Dynamins are a family of high molecular mass (100 kDa), peripheral membrane-associated GTPases that function in the early stages of endocytosis (502, 586). Examination of dynamin function in transfected or microinjected mammalian cells has indicated its participation in clathrin-mediated endocytosis (106, 568) and suggested a role for dynamin in mediating ssion of caveolae from the PM (223, 416). A role for dynamin in transcytosis has best been established in endothelial cells. Not only was dynamin found associated with caveolae in these cells, but it was also identied as a required factor for the release of caveolae from endothelial PM preparations in vitro (416). Caveolae-mediated internalization of cholera toxin B was also impaired in cultured endothelial cells expressing dynamin dominant negative constructs (416). Although these results imply that dynamin is a general regulator of caveolae-mediated internalization in endothelial cells, the effects of the dominant negative dynamins on transcytosing proteins were not analyzed. Examination of dynamin isoforms in other polarized mammalian epithelial cells has been limited to cultured MDCK cells. Dynamin-2 was found at both PM domains
Physiol Rev VOL

and corresponding defects in IgA and Tf internalization from both domains were observed in cells expressing dominant negative dynamin mutants (7). Interestingly, dynamin-1 (the brain-specic isoform) localized only to the apical PM, and when the dominant negative dynamin-1 was expressed, internalization was inhibited only from that domain (7). Whether this is a physiologically relevant nding is not yet clear but may point to similarities in PM dynamics at the apical domain in epithelial cells and at the synapse in neurons. Nonetheless, many transcytosing receptors (including pIgA-R) are internalized via clathrin-coated vesicles, a process requiring dynamin activity. The prediction is that dynamin is required for the internalization of transcytosing molecules in polarized epithelial cells. Does dynamin regulate internalization of all transcytosing molecules, through caveolae, clathrin-coated, or noncoated endocytic vesicles, and at both PM domains? B. Cytoskeleton 1. Microtubules and microtubule-based motors In addition to the asymmetric distribution of PM proteins, the polarity of epithelial cells is also reected in the organization of the cytoskeleton. In nonpolarized cells, microtubules emanate from a juxtanuclear microtubule organizing center (MTOC). In polarized cells, there is accumulating evidence that microtubules are instead (or additionally?) organized from sites at or near the apical PM such that the emanating microtubules are oriented with their minus ends at the apical PM and their plus ends attached to or near the basolateral PM (22, 131, 374, 475). Such an arrangement also dictates the placement of organelles within the interior of the epithelial cell (410, 434). In particular, in many epithelial cells, the compartments of the transcytotic pathway are linearly situated along the parallel microtubules. However, this arrangement is not universal to all polarized cells. For example, the microtubules in endothelial cells are arranged parallel, perpendicular, and obliquely to the long axis and in some cases even form criss-crossed helical arrays (53, 481). In general, disruption of microtubules does not inhibit internalization of molecules (either soluble of membrane-associated) into early endosomal compartments, but does impair their movement to other compartments. This is true for molecules destined for transcytosis and internalized from the basolateral side in epithelial cells where the transcellular path is long, such as in colchicineor nocodazole-treated MDCK cells, isolated hepatocytes or Caco-2 cells (56, 110, 137, 190, 222, 246, 346, 362). Interestingly, addition of colchicine impaired transcytotic delivery of albumin in endothelial cells in situ despite the comparatively short distance between the apical and basolateral surfaces (13). However, addition of microtubule
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

910

PAMELA L. TUMA AND ANN L. HUBBARD

disrupting agents does not impair transcytosis of molecules internalized from the apical surface in MDCK or Caco-2 cells (246, 362). The reasons for this differential dependence on microtubules to the two domains are not yet understood. One possibility is that basolaterally destined vesicles have a shorter path to the lateral surface and thus a higher probability of encountering their target by simple diffusion. Alternatively, the machinery associated with basolaterally destined vesicles may promote more efcient and specic binding interactions with their target. Microtubules are probably not a direct requirement for transcytosis; they likely facilitate delivery by providing the tracks upon which vesicles are translocated (43). Thus, when microtubules are disrupted, the kinetics of delivery are slowed, i.e., it takes a vesicle longer to encounter its appropriate target membrane by diffusion than when tracked along microtubules. However, this passive role for microtubules does not account for the mistargeting observed for some transcytosing molecules upon microtubule disruption. For example, the apical PM proteins, aminopeptidase N, DPP IV, and alkaline phosphatase, lost their polarized expression patterns in Caco-2 cells treated with nocodazole or colchicine (56, 137, 190). One explanation for the mistargeting is that different vesicle populations require the activities of distinct assemblies of docking and fusion molecules that differ in their binding specicities. Those vesicles with more promiscuous docking capabilities are able to associate with the improper domain upon microtubule disruption (e.g., apically targeted vesicles) resulting in apparent missorting. Alternatively, the mistargeting may reect normal basolateral delivery of these PM proteins and the subsequent need for microtubules to facilitate transport to the apical PM domain. The microtubule-based motor molecule that has received the most attention as a possible regulator of transcytosis is cytoplasmic dynein. In vitro analysis indicated that this megadalton, multisubunit molecule translocates vesicles in an ATP-dependent manner toward microtubule minus ends and that its activity is enhanced by another megadalton, multisubunit molecule, dynactin (279, 373). Since the microtubule minus ends are anchored in the apical PM in many epithelial cells, it is thought that dynein mediates delivery of vesicles from the basolateral to the apical cell surface. Although this is an attractive proposal, the evidence conrming it is only circumstantial and largely stems from the effects of microtubule disruption on transcytosis. Additional evidence comes from the observation that transport in this direction is NEM sensitive. Although NEM sensitivity is a litmus test for the involvement of NSF in transport, this alkylating agent is also a potent inhibitor of dynein ATPase activity at similar concentrations (93). Despite the absence of direct evidence that dynein
Physiol Rev VOL

regulates transcytotic vesicle delivery, a recent study examining the trafcking of transfected rhodopsin in MDCK cells implicates this motor in direct apical vesicle targeting. The 14-kDa endogenous dynein light chain Tctex-1 directly binds transfected rhodopsin in MDCK cells. When RP3, a non-rhodopsin-binding Tctex-1 homolog, was overexpressed in MDCK cells, it displaced the endogenous Tctex-1 in the dynein complex and disrupted apical delivery of rhodopsin (567). Interestingly, the apical distributions of HA and gp135 were not changed in these cells, nor were any basolateral antigens, suggesting that distinct light chains might regulate vesicle translocation. In permeabilized MDCK cells, when cytoplasmic dynein activity was abolished by either ultraviolet/vanadate photocleavage or immunodepletion from cytosolic extracts, direct apical delivery was impaired (298). On the other hand, immunodepletion of kinesin, a plus-end directed microtubule motor, inhibited transport to both the basolateral (to the plus ends as expected) and the apical (to the minus ends which was unexpected) domains. One possible explanation for this last result is that kinesin may be required to translocate vesicles through the microtubule meshwork that is postulated to exist between the Golgi and apical PM in MDCK cells (22). Finally, a surprising nding is that apical-organized microtubules were not required for transport from the TGN to either cell surface domain in MDCK cells (199). This implies that microtubules of both polarities, and by extension, both motor proteins facilitate transport to either PM domain, a report that contradicts numerous reports. Such continued confusion highlights the need for more information from multiple systems before we can condently assign specic and/or generalizable roles for microtubules and their motors in transcytosis. 2. Actin and actin-based motors The actin cytoskeleton also has a unique organization in many polarized cells. In general, actin microlaments extend to the basolateral PM and form attachments through interactions with proteins of zonulae adherens, tight junctions, and focal adhesions. At the apical surface, actin is found as the core lament of microvilli and also as a dense subcortical web (58, 146, 366). At the basolateral domain, the actin-associated proteins fodrin and ankyrin form a scaffold that restricts the movements of certain integral PM proteins, including the Na-K-ATPase, thereby stabilizing the basolateral population (208, 366, 386). At the apical surface, actin was shown to be an important factor in maintaining the apical distribution of gp135, a membrane glycoprotein in MDCK cells, even in cells not in contact with their neighbors (419). In general, actin is thought to be an important regulator of endocytosis from the apical, but not basolateral, PM (16). In MDCK, Caco-2, and pancreatic acinar cells,
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS

911

addition of the actin disrupting agent cytochalasin D impaired internalization only from the apical domain (197, 261, 528). Clathrin-coated pits accumulated at the apical PM in each cell type, suggesting a block in clathrin-mediated internalization. In treated Caco-2 cells, apical internalization of folate was also decreased, implying that caveolae-mediated internalization was altered, and in MDCK cells, apical uptake of Lucifer yellow, which is internalized via noncoated vesicles, was decreased (197, 261). Thus actin may be important in many modes of apical internalization. However, there are exceptions to this generalization, as it was observed that the receptormediated internalization of ligand from the basolateral domain of hepatocytes was impaired by cytochalasin B treatment (280). Furthermore, treatment of Caco-2 cells with latrunculin B, another actin-disrupting agent, misdirected basolaterally internalized Tf-R and LDL-R into the apically directed transcytotic pathway (136). Limited evidence from studies in MDCK cells suggests that actin is also involved in facilitating steps in postendocytic transcytotic trafcking. In the presence of cytochalasin D, the delivery of transcytosing dIgA from the basolateral early endosome to the apical recycling endosome was impaired 45% (346). Interestingly, dIgA transcytosis was completely blocked when both cytochalasin D and nocodazole were added, suggesting that microlaments and microtubules work in concert to facilitate transport to the apical cell surface (16, 346). In support of the evidence indicating a role for actin in regulating membrane dynamics, recent studies have also implicated specic actin-based motors as important players in polarized vesicle trafcking. In particular, many classes of myosin motors have been localized to the subcortical actin network in many different epithelial cell types. The single-headed, short-tailed myosin I isoform has been localized to the apical brush border of intestinal (440) and kidney cells (94). They have also been found in association with zymogen granules at the apical aspect of pancreatic acinar cells (452) and in hepatocytes (25, 95). Also present at the intestinal brush border, although much less abundant, are the related myosin isoforms V and VI (218). Myosin VI has also been placed at the apical brush border of the proximal tubule cell line LLC-PK1 (215), whereas myosin Va has been also localized to subapical structures in polarized hepatic WIF-B cells (321) and myosin Vb at the apical recycling endosome in MDCK cells. Functional studies on two different myosin isoforms have also placed them as potential regulators of transcytosis. Brush-border myosin 1 (BBM1) plays a role in postendocytic trafc at the basolateral pole, whereas myosin Vb is functioning at the apical pole (136, 304, 315). RhoA and rac1, two small GTPases that regulate actin cytoskeletal dynamics, also have also been implicated as regulators of polarized membrane transport with rhoA functionPhysiol Rev VOL

ing at the basolateral surface and rac1 at the apical PM (270, 315). In particular, basolateral-to-apical transcytosing IgA accumulated in basolateral early endosomes in MDCK cells expressing inactivated forms of rhoA, whereas it accumulated in apical early endosomes in cells expressing dominant negative forms of myosin Vb or a constitutively activated form of rac1 (270, 304, 315). The mechanisms by which these motors, GTPases, and actin regulate membrane transport in polarized epithelial cells are not yet known but are the subject of a recent review (16). C. Lipids and Transcytosis The identication of proteins as important regulators of membrane transport is widely accepted and nearly indisputable. In the last several years, it has become more accepted that lipids also play signicant roles in membrane transport. In particular, phosphoinositides, PC, cholesterol, and glycosphingolipids have been shown to be important players. Here we will focus on the roles of phosphoinositide 3-phosphate [PI(3)P], cholesterol, and glycosphingolipids in polarized membrane transport. Other lipids and their modifying enzymes have been the subject of many recent reviews (99, 242, 353). 1. PI(3)P In the past several years, the role PI(3)P lipids play in regulating membrane transport has received considerable attention. This interest arose from early studies examining the effects of the selective phosphoinositide 3-kinase (PI 3-kinase) inhibitors wortmannin and LY294002 on membrane trafcking (98, 524, 584). To date, many transport pathways, including transcytosis, have been shown to be wortmannin and/or LY294002 sensitive. In MDCK cells, wortmannin treatment impaired basolateral to apical dIgA transcytosis (78, 212). Likewise, both wortmannin and LY294002 disrupted transcytosis of pIgA-R and three newly synthesized resident apical PM proteins to the WIF-B apical domain (582) while wortmannin perfusion in isolated rat livers decreased the biliary release of basolaterally internalized HRP (156). Both agents were also observed to impair transcytosis in both directions of ricin in FRT cells and of neonatal FcR in IMCD cells (212, 370). Although the transcytosing proteins in treated WIF-B cells eventually reached their nal destination (the apical PM), they transiently accumulated in basolateral early endosomes, indicating a block early in the transcytotic pathway (583). Mammalian cells encode at least three different classes of PI 3-kinase isoforms (23, 162). Class I includes the p85/p110 heterodimeric kinases which consist of an 110-kDa catalytic subunit associated with a regulatory 85-kDa subunit. The other member is PI 3-kinase-, whose
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

912

PAMELA L. TUMA AND ANN L. HUBBARD

catalytic activity is regulated by the -subunits of heterotrimeric G proteins (80, 162, 591a). Class II PI 3-kinases include higher molecular weight kinases that contain C2 domains and class III kinases share the highest sequence similarity with the sole isoform identied in yeast, Vps34p (224, 604). The last kinase is also under the control of a regulatory subunit, p150 (in mammalian cells) or Vps15p (in yeast) (225, 427). All mammalian PI 3-kinase isoforms are sensitive to wortmannin and LY294002 (class II kinases at higher concentrations), which has led to ambiguity in distinguishing the roles that specic PI 3-kinases play in membrane transport. However, more recent studies that examined the effects of microinjection of specic inhibitory reagents on membrane transport have helped to begin assigning specic roles to specic kinase (529). Using these inhibitory reagents in polarized hepatic cells, we found that specic inhibition of the class III PI 3-kinase Vps34p led to the formation of prelysosomal vacuoles containing endocytosed resident apical PM proteins and to the transient accumulation of transcytosing apical proteins in basolateral early endosomes (583). These results indicate that the lipid product of Vps34p, PI(3)P, regulates the two endocytic pathways differentially, at an early endosomal stage from the basolateral surface and from prelysosomes to lysosomes from the apical surface. The current model of PI(3)Ps role in endocytosis invokes recruitment of Vps34p/p150 by activated rab5 to the sites of endosome-endosome fusion and local production of PI(3)P (98, 559, 625). The PI(3)Pbinding protein early endosomal antigen 1 (EEA1) is recruited to these sites where PI(3)P and rab5 binding stabilize its membrane association. The stabilized EEA1 molecules then form oligomers that coordinate the formation of a large vesicle docking site also containing NSF and syntaxin13, all of which drive endosome fusion. Thus, in nonpolarized cells, when PI(3)P lipids were depleted by wortmannin, EEA1 association with early endosomes was lost and the subsequent events were affected. Likewise, we found that EEA1 dissociated from basolateral early endosomes in treated WIF-B cells; concomitantly, we observed delayed basolateral to apical transcytosis. All of these results are consistent with the current model. In contrast, the block we observed at a late endocytic step in the apical pathway is not consistent with the existing model (583). Disruption of EEA1 function in the fusion of early endosomes arising from the apical surface should have blocked an earlier step in the pathway. However, a subpopulation of EEA1 near the apical surface remained membrane-bound under PI(3)P-depleting conditions, suggesting that the protein remained active and allowed progression of endocytosed proteins along the apical route thereby revealing a block downstream. Injection of p110 inhibitory antibodies into WIF-B cells was also found to impair basolateral to apical transPhysiol Rev VOL

cytosis as did anti-Vps34p injection (583). However, increased basolateral surface staining of the transcytosing markers was observed with no corresponding intracellular accumulations, suggesting that Vps34p and p110 act at separate steps of the pathway, with p110 possibly acting at internalization. Together, these results lead to a number of questions. Are the apical and basolateral pathways in other polarized epithelial cells also differentially regulated by PI(3P)? Are the functions of p110 and Vps34p in basolateral-to-apical transcytosis conserved among other epithelial cells? What PI(3)P-binding proteins are required for membrane transport at the distinct steps? 2. Cholesterol and glycosphingolipids
Downloaded from physrev.physiology.org on November 4, 2009

Glycosphingolipids and cholesterol are enriched in cell surface membranes in all eukaryotic cells. In polarized epithelial cells, the apical surface is even further enriched for these lipid species (158, 281, 295, 597). One possible function of cholesterol and glycosphingolipids is to impart structural rigidity and decreased permeability to the apical domain, which in turn protects the cell against the harsh external environment it faces (e.g., the detergent-like bile or high acidity) (295). The differences in these environments would therefore dictate the lipid compositions required for appropriate protection and function in different epithelial cell types. The intrinsic properties of glycosphingolipids and cholesterol promote their assembly into specialized membrane domains called rafts (63, 214). Within these cell surface domains are selected proteins that are recruited based on their biophysical properties. In particular, GPI-anchored proteins that are predominantly expressed at the apical surfaces of epithelial cells localize to rafts. These observations in combination with studies performed mainly in MDCK cells have led to the raft hypothesis for protein sorting. According to this hypothesis, rafts form in the biosynthetic pathway where they recruit apically destined proteins (especially GPI-linked proteins); the rafts with their recruited cargo are then transported directly to the apical domain in vesicles. There is considerable experimental evidence to support this hypothesis. For example, GPI anchors have been shown to be sufcient to target proteins to the apical domain (62, 325). Furthermore, rafts have been isolated based on their insolubility in nonionic detergents (especially Triton X-100) at 4C, GPI-anchored proteins copurify with them (297), and cholesterol-depleting drugs and sphingolipid synthesis inhibitors disrupt delivery of apical PM residents (252, 297). Recent work has also suggested that caveolins and caveolae-like vesicles are important for PM delivery of GPI-anchored proteins. In caveolin-1 or caveolin-3 knock-out mice, GPI-anchored proteins were rewww.prv.org

83 JULY 2003

TRANSCYTOSIS

913

tained at the TGN in mouse embryo broblasts or muscle tissue, respectively (551). However, there are many observations that are inconsistent with the raft hypothesis of sorting (612). In FRT cells, both glycosphingolipids and GPI-anchored proteins are sorted to the basolateral domain, whereas in certain MDCK strains, they are evenly distributed between the two domains. However, in both cases, other apical PM proteins are sorted properly to the apical surface. In hepatocytes, GPI-anchored proteins, such as 5nucleotidase, are rst transported to the basolateral domain before apical delivery (499). Furthermore, many nonapical proteins have also been detected in puried raft fractions. Despite the debate, the question still remains. Do rafts sort apically destined proteins in the transcytotic pathway? If so, are rafts present at the basolateral domain or in other transcytotic intermediates? Two approaches have been taken to begin answering these questions. First is examining whether transcytosing proteins are present in detergent-insoluble fractions and second is whether they are internalized from the cell surface via caveolae, specialized raft domains. So far, the rst approach has yielded conicting results from studies performed in enterocytes as well as FRT and MDCK cells (210, 493). In polarized enterocytes from mouse intestinal explants, a signicant proportion of basolateral to apical transcytosing IgA (secreted from neighboring mucosal plasma cells) was found in detergent-insoluble rafts (210). Because IgA is internalized and delivered to the apical surface via the pIgA-R, the data imply that the receptor must also be raft associated. Accordingly, 50% of the pIgA-R was recovered in detergent-insoluble fractions (210). This result contradicts that reported for pIgA-R in MDCK and FRT cells; pIgA-R was not found in Triton X-100-insoluble fractions at any point during its life cycle (493). The reasons for these opposing observations are likely not due to differences in raft preparation, since the methods used were very similar. Instead, the differences may be related to the intrinsic differences between cell types, between in vitro versus in vivo systems, or between endogenously (in enterocytes) or exogenously (in FRT and MDCK cells) expressed molecules. None of these possibilities has yet been well explored. The second approach to determine whether rafts sort apically destined proteins in the transcytotic pathway has focused on examining interactions of transcytosing proteins with caveolae. Because not all rafts are associated with caveolin, caveolae have more recently been classied as specialized rafts. Much of what we know about caveolae in polarized epithelial cells comes from studies in endothelial cells where these structures are highly abundant (see sect. IV). Otherwise, caveolae have been examined in only a limited number of polarized cells. Despite the enrichment of cholesterol and glycosphingoPhysiol Rev VOL

lipids at the apical domain, caveolae have only been observed at the basolateral domain of MDCK cells and in kidney epithelial cells in situ (57, 435, 498). Like endothelial cells, MDCK caveolae formation is dependent on cholesterol levels (205, 267, 511). In both cases, no morphologically denable caveolae were observed in cells treated with cholesterol-depleting drugs such as lipin or cyclodextrin. In endothelial cells, the cholesterol-dependent loss of caveolae corresponded to decreases in albumin transcytosis both in vitro and in vivo (267, 511). When the drugs were withdrawn and cholesterol synthesis stimulated by the addition of mevalonate in MDCK cells, or the addition of 10 20% serum in endothelial cells, the caveolae reformed (205, 511). FRT and Caco-2 cells express little to no detectable caveolin, and no caveolae have been observed (322, 603). However, when caveolin-1 was overexpressed in these cells, caveolae were observed in both domains in FRT cells and only at the basolateral domain of Caco-2 cells (322, 603). Interestingly, the formation of caveolae in FRT cells did not promote apical sorting of GPI-anchored proteins or their sorting into rafts (322), suggesting that rafts are not responsible for apical targeting in these cells. Alternatively, other components may be lacking in FRT cells that are necessary for sorting into rafts and/or subsequent apical delivery. Our challenge is to begin carefully examining the detergent solubility properties of multiple endogenous apical PM protein types (e.g., GPI-linked, single transmembrane, polytopic) during their entire life cycles within one cell type to determine the role of rafts in transcytosis. The detergents used should not be limited to Triton X-100, as it has been recently reported that in PC12 cells, subpopulations of rafts exist with different solubility properties (482). The effects of both cholesterol and glycosphingolipid disrupters on transcytosis should be directly tested. Also, examination of puried organellar intermediates for raft components and protein insolubility properties may help clarify at which step(s) rafts are required. Such a strategy has shown that recycling endosomes (immunoisolated with anti-Tf-R antibodies) are enriched for raft components including glycosphingolipids, cholesterol, caveolin, and another raft-associated protein, otillin (167). Such careful and consistent experimentation will more clearly determine the role of rafts in apical vesicle targeting. Recent evidence also suggests that the PM-associated t-SNAREs are organized in cholesterol-dependent surface domains in nonpolarized cells (87, 303). Are these domains required for vesicle delivery to the apical PM? D. Perturbations of Transcytosis The use of perturbants has long been a way to begin dissecting the mechanisms and molecules involved in
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

914

PAMELA L. TUMA AND ANN L. HUBBARD

regulating complex cellular processes. Chemical perturbants are commonly used and have provided insight into understanding vesicle transport in mammalian cells. Of particular note are alkylating agents (e.g., NEM), drugs that specically disrupt the cytoskeletal systems of the cell and specic lipid kinase inhibitors which were discussed in section V, AC. In this section, we focus on the perturbation of acute regulation of vesicle transport by agents that alter the functions of heterotrimeric G proteins, intracellular calcium (Cai) homeostasis, protein kinase A (PKA), or protein kinase C (PKC) activity (Table 6). 1. Heterotrimeric G proteins and PKA Accumulating evidence supports a role for heterotrimeric G proteins in regulating vesicle transport in both the endocytic and exocytic pathways (4, 41, 42, 220, 412, 445, 561). Examination of the effects of various agents on epithelial cells, especially MDCK cells, have indicated that transcytosis is also regulated by G proteins. The rst clues came from treating cells with nonselective G protein activators, guanosine 5-O-(3-thiotriphosphate) (GTPS) and AlF. In both cases, transcytosis to the apical, but not basolateral, domain was slightly enhanced (42). Addition of specic G protein ADP-ribosylating toxins (cholera or pertussis toxins) further indicated that transport to the apical domain (from both the TGN and via transcytosis) is regulated by the Gs -subunit (31, 42, 211). Mastoparan, (a Gi-activating peptide) had no effect on pIgA-R apical delivery while anti-Gs -antibodies were slightly inhibitory, conrming a role for Gs in transcytosis to the apical domain (42). In intact cells, the overexpression of the wild-type or constitutively active Gs -subunit led to a modest increase in transcytosis (211). Interestingly, the addition of either the Gs - or -subunits to an in vitro system led to a small increase in the formation of transcytotic vesicles (42). However, it is not known whether the -subunits were acting to inhibit another G protein that negatively regulates transcytosis or in concert with the -subunit. One well-studied result of Gs activation is the activation of adenylyl cyclase and the subsequent increased production of cAMP. PKA is then activated by the increased cAMP levels, which puts in motion numerous (but not well-dened) cellular processes. To determine whether this cascade of events is involved in vesicle transport, another handful of chemical perturbants has been useful. In particular, forskolin (a direct activator of adenylyl cyclase) has been shown to enhance transcytosis to the apical, but not basolateral, domain in MDCK cells (211). This effect was also seen by the addition of exogenous cAMP to both MDCK cells and intact rat hepatocytes (211, 217). A PKA inhibitor, H-89, produced the opposite effect, consistent with a role for PKA in regulatPhysiol Rev VOL

ing transcytosis (211). Some evidence suggests that G protein/PKA regulates transcytosis from apical endosomal compartments to the apical PM (31). Interestingly, this is also the site(s) at which at least nine small-molecular-weight GTP-binding proteins have been proposed to function, further exposing the complexity of vesicle transport events in this region of a polarized epithelial cell. Trimeric G proteins are also important regulators of endothelial transcytosis, but in this case, Gi has been examined most extensively. In polarized endothelial cells in vitro, addition of pertussis toxin (a Gi inhibitor) or expression of a dominant negative Gi peptide inhibited apical to basolateral albumin transcytosis mediated by activated gp60 (379). Previously, treatment of endothelial cells with tyrosine kinase inhibitors suggested that a srcmediated signaling pathway regulated transcytosis (Ref. 576 and discussion in sect. IVA3); thus Minshall and colleagues examined whether Gi and src signaling were coupled. Consistent with this hypothesis, overexpression of dominant negative src prevented the association of Gi with caveolin (caveolae?) in caveolin-1-overexpressing cells. Furthermore, dominant negative src also inhibited albumin transcytosis. From these results, the authors suggested that gp60 activation recruits Gi to caveolae that in turn sets off a src-mediated signaling cascade that activates transcytosis (379). This situation is somewhat analogous to the activation of pIgA-R transcytosis by ligand binding where another tyrosine kinase, p62yes, is involved (341). Thus tyrosine phosphorylation may be a common mechanism for regulating activated transcytotic pathways. 2. Calcium, calmodulin, and PKC The use of another set of pharmacological agents has indicated that vesicle trafcking is also acutely regulated by changes in Cai levels. Thapsigargin, a selective inhibitor of sarco/endoplasmic reticulum Ca2-ATPases, enhanced transcytosis to the apical domain in MDCK cells, whereas BAPTA, a Ca2-chelating compound, inhibited it (79). Neither agent altered transcytosis in the opposite direction. The mechanism whereby Cai uxes are manifested in changes in vesicle transport is not known, but likely includes alterations in Ca2-dependent enzyme activities. This has been substantiated by studies examining the effects of the calmodulin antagonists W-7, W-13, and triuoperazine in MDCK cells (18, 244, 329). All three agents signicantly impaired basolateral to apical transcytosis of dIgA while transcytosis in the same direction of the uid-phase marker ricin was enhanced. In both cases, endocytosis from the basolateral surface was unchanged, suggesting that the agents were acting later in the pathway. Consistent with this is the nding that dIgA accumulated in large endosomal structures located in the apical region of cells treated with W-13 (18). Interestingly, apical
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS
TABLE

915

6. Effects of pharmacological agents on transcytosis


Mode of Action BL-A A-BL Epithelial Cell Types Examined SNAREs Comments Reference No.

Agent

NEM Bacterial toxins

Alkylates sulfhydryls Proteolytically cleaves v- and t-SNAREs 2

N/D N/D

MDCK and endothelial cells, hepatocytes MDCK cells Lipids

Very nonselective and will inhibit multiple ATPases Each toxin is highly selective for its target

17, 253, 505, 564 17, 26, 27, 314

Wortmannin

PI 3-kinase inhibitor

2 2 2

/2 2 N/D

LY294002 Filipin

PI 3-kinase inhibitor Binds and removes cholesterol from PM

MDCK, FRT, IMCD and WIF-B cells, hepatocytes IMCD and WIF-B cells Endothelial cells Cytoskeleton

Class I and III kinases inhibited at low concentrations (nM), all classes inhibited at high concentrations (M), irreversible All classes inhibited at high concentrations (M), reversible Specic for unesteried cholesterol, reversible

78, 156, 212, 370, 582

370, 582 509

Nocodazole

Microtubule depolymerizer

2 2 2 2 1 1 1/2 /1 /1 2 1 2 1/2 2 1 1

/2 N/D N/D N/D

Many cell types

Specic, cell permeant, reversible

Colchicine UV/vanadate Cytochalasin D GTPS AIF Cholera toxin

Inhibits tubulin polymerization ATPase inhibitor Actin depolymerizer

Many cell types MDCK cells Many cell types

Specic, irreversible Not readily cell permeant, potent inhibitor of many ATPases, especially ion pump ATPases Specic, cell permeant, reversible

56, 110, 137, 222, 246, 346, 362, 370, 453, 548 13, 390 298 197, 261, 346, 528

Downloaded from physrev.physiology.org on November 4, 2009

G proteins and PKA G protein activator G protein activator ADP-ribosylates and activates Gs -subunit ADP-ribosylates and activates Gi, Go, and Gt -subunits Gi, Go, and Gt activator Adenylyl cyclase activator PKA activator PKA inhibitor Ca2-ATPase inhibitor Calcium chelator CaM antagonist CaM antagonist CaM antagonist MDCK cells MDCK cells MDCK cells Activates all GTPases Activates all heterotrimeric G proteins Holotoxin must bind GM1 to be internalized, A subunit (with ADP ribosylation activity) is not cell permeant Holotoxin must bind cell surface to be internalized, A protomer (with ADP ribosylation activity) is not cell permeant Cell permeant, also inhibits calmodulin and activates phospholipase A2 Specic Not readily cell permeant Potent, selective 41 41 31, 41, 211, 379

Pertussis toxin

N/D

MDCK cells

31, 42, 211

Mastoparan Forskolin cAMP H-89

N/D N/D N/D 2/ N/D 2/ N/D N/D

MDCK cells MDCK cells MDCK cells and hepatocytes MDCK cells

42 211 211, 217 211

Calcium, CaM, and PKC Thapsigargin BAPTA W-7 W-13 Triuoperazine MDCK cells MDCK cells FRT and MDCK cells MDCK cells FRT and MDCK cells Potent and specic for sarco endoplasmic reticulum Ca2-ATPases 105-fold greater afnity for Ca2 than Mg2; available in a cell permeant form Cell permeant Cell permeant Elevates intracellular Ca levels at low concentrations, antagonizes CaM at higher concentrations, cell permeant Potent, selective Cell permeant, potent Nonselective, cell permeant 79 79 244, 329, 349 18, 349 329

KN-62 PMA H-7

CaM kinase II inhibitor PKC activator Serine/threonine kinase inhibitor

MDCK cells MDCK cells MDCK cells Tyrosine kinases

329 79 18

Herbimycin Genestein PP1

Tyrosine kinase inhibitor Tyrosine kinase inhibitor Tyrosine kinase inhibitor

2 2 2

N/D N/D N/D

Endothelial and MDCK cells MDCK and endothelial cells MDCK cells

Highly selective for tyrosine kinases, cell permeant, irreversible Nonselective Selective for Src tyrosine kinases

340, 576 340, 576 340

1, Increases; 2, decreases; , no change; CaM, calmodulin; IMCD, inner medullary collecting duct; N/D, not determined; NEM, Nethylmaleimide; GTPS, guanosine 5-O-(3-thiotriphosphate); PKC, protein kinase C; PKA, protein kinase A; PI, phosphatidylinositol.

to basolateral transcytosis of ricin was not altered by these agents, yet an increase in its endocytosis from the apical domain was observed (329). However, megalinmediated transcytosis of thyroglobulin in the same direction was inhibited in thyroid cells (FRTL-5) treated with W-7 and triuoperazine (349). Thus CaM regulation is
Physiol Rev VOL

important for many steps in transcytosis. The challenge is to pinpoint the specic calmodulin-dependent enzymes that are functioning at these transport steps to understand the differential effects of these agents. PKC has also received attention as a possible regulator of transcytosis based both on the effects of changing Cai
www.prv.org

83 JULY 2003

916

PAMELA L. TUMA AND ANN L. HUBBARD

levels and on the use of phorbol esters, potent PKC activators. When one such phorbol ester, phorbol 12-myristate 13-acetate (PMA), was applied to MDCK cells, both apical recycling and basolateral to apical transcytosis of dIgA and transferrin were enhanced, suggesting PKC was acting at an apical recycling compartment (79). PMA treatment also led to the membrane recruitment of and PKC isoforms (79). Interestingly, dIgA binding to its receptor (conditions that activate its own transcytosis) also activated PKC-, which led to increased levels of inositol 1,4,5-trisphosphate and Cai, the latter of which stimulated transcytosis (79, 548). The rise in Cai is likely mediated via inositol 1,4,5-trisphosphatesensitive intracellular stores; thus ligand binding initiates a signal that is propagated across the cell independent of the ligand-receptor complex itself (340). Paradoxically, treatment of MDCK cells with H-7, a specic PKC inhibitor, did not inhibit dIgA or ricin transcytosis in MDCK cells (18, 329). At present, there is no good explanation for these disparate results. 3. Possible mechanisms In all the studies cited above, it is important to point out that in many cases, the inhibitory and stimulatory effects of transfection or addition of pharmacological agents on PM targeting were small. Does the size of the response in vitro reect loss of normal regulation that would be observed in vivo or does it represent ne-tuning that may be critical for proper organ function? If it is the latter, what appears to be a minimal change in vitro may have a large impact on the overall homeostatic balance of the organism. Thus physiological studies are needed. Specically, our challenge is to identify the molecules in membrane transport that are regulated by phosphorylation or calmodulin/Ca2 binding. Few if any direct links have yet been established, but there are some examples of where the modications of molecules implicated in transcytosis correlate with their proposed functions. Many of the SNARE molecules are phosphorylated in vitro by puried kinases, and the modication alters their binding properties. In particular, -SNAP is a substrate for PKA and when phosphorylated, its ability to bind the core docking and fusion complex was decreased 10-fold (230). In vitro, syntaxin 4 was shown to be phosphorylated by PKA, casein kinase II (CKII), and PKC, and this phosphorylation disrupted its binding to SNAP23 or SNAP25 (92, 159, 474). When syntaxin 4 was used as bait in a yeast two-hybrid screen, a novel SNARE kinase (SNAK) was identied, but surprisingly, SNAP-23 was overwhelmingly its preferred substrate in vitro and in vivo (73). SNAKphosphorylated SNAP-23 was not associated with the ternary complex, whereas phosphorylation of syntaxin 1 by CKII enhanced t-SNAREs association with SNAP-25 (154). Interestingly, by using phosphospecic antibodies to stain neurons, it was found that the phosphorylated
Physiol Rev VOL

form of syntaxin 1 was localized to discrete regions along the axonal PM that did not colocalize with synaptic vesicles (154). Another SNARE hypothesis molecule, Munc18 1, is a substrate for PKC and cyclin-dependent kinase 5, and phosphorylation in this case inhibited binding to syntaxin 1 (123a, 164, 527). Several rabs and rab effector proteins have also been shown to be phosphorylated. In vitro, GDI phosphorylation is mediated by PKA (555). In vivo, the phosphorylated GDI associated to the cytosolic form of rab5, while the unphosphorylated GDI was bound to the membrane-associated rab5. These data suggest that the cycling of rab proteins between donor and acceptor membranes is also a regulated process. The rab effector protein rabphilin 3A is phosphorylated by PKA in vitro (411). Another rab effector, rab8ip, is a serine/threonine protein kinase itself (GC kinase) that is activated by the stress response in lymphocytes (466). The phosphorylation state of rip11, a rab11 effector, may regulate its membrane binding properties. In polarized MDCK cells, conditions that decrease rip11 phosphorylation (e.g., staurosporine treatment) enhanced its binding to membranes (458). Caveolin, the major structural protein of caveolae, has been shown to be tyrosine phosphorylated in endothelial cells under conditions where transcytosis was stimulated (191, 318, 358). Both kinesin and cytoplasmic dynein are phosphorylated in vitro and in vivo, and this modication has been correlated with their ability to transport vesicles (including transcytotic vesicles?) along microtubules (320, 486, 494). The light chain of the actin based myosin I motor proteins is calmodulin, which is thought to regulate motor activity (449). Annexin binding to membrane lipids, and by extension, ability to promote intermembrane associations, is dependent on Ca2 (103). E. Transcytosis Versus Direct PM Delivery Transcytosis is only one pathway that molecules take to a specic PM domain. Both newly synthesized PM proteins and secreted molecules can also be delivered directly from the TGN to either PM domain. How different are the mechanisms regulating vesicle transport along these pathways? As expected, transport directly from the TGN to either the apical or basolateral domains is regulated differently. In particular, differences in the involvement of SNARE molecules were observed in permeabilized MDCK cells (17, 253, 332). Addition of anti-NSF antibodies, NEM, mutant NSF, rab-GDI, or tetanus and botulinum F neurotoxins all inhibited basolateral targeting of VSV-G protein, whereas targeting from the TGN to the apical domain of HA was not changed (17, 253). This implies that basolateral targeting requires NSF, rab proteins, and VAMP 2. Conversely, syntaxin 3 overexpression or anti-syntaxin 3 antibodies inhibited only apical delivery
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS

917

(300, 332). Addition of -SNAP antibodies and treatment of cells with botulinum E inhibited transport to both domains (17, 253), whereas TI-VAMP antibodies inhibited apical transport (the effects of theses antibodies on basolateral targeting were not examined) (300). Taken together, targeting to both domains requires SNARE molecules, but in different combinations. Surprisingly, apical targeting is NSF independent, suggesting the involvement of an as yet unidentied homolog. Microtubule and actin lament disruption also has differential effects on direct delivery to either domain in MDCK cells; apical delivery is inhibited, whereas, in most cases, basolateral is not (56, 137, 190, 362, 420, 598). In addition, PKA, PKC, and calmodulin-mediated mechanisms appear to acutely regulate delivery mainly to the apical domain (79, 329, 447). How different are the targeting mechanisms regulating transcytosis? This question has so far been best addressed in studies performed in MDCK cells. Unlike TGN to apical delivery of HA-containing vesicles, pIgA-R-mediated transcytosis appears to require NSF activity (17, 253). Furthermore, syntaxin 3 (required for direct apical targeting) is not involved in mediating transcytosis of IgA (332). SNAP-23, on the other hand, is involved in both basolateral to apical transcytosis, and direct apical and basolateral targeting (332). Also, both direct and transcytotic delivery to the basolateral domain does not require microtubules (see sect. VB). Based on the effects of pertussis toxin on MDCK cells, transport from the TGN to the basolateral PM was found to involve the Gi subunit of heterotrimeric G proteins, whereas transcytosis to this domain was not (31, 211, 446). From these results (and others) it is clear that the mechanisms cells use to regulate cell surface delivery are complex and are dependent on factors that are not yet understood. Another interesting twist to apical PM sorting has come from studies looking at the raft-associated protein referred to as MAL. This 17-kDa tetra-spanning TMD protein, rst identied in myelin and lymphocytes (hence MAL), is also expressed in many epithelial cell types where it is concentrated at the TGN (287). With the use of an antisense approach, it was shown that MDCK cells lacking MAL showed decreased specic apical delivery of a single TMD apical protein surrogate, the inuenza HA the ectopic expression of human MAL rescued the defect (459). Thus MAL has been implicated as an important player in apical sorting. Interestingly, liver does not express this isoform of MAL, a nding consistent with the absence of a direct apical delivery mechanism for the single-TMD class of apical PM proteins in hepatocytes. Recently, another MAL family member has been identied, MAL2, that is enriched in hepatic cells (120). In HepG2 cells treated with antisense MAL2 oligonucleotides, transcytosis of pIgA was impaired from early endosomes to a juxta-apical compartment. Thus different MAL isoforms may be responsible for specialized domain-spePhysiol Rev VOL

cic protein sorting. This is further suggested by the nding that yet another MAL family member, BENE, is expressed in endothelial cells where it is associated with caveolae (119). VI. CONCLUSION We have learned a lot about transcytosis since its existence was rst postulated over 50 years ago. We have identied cargo, uncovered pathways, and determined possible mechanisms. Nonetheless, many things remain mysterious. How does the cell discriminate between cargo destined for transcytosis versus degradation in lysosomes? In peripheral endothelial cells, it appears that separate entry points dictate different fates; internalization via coated pits sends cargo to the endocytic pathway, whereas caveolae-mediated internalization ensures a transcytotic fate. However, we need further study to make a denitive conclusion on this point. In other epithelial cells, the transcytotic pathway is a branch of the endocytic pathway. Where and how is transcytotic cargo sorted in these cells? Signals have been identied in the pIgA-R cytoplasmic tail that prevent its degradation. How are they recognized? Are they universal? In enterocytes, megalin recycles to the PM from endosomes after internalization and dissociation from its transcytotic cargo, whereas in thyroid, the same membrane receptor escorts thyroglobulin across the cell. What signals navigate megalin along these different itineraries and how do cells discriminate between the possible fates? Another unsolved problem is understanding how the cell determines what should be transcytosed versus that which should be diverted for its own use. Are the acute regulatory mechanisms described in section VD3 important here? How might transcytosis dysfunction or corruption contribute to human disease? For example, do mistakes in self-apportionment lead to vitamin B12 deciency? Do mistakes in cargo selection cause disease? In enterocytes, the transcytosis of undigested food antigens to underlying interstitial cells has been linked to food hypersensitivity and allergies (630). The antigenic epitopes stimulate the immune system leading to the production of cytokines that ultimately result in the loss of enterocyte barrier function and diarrhea. Similarly, certain pathogens co-opt the transcytotic pathway of M cells for infection. Recent evidence suggests that Streptococcus pneumoniae binds unoccupied, uncleaved pIgA-R that has been transcytosed to the apical surfaces of nasopharyngeal epithelial cells. The receptor-pneumonia complex is thought to be apically internalized and transcytosed to the basolateral surface where the pathogen is released. Finally, how is transcytosis used or adapted in development? This is a huge unexplored area where there will undoubtedly be surprises. Let us hope that the next 50 years will provide us with some exciting answers.
www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

918

PAMELA L. TUMA AND ANN L. HUBBARD MDCK cells: existence of an NSF-dependent fusion mechanism with the apical surface of MDCK cells. EMBO J 15: 14711481, 1996. APODACA G, ENRICH C, AND MOSTOV KE. The calmodulin antagonist, W-13, alters transcytosis, recycling, and the morphology of the endocytic pathway in Madin-Darby canine kidney cells. J Biol Chem 269: 1900519013, 1994. AROETI B, KOSEN PA, KUNTZ ID, COHEN FE, AND MOSTOV KE. Mutational and secondary structural analysis of the basolateral sorting signal of the polymeric immunoglobulin receptor. J Cell Biol 123: 1149 1160, 1993. AROETI B AND MOSTOV KE. Polarized sorting of the polymeric immunoglobulin receptor in the exocytotic and endocytotic pathways is controlled by the same amino acids. EMBO J 13: 22972304, 1994. AROETI B, OKHRIMENKO H, REICH V, AND ORZECH E. Polarized trafcking of plasma membrane proteins: emerging roles for coats, SNAREs, GTPases and their link to the cytoskeleton. Biochim Biophys Acta 1376: 5790, 1998. BACALLAO R, ANTONY C, DOTTI C, KARSENTI E, STELZER EHK, AND SIMONS K. The subcellular organization of Madin-Darby canine kidney cells during formation of a polarized epithelium. J Cell Biol 109: 28172832, 1989. BACKER JM. Phosphoinositide 3-kinases and the regulation of vesicular trafcking. Mol Cell Biol 3: 193204, 2000. BALIN BJ AND BROADWELL RD. Trancytosis of protein through the mammalian cerebral epithelium and endothelium. I. Choroid plexus and the blood-cerebrospinal uid barrier. J Neurocytol 17: 809 826, 1988. BALISH MF AND COLUCCIO LM. Identication of brush border myosin-I in liver and testis. Biochem Biophys Res Commun 211: 331 339, 1995. BANERJEE A, LI G, ALEXANDER EA, AND SCHWARTZ JH. Role of SNAP-23 in trafcking of H-ATPase in cultured inner medullary collecting duct cells. Am J Physiol Cell Physiol 280: C775C781, 2001. BANERJEE A, SHIH T, ALEXANDER EA, AND SCHWARTZ JH. SNARE proteins regulate H()-ATPase redistribution to the apical membrane in rat renal inner medullary collecting duct cells. J Biol Chem 274: 26518 26522, 1999. BANKER GA AND COWAN WM. Rat hippocampal neurons in dispersed cell culture. Brain Res 126: 397342, 1977. BARR VA AND HUBBARD AL. Newly synthesized hepatocyte plasma membrane proteins are transported in transcytotic vesicles in the bile duct-ligated rat. Gastroenterology 105: 554 571, 1993. BARR VA, SCOTT LJ, AND HUBBARD AL. Immunoadsorption of hepatic vesicles carrying newly synthesized dipeptidyl peptidase IV and polymeric IgA receptor. J Biol Chem 270: 27834 27844, 1995. BARROSO M AND SZTUL ES. Basolateral to apical transcytosis in polarized cells is indirect and involves BFA and trimeric G protein sensitive passage through the apical endosome. J Cell Biol 124: 83100, 1994. BARTLES JR, FERACCI HM, STIEGER B, AND HUBBARD AL. Biogenesis of the rat hepatocyte plasma membrane in vivo: comparison of the pathways taken by apical and basolateral proteins using subcellular fractionation. J Cell Biol 105: 12411251, 1987. BARTLES JR AND HUBBARD AL. Plasma membrane protein sorting in epithelial cells: do secretory pathways hold the key? Trends Biochem Sci 13: 181184, 1988. BASTAKI M, BRAITERMAN LT, JOHNS DC, CHEN YH, AND HUBBARD AL. Absence of direct delivery for single transmembrane apical proteins or their secretory forms in polarized hepatic cells. Mol Biol Cell 13: 225237, 2002. BEGLEY DJ. Peptides and the blood-brain barrier: the status of our understanding. Ann NY Acad Sci 739: 89 100, 1994. BERKELS R, HASS U, AND KLAUS W. The calcium antagonist nifedipine inhibits the uptake of acetylated LDL into endothelial cells. Naunyn-Schmiedebergs Arch Pharmacol 362: 9195, 2000. BERRY MN AND FRIEND DS. High-yield preparation of isolated rat liver parenchymal cells: a biochemical and ne structural study. J Cell Biol 43: 506 520, 1969. BIDEY SP, CHIOVATO L, DAY A, TURMAINE M, GOULD RP, EKINS RP, AND MARSHALL NJ. Evaluation of the rat thyroid cell strain FRTL-5 as an in-vitro bioassay system for thyrotrophin. J Endocrinol 101: 269 276, 1984. www.prv.org

We thank R. Fuchs, L. Ghitescu, M. Lisanti, M. Molliver, M. Neutra, S. C. Silverstein, and M. Wessling-Resnick for sharing their expertise in areas of transcytosis less familiar to us. Thanks also to the Editorial Board (Susan Hamilton) for their patience during the long gestation period before seeing this review. Our research has been supported by National Institutes of Health Grants P01-DK-44375, R01-GM-29185, NRSA-DK-09620, and T32-DK-07632. Present address of P. L. Tuma: Biology Department, The Catholic University of America, Washington, DC 20064. Address for reprint requests and other correspondence: A. L. Hubbard, Hunterian 119, Dept. of Cell Biology, 725 N. Wolfe St., Baltimore, MD 21205 (E-mail: alh@jhmi.edu).

18.

19.

20.

21.

22.

REFERENCES
23. 1. ABRAHAMSON DR AND RODEWALD R. Evidence for the sorting of endocytic vesicle contents during the receptor-mediated transport of IgG across the newborn rat intestine. J Cell Biol 91: 270 280, 1981. 2. ADACHI H, TSUJIMOTO M, ARAI H, AND INOUE K. Expression cloning of a novel scavenger receptor from human endothelial cells. J Biol Chem 272: 3121731220, 1997. 3. ADEN DP, FOGEL A, PLOTKIN S, DAMJANOV I, AND KNOWLES B. Controlled synthesis of HBsAg in a differentiated human liver carcinoma-derived cell line. Nature 282: 615 616, 1979. 4. AHNERT-HILGER G, MACH W, FOHR KJ, AND GRATZL M. Poration by alpha-toxin and streptolysin O: an approach to analyze intracellular processes. Methods Cell Biol 31: 6390, 1989. 5. AISEN P, ENNS C, AND WESSLING-RESNICK M. Chemistry and biology of eukaryotic iron metabolism. Int J Biochem Cell Biol 33: 940 959, 2001. 6. ALLAN CH, MENDRICK DL, AND TRIER JS. Rat intestinal M cells contain acidic endosomal-lysosomal compartments and express class II major histocompatibility complex determinants. Gastroenterology 104: 698 708, 1993. 7. ALTSCHULER Y, BARBAS SM, TERLECKY LJ, TANG K, HARDY S, MOSTOV KE, AND SCHMID SL. Redundant and distinct functions for Dynamin-1 and Dynamin-2 isoforms. J Cell Biol 143: 18711881, 1998. 8. AMERONGEN HM, WELTZIN R, FARNET CM, MICHETTI P, HASELTINE WA, AND NEUTRA MR. Transepithelial transport of HIV-1 by intestinal M cells: a mechanism for transmission of AIDS. J Acquir Immune Dec Syndr 4: 760 765, 1991. 9. ANDERSON RG. The caveolae membrane system. Annu Rev Biochem 67: 199 225, 1998. 10. ANDERSON RGW. Plasmalemmal caveolae and GPI-anchored membrane proteins. Curr Opin Cell Biol 5: 647 652, 1993. 11. ANDREWS NC AND LEVY JE. Iron is hot: an update on the pathophysiology of hemochromatosis. J Am Soc Hematol 92: 18451851, 1998. 12. ANDREWS PA. Disorders of iron metabolism. N Engl J Med 342: 12931294, 2000. 13. ANTOHE F, DOBRILA L, HELTIANU C, SIMIONESCU N, AND SIMIONESCU M. Albumin-binding proteins function in the receptor-mediated binding and transcytosis of albumin across cultured endothelial cells. Eur J Cell Biol 60: 268 275, 1993. 14. ANTOHE F, RADULESCU L, GAFENCU A, GHETIE V, AND SIMIONESCU M. Expression of functionally active FcRn and the differentiated bidirectional transport of IgG in human placental endothelial cells. Hum Immunol 62: 93105, 2001. 15. ANTONIN W, HOLROYD C, FASSHAUER D, PABST S, VON MOLLARD GF, AND JAHN R. A SNARE complex mediating fusion of late endosomes denes conserved properties of SNARE structure and function. EMBO J 19: 6453 6464, 2000. 16. APODACA G. Endocytic trafc in polarized epithelial cells: role of the actin and microtubule cytoskeleton. Trafc 2: 149 159, 2001. 17. APODACA G, CARDONE MH, WHITEHEART SW, DASGUPTA BR, AND MOSTOV KE. Reconstitution of transcytosis in SLO-permeabilized Physiol Rev VOL 24.

Downloaded from physrev.physiology.org on November 4, 2009

25.

26.

27.

28. 29.

30.

31.

32.

33.

34.

35. 36.

37.

38.

83 JULY 2003

TRANSCYTOSIS 39. BLOXAM DL, BAX BE, AND BAX CM. Culture of syncytiotrophoblast for the study of human placental transfer. II. Production, culture and use of syncytiotrophoblast. Placenta 18: 99 108, 1997. 40. BOLENDER RP. Stereological analysis of the guinea pig pancreas. I. Analytical model and quantitative description of nonstimulated pancreatic exocrine cells. J Cell Biol 61: 269 287, 1974. 41. BOMSEL M AND MOSTOV KE. Both the Gs alpha and beta gamma subunits of the heterotrimeric G protein, Gs, control the sorting of the polymeric immunoglobulin receptor into transcytotic vesicles. Biochem Soc Trans 22: 463 468, 1994. 42. BOMSEL M AND MOSTOV KE. Possible role of both the a and bg subunits of the heterotrimeric G protein, Gs, in transcytosis of the polymeric immunoglobulin receptor. J Biol Chem 268: 25824 25835, 1993. 43. BOMSEL M, PARTON R, KUZNETSOV SA, SCHROER TA, AND GRUENBERG J. Microtubule- and motor-dependent fusion in vitro between apical and basolateral endocytic vesicles from MDCK cells. Cell 62: 719 731, 1990. 44. BOMSEL M, PRYDZ K, PARTON RG, GRUENBERG J, AND SIMONS K. Endocytosis in lter-grown Madin-Darby canine kidney cells. J Cell Biol 109: 32433258, 1989. 45. BONILHA VL, MARMORSTEIN AD, COHEN-GOULD L, AND RODRIGUEZ-BOULAN E. Apical sorting of inuenza hemagglutinin by transcytosis in retinal pigment epithelium. J Cell Sci 110: 17171727, 1997. 46. BORST P, ZELCER N, AND VAN HELVOORT A. ABC transporters in lipid transport. Biochim Biophys Acta 1486: 128 144, 2000. 47. BORVAK J, RICHARDSON J, MEDESAN C, ANTOHE F, RADU C, SIMIONESCU M, GHETIE V, AND WARD ES. Functional expression of the MHC class I-related receptor, FcRn, in endothelial cells of mice. Int Immunol 10: 1289 1298, 1998. 48. BOSE S, SEETHARAM S, DAHMS NM, AND SEETHARAM B. Bipolar functional expression of transcobalamin II receptor in human intestinal epithal Caco-2 cells. J Biol Chem 272: 3538 3543, 1997. 49. BOSE S, SEETHARAM S, AND SEETHARAM B. Membrane expression and interactions of human transcobalamin II receptor. J Biol Chem 270: 8152 8157, 1995. 50. BOYER JL. Isolated hepatocyte couplets and bile duct unitsnovel preparations for the in vitro study of bile secretory function. Cell Biol Toxicol 13: 289 300, 1997. 51. BOYER JL, PHILLIPS JM, AND GRAF J. Preparation and specic applications of isolated hepatocyte couplets. Methods Enzymol 192: 501516, 1990. 52. BRADBURY MW. Transport of iron in the blood-brain-cerebrospinal uid system. J Neurochem 69: 443 454, 1997. 53. BRAET F, DE ZANGER R, KALLE W, RAAP A, TANKE H, AND WISSE E. Comparative scanning, transmission and atomic force microscopy of the microtubular cytoskeleton in fenestrated liver endothelial cells. Scanning Microsc Suppl 10: 225235, 1996. 54. BRAMBELL FWR. The transmission of immunity from mother to young and the catabolism of immunoglobulins. Lancet 2: 1087 1093, 1966. 55. BREITFELD PP, CASANOVA JE, MCKINNON WC, AND MOSTOV KE. Deletions in the cytoplasmic domain of the polymeric immunoglobulin receptor differentially affect endocytotic rate and postendocytotic sorting. J Biol Chem 265: 13750 13757, 1990. 56. BREITFELD PP, MCKINNON WC, AND MOSTOV KE. Effect of nocodazole on vesicular trafc to the apical and basolateral surfaces of polarized Madin-Darby canine kidney cells. J Cell Biol 111: 23652373, 1990. 57. BRETON S, LISANTI MP, TYSZKOWSKI R, MCLAUGHLIN M, AND BROWN D. Basolateral distribution of caveolin-1 in the kidney. Absence from H-ATPase-coated endocytic vesicles in intercalated cells. J Histochem Cytochem 46: 205214, 1998. 58. BRETSCHER A. Microlament structure and function in the cortical cytoskeleton. Annu Rev Cell Biol 7: 337374, 1991. 59. BREUZA L, FRANSEN J, AND LE BIVIC A. Transport and function of syntaxin 3 in human epithelial intestinal cells. Am J Physiol Cell Physiol 279: C1239 C1248, 2000. 60. BRIGHTMAN MW AND REESE TS. Junctions between intimately apposed cell membranes in the vertebrate brain. J Cell Biol 40: 648 677, 1969. 61. BRONNER F. Calcium absorptiona paradigm for mineral absorption. J Nutr 128: 917920, 1998. Physiol Rev VOL

919

62. BROWN DA, CRISE B, AND ROSE JK. Mechanism of membrane anchoring affects polarized expression of two proteins in MDCK cells. Science 245: 1499 1501, 1989. 63. BROWN RE. Sphingolipid organization in biomembranes: what physical studies of model membranes reveal. J Cell Sci 111: 19, 1998. 64. BROWN WR, ISOBE Y, AND NAKANE PK. Studies on translocation of immunoglobulins across intestinal epithelium. II. Immunoelectronmicroscopic localization of immunoglobulins and secretory component in human intestinal mucosa. Gastroenterology 71: 985995, 1976. 65. BRUNS RR AND PALADE GE. Studies on blood capillaries. I. General organization of blood capillaries in muscle. J Cell Biol 37: 244 276, 1968. 66. BRUNS RR AND PALADE GE. Studies on blood capillaries. II. Transport of ferritin molecules across the wall of muscle capillaries. J Cell Biol 37: 277299, 1968. 67. BUCCI C, WANDINGER-NESS A, LUTCKE A, CHIARIELLO M, BRUNI CB, AND ZERIAL M. Rab5a is a common component of the apical and basolateral endocytic machinery in polarized epithelial cells. Proc Natl Acad Sci USA 91: 50615065, 1994. 68. BURACK MA, SILVERMAN MA, AND BANKER G. The role of selective transport in neuronal protein sorting. Neuron 26: 465 472, 2000. 69. BURGER RL, MCHLMAN CS, AND ALLEN RH. Human placenta R-type vitamin B12-binding proteins. I. Isolation and characterization of transcobalamin I, transcobalamin III, and the normal granulocyte vitamin B12-binding protein. J Biol Chem 250: 7700 7706, 1975. 70. BURNS JL, GRIFFITH A, BARRY JJ, JONAS M, AND CHI EY. Transcytosis of gastrointestinal epithelial cells by Escherichia coli K1. Pediatr Res 49: 30 37, 2001. 71. BUTOR C AND DAVOUST J. Apical to basolateral surface area ratio and polarity of MDCK cells grown on different supports. Exp Cell Res 203: 115127, 1992. 72. BYE WA, ALLAN CH, AND TRIER JS. Structure, distribution, and origin of M cells in Peyers Patches of mouse ileum. Gastroenterology 86: 789 801, 1984. 73. CABANIOLS JP, RAVICHANDRAN V, AND ROCHE PA. Phosphorylation of SNAP-23 by the novel kinase SNAK regulates t-SNARE complex assembly. Mol Biol Cell 10: 4033 4041, 1999. 74. CALHOUN BC AND GOLDENRING JR. Two Rab proteins, vesicle-associated membrane protein 2 (VAMP-2) and secretory carrier membrane proteins (SCAMPs), are present on immunoisolated parietal cell tubulovesicles. Biochem J 325: 559 564, 1997. 75. CALVO M, POL A, LU A, ORTEGA D, PONS M, BLASI J, AND ENRICH C. Cellubrevin is present in the basolateral endocytic compartment of hepatocytes and follows the transcytotic pathway after IgA internalization. J Biol Chem 275: 7910 7917, 2000. 76. CAMPBELL L, HOLLINS AJ, AL-EID A, NEWMAN GR, VON RUHLAND C, AND GUMBLETON M. Caveolin-1 expression and caveolae biogenesis during cell transdifferentiation in lung alveolar epithelial primary cultures. Biochem Biophys Res Commun 262: 744 751, 1999. 77. CARDELL RR JR, BADENHAUSEN S, AND PORTER KR. Intestinal triglyceride absorption in the rat. An electron microscopical study. J Cell Biol 34: 123155, 1967. 78. CARDONE M AND MOSTOV K. Wortmannin inhibits transcytosis of dimeric IgA by the polymeric immunoglobulin receptor. FEBS Lett 376: 74 76, 1995. 79. CARDONE MH, SMITH BL, SONG W, MOCHLY-ROSEN D, AND MOSTOV KE. Phorbol myristate acetate-mediated stimulation of transcytosis and apical recycling in MDCK cells. J Cell Biol 124: 717727, 1994. 80. CARPENTER CL AND CANTLEY LC. Phosphoinositide kinases. Curr Opin Cell Biol 8: 153158, 1996. 81. CASANOVA JE, APODACA G, AND MOSTOV KE. An autonomous signal for basolateral sorting in the cytoplasmic domain of the polymeric immunoglobulin receptor. Cell 66: 6575, 1991. 82. CASANOVA JE, WANG X, KUMAR R, BHARTUR SG, NAVARRE J, WOODRUM JE, ALTSCHULER Y, RAY GS, AND GOLDENRING JR. Association of Rab25 and Rab11a with the apical recycling system of polarized Madin-Darby canine kidney cells. Mol Biol Cell 10: 47 61, 1999. 83. CASSIO D, HAMON-BENAIS C, GUERIN M, AND LECOQ O. Hybrid cell lines constitute a potential reservoir of polarized cells: isolation and study of highly differentiated hepatoma-derived hybrid cells able to form functional bile canaliculi in vitro. J Cell Biol 115: 13971408, 1991. www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

920

PAMELA L. TUMA AND ANN L. HUBBARD placental (BeWo) cells: evidence for an endogenous Cu oxidase. Placenta 21: 805 812, 2000. 108. DAUGHERTY A, CORNICELLI JA, WELCH K, SENDOBRY SM, AND RATERI DL. Scavenger receptors are present on rabbit aortic endothelial cells in vivo. Arterioscler Thromb Vasc Biol 17: 2369 2375, 1997. 109. DE ALMEIDA JB AND STOW JL. Disruption of microtubules alters polarity of basement membrane proteoglycan secretion in epithelial cells. Am J Physiol Cell Physiol 261: C691C700, 1991. 110. DEFFEBACH ME, BRYAN CJ, AND HOY CM. Protein movement across cultured guinea pig trachea: specicity and effect of transcytosis inhibitors. Am J Physiol Lung Cell Mol Physiol 271: L744 L752, 1996. 111. DEHOUCK B, DEHOUCK MP, FRUCHART JC, AND CECCHELLI R. Upregulation of the low density lipoprotein receptor at the blood-brain barrier: intercommunications between brain capillary endothelial cells and astrocytes. J Cell Biol 126: 465 473, 1994. 112. DEHOUCK B, FENART L, DEHOUCK-P, PIERCE A, TORPIER G, AND CECCHELLI R. A new function for the LDL receptor: transcytosis of LDL across the blood-brain barrier. J Cell Biol 138: 877 889, 1997. 113. DEHOUCK MP, JOLLIET-RIANT P, BREE F, FRUCHART J-C, CECCHELLI R, AND TILLEMENT J-P. Drug transfer across the blood-brain barrier: correlation between in vitro and in vivo models. J Neurochem 58: 1790 1797, 1992. 114. DEHOUCK MP, MERESSE S, DELORME P, FRUCHART J-C, AND CECCHELLI R. An easier, reproducible, and mass-production method to study the blood-brain barrier in vitro. J Neurochem 54: 1798 1801, 1990. 115. DEHOUCK MP, MERESSE S, DEHOUCK B, FRUCHART JC, AND CECCHELLI R. In vitro reconstituted blood-brain barrier. J Controlled Release 21: 8192, 1992. 116. DEHOUCK MP, MERESSE S, DELORME P, TORPIER G, FRUCHART J-C, AND CECCHELLI R. The blood-brain barrier in vitro: co-cultured of brain capillary endothelial cells and astrocytes. Circ Metab Cerveau 7: 151162, 1990. 117. DELGROSSI MH, BREUZA L, MIRRE C, CHAVRIER P, AND LEBIVIC A. Human syntaxin 3 is localized apically in human intestinal cells. J Cell Sci 110: 22072214, 1997. 118. DEL VECCHIO PJ, SIFLINGER-BIRNBOIM A, BELLONI PN, HOLLERAN LA, LUM H, AND MALIK AB. Culture and characterization of pulmonary microvascular endothelial cells. In Vitro Cell Dev Biol 28: 711715, 1992. 119. DE MARCO MC, KREMER L, ALBAR JP, MARTINEZ-MENARGUEZ JA, BALLESTA J, GARCIA-LOPEZ MA, MARAZUELA M, PUERTOLLANO R, AND ALONSO MA. BENE, a novel raft-associated protein of the MAL proteolipid family, interacts with caveolin-1 in human endothelial-like ECV304 cells. J Biol Chem 276: 23009 23017, 2001. 120. DE MARCO MC, MARTIN-BELMONTE F, KREMER L, ALBAR JP, CORREAS I, VAERMAN JP, MARAZUELA M, BYRNE JA, AND ALONSO MA. MAL2, a novel raft protein of the MAL family, is an essential component of the machinery for transcytosis in hepatoma HepG2 cells. J Cell Biol 159: 37 44, 2002. 122. DESCAMPS L, DEHOUCK M-P, TORPIER G, AND CECCHELLI R. Receptormediated transcytosis of transferrin through blood-brain barrier endothelial cells. Am J Physiol Heart Circ Physiol 270: H1149 H1158, 1996. 123. DESHPANDE D, TOLEDO-VELASQUEZ D, WANG LY, MALANGA CJ, MA JK, AND ROJANASAKUL Y. Receptor-mediated peptide delivery in pulmonary epithelial monolayers. Pharm Res 11: 11211126, 1994. 123a.DE VRIES KJ, GEIJTENBEEK A, BRIAN EC, DE GRAAN PN, GHIJSEN WE, AND VERHAGE M. Dynamics of munc18 1 phosphorylation/dephosphorylation in rat brain nerve terminals. Eur J Neurosci 12: 385 390, 2000. 124. DHARMSATHAPHORN K AND MADARA JL. Established intestinal cell lines as model systems for electrolyte transport studies. Methods Enzymol 192: 354 389, 1990. 125. DICKINSON BL, BADIZADEGAN K, WU Z, AHOUSE JC, ZHU X, SIMISTER NE, BLUMBERG RS, AND LENCER WI. Bidirectional FcRn-dependent IgG transport in a polarized human intestinal epithelial cell line. J Clin Invest 104: 903911, 1999. 126. DICKINSON TK AND CONNOR JR. Cellular distribution of iron, transferrin, and ferritin in the hypotransferrinemic (Hp) mouse brain. J Comp Neurol 355: 67 80, 1995. 127. DIETSCHY JM AND TURLEY SD. Cholesterol metabolism in the brain. Curr Opin Lipidol 12: 105112, 2001. www.prv.org

84. CASSIO D AND WEISS MC. Expression of fetal and neonatal hepatic functions by mouse hepatoma-rat hepatoma hybrids. Somatic Cell Genet 5: 719 738, 1979. 85. CECCHELLI R, DEHOUCK B, DESCAMPS L, FENART L, BUEE-SCHERRER VV, DUHEM C, LUNDQUIST S, RENTFEL M, TORPIER G, AND DEHOUCK MP. In vitro model for evaluating drug transport across the blood-brain barrier. Adv Drug Deliv Rev 36: 165178, 1999. 86. CEREIJIDO M, ROBBINS ES, DOLAN WJ, ROTUNNO CA, AND SABATINI DD. Polarized monolayers formed by epithelial cells on a permeable and translucent support. J Cell Biol 77: 853 880, 1978. 87. CHAMBERLAIN LH, BURGOYNE RD, AND GOULD GW. SNARE proteins are highly enriched in lipid rafts in PC12 cells: implications for the spatial control of exocytosis. Proc Natl Acad Sci USA 98: 5619 5624, 2001. 88. CHEN D AND WHITEHEART SW. Intracellular localization of SNAP-23 to endosomal compartments. Biochem Biophys Res Commun 255: 340 346, 1999. 89. CHINNASWAMY T, MAGNUS WS, BERGENFELDTS M, SASS P, AND MALIK AB. Gp60 activation mediates albumin transcytosis in endothelial cells by tyrosine kinase-dependent pathway. J Biol Chem 272: 25968 25975, 1997. 90. CHIU JH, HU CP, LUI WY, LO SJ, AND CHANG C. The formation of bile canaliculi in human hepatoma cell lines. Hepatology 11: 834 842, 1990. 91. CHRISTENSEN EI AND BIRN H. Megalin and cubilin: synergistic endocytic receptors in renal proximal tubule. Am J Physiol Renal Physiol 280: F562F573, 2001. 92. CHUNG SH, POLGAR J, AND REED GL. Protein kinase C phosphorylation of syntaxin 4 in thrombin-activated human platelets. J Biol Chem 275: 25286 25291, 2000. 93. COLLINS CA. Dynein-based organelle movement. In: Microtubules, edited by Hymans JS and Lloyd CW. New York: Wiley-Liss, 1994, p. 367380. 94. COLUCCIO LM. Identication of the microvillar 110-kDa calmodulin complex (Myosin-1) in kidney. Eur J Cell Biol 56: 286 294, 1991. 95. COLUCCIO LM AND CONATY C. Myosin-I in mammalian liver. Cell Motil Cytoskeleton 24: 189 199, 1993. 96. CORNFORD EM AND HYMAN S. Blood-brain barrier permeability to small and large molecules. Adv Drug Deliv Rev 36: 145163, 1999. 97. CORNICELLI JA, WITTE LD, AND GOODMAN DS. Inhibition of LDL degradation in cultured human broblasts induced by endothelial cellconditioned medium. Arteriosclerosis 3: 560 567, 1983. 98. CORVERA S AND CZECH MP. Direct targets of phosphoinositide 3-kinase products in membrane trafc and signal transduction. Trends Cell Biol 8: 442 446, 1998. 99. CORVERA S, ARRIGO AD, AND STENMARK H. Phosphoinositides in membrane trafc. Curr Opin Cell Biol 11: 460 465, 1999. 100. COX DS, RAJE S, GAO H, SALAMI NN, AND EDDINGTON ND. Enhanced permeability of molecular weight markers and poorly bioavailable compounds across Caco-2 cell monolayers using the absorption enhancer, zonula occludens toxin. Pharm Res 19: 1680 1688, 2002. 101. CRAIG LE, SPELMAN JP, STRANDBERG JD, AND ZINK MC. Endothelial cells from diverse tissues exhibit differences in growth and morphology. Microvasc Res 55: 6576, 1998. 102. CRAWFORD JM, VINTER DW, AND GOLLAN JL. Taurocholate induces pericanalicular localization of C6-NBD-ceramide in isolated hepatocyte couplets. Am J Physiol Gastrointest Liver Physiol 260: G119 G132, 1991. 103. CREUTZ CE. The annexins and exocytosis. Science 258: 924 931, 1992. 104. CROWE A AND MORGAN EH. Iron and transferrin uptake by brain and cerebrospinal uid in the rat. Brain Res 592: 8 16, 1992. 105. CUCULLO L, MCALLISTER MS, KIGHT K, KRIZANAC-BENGEZ L, MARRONI M, MAYBERG MR, STANNESS KA, AND JANIGRO D. A new dynamic in vitro model for the multidimensional study of astrocyte-endothelial cell interactions at the blood-brain barrier. Brain Res 951: 243254, 2002. 106. DAMKE H, BABA T, WARNOCK DE, AND SCHMID SL. Induction of mutant dynamin specically blocks endocytic coat formation. J Cell Biol 127: 915934, 1994. 107. DANZEISEN R, PONNAMBALAM S, LEA RG, PAGE K, GAMBLING L, AND MCARDLE HJ. The effect of ceruloplasmin on iron release from Physiol Rev VOL

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS 128. DONALDSON RM JR. How does cobalamin (vitamin B12) enter and traverse the ileal cell? Gastroenterology 88: 1069 1071, 1985. 129. DONALDSON RMJ. Transcellular transport of vitamin B12. Gastroenterology 100: 291, 1991. 130. DRAB M, VERKADE P, ELGER M, KASPER M, LOHN M, LAUTERBACH B, MENNE J, LINDSCHAU C, MENDE F, LUFT FC, SCHEDL A, HALLER H, AND KURZCHALIA TV. Loss of caveolae, vascular dysfunction, and pulmonary defects in caveolin-1 gene-disrupted mice. Science 293: 2449 2452, 2001. 131. DRENCKHAHN D AND JONS T. Cytoskeletonal basis for epithelial polarity. Kidney Blood Press Res 21: 238 240, 1998. 132. DREWES LR. What is the blood-brain barrier? A molecular perspective. Cerebral vascular biology. Adv Exp Med Biol 474: 111122, 1999. 133. DUFFY KR AND PARDRIDGE WM. Blood-brain barrier transcytosis of insulin in developing rabbits. Brain Res 420: 3238, 1987. 134. DUMAN JG, TYAGARAJAN K, KOLSI MS, MOORE HP, AND FORTE JG. Expression of rab11a N124I in gastric parietal cells inhibits stimulatory recruitment of the H-K-ATPase. Am J Physiol Cell Physiol 277: C361C372, 1999. 135. DUNN JC, YARMUSH ML, KOEBE HG, AND TOMPKINS RG. Hepatocyte function and extracellular matrix geometry: long-term culture in a sandwich conguration. FASEB J 3: 174 177, 1989. 136. DURRBACH A, RAPOSO G, TENZA D, LOUVARD D, AND COUDRIER E. Truncated brush border myosin I affects membrane trafc in polarized epithelial cells. Trafc 1: 411 424, 2000. 137. EILERS U, KLUMPERMAN J, AND HAURI HP. Nocodazole, a microtubuleactive drug, interferes with apical protein delivery in cultured intestinal epithelial cells (Caco-2). J Cell Biol 108: 1322, 1989. 138. ELFERINK RO AND GROEN AK. Genetic defects in hepatobiliary transport. Biochim Biophys Acta 1586: 129 145, 2002. 139. ELLINGER I, ROTHE A, GRILL M, AND FUCHS R. Apical to basolateral transcytosis and apical recycling of immunoglobulin G in trophoblast-derived BeWo cells: effects of low temperature, nocodazole, and cytochalasin D. Experientia 269: 322331, 2001. 140. ELLINGER I, SCHWAB M, STEFANESCU A, HUNZIKER W, AND FUCHS R. IgG transport across trophoblast-derived BeWo cells: a model system to study IgG transport in the placenta. Eur J Immunol 29: 733744, 1999. 141. ELLIS JA AND LUZIO JP. Identication and characterization of a novel protein (p137) which transcytoses bidirectionally in Caco-2 cells. J Biol Chem 270: 2071720723, 1995. 142. EMANS N, GORVEL JP, WALTER C, GERKE V, KELLNER R, GRIFFITHS G, AND GRUENBERG J. Annexin II is a major component of fusogenic endosomal vesicles. J Cell Biol 120: 13571369, 1993. 143. ERICSON LE AND NILSSON M. Structural and functional aspects of the thyroid follicular epithelium. Toxicol Lett 64 65: 365373, 1992. 144. FASSHAUER D, SUTTON RB, BRUNGER AT, AND JAHN R. Conserved structural features of the synaptic fusion complex: SNARE proteins reclassied as Q-and R-SNAREs. Proc Natl Acad Sci USA 95: 1578115786, 1998. 145. FATH KR AND BURGESS DR. Golgi-derived vesicles from developing epithelial cells bind actin laments and possess myosin-I as a cytoplasmically oriented peripheral membrane protein. J Cell Biol 120: 117127, 1993. 146. FATH KR, MAMAJIWALLA SN, AND BURGESS DR. The cytoskeleton in development of epithelial cell polarity. J Cell Sci Suppl 17: 6573, 1993. 147. FAWCETT DW. Surface specializations of absorbing cells. J Histochem Cytochem 13: 7591, 1965. 148. FERACCI H, RIGAL A, AND MAROUX S. Biosynthesis and intracellular pool of aminopeptidase N in rabbit enterocytes. J Membr Biol 83: 139 146, 1985. 149. FIEDLER K, KELLNER R, AND SIMONS K. Mapping the protein composition of trans-Golgi network (TGN)-derived carrier vesicles from polarized MDCK cells. Electrophoresis 18: 26132619, 1997. 150. FIEDLER K, LAFONT F, PARTON RG, AND SIMONS K. Annexin XIIIb: a novel epithelial specic annexin is implicated in vesicular trafc to the apical plasma membrane. J Cell Biol 128: 10431053, 1995. 150. FIELDING CJ AND FIELDING PE. Caveolae and intracellular trafcking of cholesterol. Adv Drug Delivery Rev 49: 251264, 2001. 151. FILLEBEEN C, DESCAMPS L, DEHOUCK MP, FENART L, BENAISSA M, SPIK G, CECCHELLI R, AND PIERCE A. Receptor-mediated transcytosis of Physiol Rev VOL

921

152.

153.

154.

155.

156.

157.

158.

159.

160.

161.

162. 163.

164.

165.

166.

167.

168.

169.

170.

171.

lactoferrin through the blood-brain barrier. J Biol Chem 274: 7011 7017, 1999. FISHMAN JB, RUBIN JB, HANDRAHAN JV, CONNOR JR, AND FINE RE. Receptor-mediated transcytosis of transferrin across the bloodbrain barrier. J Neurosci Res 18: 299 304, 1987. FOGH J, FOGH JM, AND ORFEO T. One hundred and twenty-seven cultured human tumor cell lines producing tumors in nude mice. J Natl Cancer Inst 59: 221226, 1977. FOLETTI DL, LIN R, FINLEY MA, AND SCHELLER RH. Phosphorylated syntaxin 1 is localized to discrete domains along a subset of axons. J Neurosci 20: 4535 4544, 2000. FOLKMAN J, HAUDENSCHILD CC, AND ZETTER BR. Long-term culture of capillary endothelial cells. Proc Natl Acad Sci USA 76: 52175221, 1979. FOLLI F, ALVARO D, GIGLIOZZI A, BASSOTTI C, KAHN CR, PONTIROLI AE, CAPOCACCIA L, JEZEQUEL AM, AND BENEDETTI A. Regulation of endocytic-transcytotic pathways and bile secretion by phosphatidylinositol 3-kinase in rats. Gastroenterology 113: 954 965, 1997. FOLSCH H, OHNO H, BONIFACINO JS, AND MELLMAN I. A novel clathrin adaptor complex mediates basolateral targeting in polarized epithelial cells. Cell 99: 189 198, 1999. FORSTNER GG AND WHERRETT JR. Plasma membrane and mucosal glycosphingolipids in the rat intestine. Biochim Biophys Acta 306: 446 459, 1973. FOSTER LJ, YEUNG B, MOHTASHAMI M, ROSS K, TRIMBLE WS, AND KLIP A. Binary interactions of the SNARE proteins syntaxin-4, SNAP23, and VAMP-2 and their regulation by phosphorylation. Biochemistry 37: 11089 11096, 1998. FREY A, GIANNASCA KT, WELSTZIN R, GIANNASCA PJ, REGGIO H, LENCER WI, AND NEUTRA MR. Role of the glycocalyx in regulating access of microparticles to apical plasma membranes of intestinal epithelial cells: implications for microbial attachment and oral vaccine targeting. J Exp Med 184: 10451059, 1996. FRIDEN PW, WALUS LR, MUSSO GF, TAYLOR MA, MALFROY B, AND STARZYK RM. Anti-transferrin receptor antibody and antibody-drug conjugates cross the blood-brain barrier. Proc Natl Acad Sci USA 88: 4771 4775, 1991. FRUMAN DA, MEYERS RE, AND CANTLEY LC. Phosphoinositide kinases. Annu Rev Biochem 67: 481507, 1998. FUJITA H, TUMA PL, FINNEGAN CM, LOCCO L, AND HUBBARD AL. Endogenous syntaxins 2, 3 and 4 exhibit distinct but overlapping patterns of expression at the hepatocyte plasma membrane. Biochem J 329: 527538, 1998. FUJITA Y, SASAKI T, FUKUI K, KOTANI H, KIMURA T, HATA Y, SUDHOF TC, SCHELLER RH, AND TAKAI Y. Phosphorylation of Munc-18/n-Sec1/ rbSec1 protein kinase C. J Biol Chem 271: 72657268, 1996. FUKUTA M, OKADA H, IINUMA S, YANAI S, AND TOGUCHI H. Insulin fragments as a carrier for peptide delivery across the blood-brain barrier. Pharm Res 11: 16811688, 1994. FURUSE M, FURUSE K, SASAKI H, AND TSUKITA S. Conversion of zonulae occludentes from tight to leaky strand type by introducing claudin-2 into Madin-Darby canine kidney I cells. J Cell Biol 153: 263272, 2001. GAGESCU R, DEMAUREX N, PARTON RG, HUNZIKER W, HUBER LA, AND GRUENBERG J. The recycling endosome of Madin-Darby canine kidney cells is a mildly acidic compartment rich in raft components. Mol Biol Cell 11: 27752791, 2000. GAISANO HY, GHAI M, MALKUS PN, SHEU L, BOUQUILLON A, BENNETT MK, AND TRIMBLE WS. Distinct cellular locations of the syntaxin family of proteins in rat pancreatic acinar cells. Mol Biol Cell 7: 2019 2027, 1996. GAISANO HY, SHEU L, WONG PP, KLIP A, AND TRIMBLE WS. SNAP-23 is located in the basolateral plasma membrane of rat pancreatic acinar cells. FEBS Lett 414: 298 302, 1997. GAISANO HY, SHEU L, GRONDIN G, GHAI M, BOUQUILLON A, LOWE A, BEAUDOIN A, AND TRIMBLE WS. The vesicle-associated membrane protein family of proteins in rat pancreatic and parotid acinar cells. Gastroenterology 111: 16611669, 1996. GALLI T, ZAHRAOUI A, VAIDYANATHAN VV, RAPOSA G, TIAN JM, KARIN M, NIEMANN H, AND LOUVARD D. A novel tetanus neurotoxin-insensitive vesicle-associated membrane protein in SNARE complexes of the apical plasma membrane of epithelial cells. Mol Biol Cell 9: 1437 1448, 1998. www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

922

PAMELA L. TUMA AND ANN L. HUBBARD 193. GOLDENRING JR, SHEN KR, VAUGHAN HD, AND MODLIN IM. Identication of a small GTP-binding protein, Rab25, expressed in the gastrointestinal mucosa, kindney and lung. J Biol Chem 268: 18419 18422, 1993. 194. GOMEZ-LECHON MJ, JOVER R, DONATO T, PONSODA X, RODRIGUEZ C, STENZEL KG, KLOCKE R, PAUL D, GUILLEN I, BORT R, AND CASTELL JV. Long-term expression of differentiated functions in hepatocytes cultured in three-dimensional collagen matrix. J Cell Physiol 177: 553562, 1998. 195. GONZALEZ L AND SCHELLER RH. Regulation of membrane trafcking: structural insights from a Rab/effector complex. Cell 96: 755758, 1999. 196. GORDON MM, HOWARS T, BECICH MJ, AND ALPERS DH. Cathepsin L mediates intracellular ileal digestion of gastric intrinsic factor. Am J Physiol Gastrointest Liver Physiol 268: G33G40, 1995. 197. GOTTLIEB TA, IVANOV IE, ADESNIK M, AND SABATINI DA. Actin microlaments play a critical role in endocytosis at the apical but not the basolateral surface of polarized epithelial cells. J Cell Biol 120: 695710, 1993. 198. GRAF GA, LI WP, GERARD RD, GELISSEN I, WHITE A, COHEN JC, AND HOBBS HH. Coexpression of ATP-binding cassette proteins ABCG5 and ABCG8 permits their transport to the apical surface. J Clin Invest 110: 659 669, 2002. 199. GRINDSTAFF KK, BACALLAO RL, AND NELSON JW. Apiconuclear organization of microtubules does not specify protein delivery from the trans-Golgi network to different membrane domains in polarized epithelial cells. Mol Biol Cell 9: 685 699, 1998. 200. GRINDSTAFF KK, YEASMAN C, ANANDASABAPATHY N, HSU S-C, RODRIGUEZ-BOULAN E, SCHELLER RH, AND NELSON WJ. Sec6/8 complex is recruited to cell-cell contacts and species transport vesicle delivery to the basal-lateral membrane in epithelial cells. Cell 93: 731 740, 1998. 201. GUMBLETON M AND AUDUS KL. Progress and limitations in the use of in vitro cell cultures to serve as a permeability screen for the blood-brain barrier. J Pharm Sci 90: 16811698, 2001. 202. GUNSHIN H, MACKENZIE B, BERGER UV, GUNSHIN Y, ROMERA MF, BORON WF, NUSSBERGER S, GOLLAN JL, AND HEDIGER MA. Cloning and characterization of a mammalian proton-coupled metal-ion transporter. Nature 388: 482 488, 1997. 203. GUTIERREZ JA AND WESSLING-RESNICK M. Molecular mechanisms of iron transport. Crit Rev Eukaryot Gene Expr 6: 114, 1996. 204. HAAS A. NSFfusion and beyond. Trends Cell Biol 8: 471 473, 1998. 205. HAILSTONES D, SLEER LS, PARTON RG, AND STANLEY KK. Regulation of caveolin and caveolae by cholesterol in MDCK cells. J Lipid Res 39: 369 379, 1998. 206. HALL CA, GREEN-COLLIGAN PD, AND BEGLEY JA. Synthesis of transcobalamin II by cultured human hepatocytes. Biochim Biophys Acta 838: 387389, 1985. 207. HAMAD AR, MARRACK P, AND KAPPLER JW. Transcytosis of staphylococcal superantigen toxins. J Exp Med 185: 14471454, 1997. 208. HAMMERTON RW, KRZEMINSKI KA, MAYS RW, RYAN TA, WOLLNER DA, AND NELSON WJ. Mechanism for regulating cell surface distribution of Na,K-ATPase in polarized epithelial cells. Science 254: 847 850, 1991. 209. HANDLER JS, PERKINS FM, AND JOHNSON JP. Studies of renal cell function using cell culture techniques. Am J Physiol Renal Fluid Electrolyte Physiol 238: F1F9, 1980. 210. HANSEN GH, NIELS-CHRISTIANSEN LL, IMMERDAL L, HUNZIKER W, KENNY AJ, AND DANIELSEN EM. Transcytosis of immunoglobulin A in the mouse enterocyte occurs through glycolipid raft- and rab17-containing compartments. Gastroenterology 116: 610 622, 1999. 211. HANSEN SH AND CASANOVA JE. Gsa stimulates transcytosis and apical secretion in MDCK cells through cAMP and protein kinase A. J Cell Biol 126: 677 687, 1994. 212. HANSEN SH, OLSSON A, AND CASANOVA JE. Wortmannin, an inhibitor of phosphoinositide 3-kinase, inhibits transcytosis in polarized epithelial cells. J Biol Chem 270: 2842528432, 1995. 213. HAO M, LIN SX, KARYLOWSKI OJ, WUSTNER D, MCGRAW TE, AND MAXFIELD FR. Vesicular and non-vesicular sterol transport in living cells. The endocytic recycling compartment is a major sterol storage organelle. J Biol Chem 277: 609 617, 2002. 214. HARDER T AND SIMONS K. Caveolae, DIGs, and the dynamics of www.prv.org

172. GAMBLING L, DANZEISEN R, GAIR S, LEA RG, CHARANIA Z, SOLANKY N, JOORY KD, SRAI SK, AND MCARDLE HJ. Effect of iron deciency on placental transfer of iron and expression of iron transport proteins in vivo and in vitro. Biochem J 356: 883 889, 2001. 173. GAN Y, MCGRAW TE, AND RODRIGUEZ-BOULAN E. The epithelial-specic adaptor AP1B mediates postendocytic recycling to the basolateral membrane. Nat Cell Biol 4: 605 609, 2002. 174. GARCIA I, SORDAT B, RAUCCIO-FARINON E, DUNAND M, KRAEHENBUHL JP, AND DIGGELMANN H. Establishment of two rabbit mammary epithelial cell lines with distinct oncogenic potential and differentiated phenotype after microinjection of transforming genes. Mol Cell Biol 6: 1974 1982, 1986. 175. GEBERT A, ROTHKOTTER HJ, AND PABST R. M cells in Peyers patches of the intestine. Int Rev Cytol 167: 91159, 1996. 176. GEUZE HJ, SLOT JW, STROUS GJAM, PEPPARD J, VON FIGURA K, HASILIK A, AND SCHWARTZ AL. Intracellular receptor sorting during endocytosis: comparative immunoelectron microscopy of multiple receptors in rat liver. Cell 37: 195204, 1984. 177. GHETIE V, HUBBARD JG, KIM JK, TSEN MF, LEE Y, AND WARD ES. Abnormally short serum half-lives of IgG in beta 2-microglobulindecient mice. Eur J Immunol 26: 690 696, 1996. 178. GHETIE V AND WARD ES. FcRn: the MHC class I-related receptor that is more than an IgG transporter. Immunol Today 18: 592598, 1997. 179. GHINEA N, ESKENASY M, SIMIONESCU M, AND SIMIONESCU N. Endothelial albumin binding proteins are membrane-associated components exposed on the cell surface. J Biol Chem 264: 4755 4758, 1989. 180. GHINEA N, FIXMAN A, ALEXANDRU D, POPOV D, HASU M, GHITESCU L, ESKENASY M, SIMIONESCU M, AND SIMIONESCU N. Identication of albumin-binding proteins in capillary endothelial cells. J Cell Biol 107: 231239, 1988. 181. GHINEA N, HAI MTV, GROYER-PICARD MT, AND MILGROM E. How protein hormones reach their target cells. Receptor-mediated transcytosis of hCG through endothelial cells. J Cell Biol 125: 8797, 1994. 182. GHINEA N AND MILGROM E. A new function for the LH/CG receptor: transcytosis of hormone across the endothelial barrier in target organs. Semin Reprod Med 19: 97101, 2001. 183. GHITESCU L, FIXMAN A, SIMIONESCU M, AND SIMIONESCU N. Specic binding sites for albumin restricted to plasmalemmal vesicles of continuous capillary endothelium: receptor-mediated transcytosis. J Cell Biol 102: 1304 1311, 1986. 184. GHITESCU LAB M. Transendothelial transport of serum albumin: a quantitative immunocytochemical study. Cell Biol 117: 745755, 1992. 185. GHITESCU Z, GAIS M, AND SIMIONESCU N. Differentiated uptake and transcytosis of albumin in successive vascular segments. J Submicrosc Cytol Pathol 20: 657 669, 1988. 186. GIANNASCA PJ, GIANNASCA KT, FALK P, GORDON JI, AND NEUTRA MR. Regional differences in glycoconjugates of intestinal M cells in mice: potential targets for mucosal vaccines. Am J Physiol Gastrointest Liver Physiol 267: G1108 G1121, 1994. 187. GIANNASCA PJ, GIANNASCA KT, LEICHTNER AM, AND NEUTRA MR. Human intestinal M cells display the sialyl Lewis A antigen. Infect Immun 67: 946 953, 1999. 188. GIARD DJ, AARONSON SA, TODARO GJ, ARNSTEIN P, KERSEY JH, DOSIK H, AND PARKS WP. In vitro cultivation of human tumors: establishment of cell lines derived from a series of solid tumors. J Natl Cancer Inst 51: 14171423, 1973. 189. GIFFROY D, LANGENDRIES A, MAURICE M, DANIEL F, LARDEUX B, COURTOY PJ, AND VAERMAN JP. In vivo stimulation of polymeric Ig receptor transcytosis by circulating polymeric IgA in rat liver. Int Immunol 10: 347354, 1998. 190. GILBERT T, LEBIVIC A, QUARONI A, AND RODRIGUEZ-BOULAN E. Microtubular organization and its involvement in the biogenetic pathways of plasma membrane proteins in Caco-2 intestinal epithelial cells. J Cell Biol 113: 275288, 1991. 191. GLENNEY JRJ AND SOPPET D. Sequence and expression of caveolin, a protein component of caveolae plasma membrane domains phosphorylated on tyrosine in Rous sarcoma virus-transformed broblasts. Proc Natl Acad Sci USA 89: 1051710521, 1992. 192. GLIEMANN J. Receptors of the low density lipoprotein (LDL) receptor family in man. Multiple functions of the large family members via interaction with complex ligands. Biol Chem 379: 951964, 1998. Physiol Rev VOL

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS sphingolipid-cholesterol microdomains. Curr Opin Cell Biol 9: 534 542, 1997. HASSON T AND MOOSEKER MS. Porcine myosin-VI: characterization of a new mammalian unconventional myosin. J Cell Biol 127: 425 440, 1994. HAY JC AND SCHELLER RH. SNAREs and NSF in targeted membrane fusion. Curr Biol 9: 505512, 1997. HAYAKAWA T, BRUCK R, NG OC, AND BOYER JL. Dibutyryladenosine 3,5-cyclic monophosphate stimulates vesicle transport and HRP excretion in isolated perfused rat liver. Am J Physiol Gastrointest Liver Physiol 259: G727G735, 1990. HEINTZELMAN MB, HASSON T, AND MOOSEKER MS. Multiple unconventional myosin domains of the intestinal brush border cytoskeleton. J Cell Sci 107: 35353543, 1994. HEISKALA M, PETERSON PA, AND YANG Y. The roles of Claudin superfamily proteins in paracellular transport. Trafc 2: 9398, 2001. HELMS JB. Role of heterotrimeric GTP binding proteins in vesicular protein transport: indications for both classical and alternative G protein cycles. FEBS Lett 369: 84 88, 1995. HEMAR A, OLIVO JC, WILLIAMSON E, SAFFRICH R, AND DOTTI CG. Dendroaxonal transcytosis of transferrin in cultured hippocampal and sympathetic neurons. J Neurosci 17: 9026 9034, 1997. HEMERY I, DURAND-SCHNEIDER AM, FELDMANN G, VAERMAN JP, AND MAURICE M. The transcytotic pathway of an apical plasma membrane protein (B10) in hepatocytes is similar to that of IgA and occurs via a tubular pericentriolar compartment. J Cell Sci 109: 12151227, 1996. HENLEY JR, KRUEGER EWA, OSWALD BJ, AND MCNIVEN MA. Dynaminmediated internalization of caveolae. J Cell Biol 141: 8599, 1998. HERMAN PK AND EMR SD. Characterization of VPS34, a gene required for vacuolar protein sorting and vacuole segregation in Saccharomyces cerevisiae. Mol Cell Biol 10: 6742 6754, 1990. HERMAN PK, STACK JH, DEMODENA JA, AND EMR SD. A novel protein kinase homolog essential for protein sorting to the yeast lysosomelike vacuole. Cell 64: 425 437, 1991. HERZOG V. Pathways of endocytosis in thyroid follicle cells. Int Rev Cytol 91: 107139, 1984. HERZOG V. Transcytosis in thyroid follicle cells. J Cell Biol 97: 607 617, 1983. HERZOG V AND FARQUHAR MG. Use of electron-opaque tracers for studies on endocytosis and membrane recycling. Methods Enzymol 98: 203225, 1983. HIRASE T, STADDON JM, SAITOU M, ANDO-AKATSUKA Y, ITOH M, FURUSE M, FUJIMOTO K, TSUKITA S, AND RUBIN LL. Occludin as a possible determinant of tight junction permeability in endothelial cells. J Cell Sci 110: 16031613, 1997. HIRLING H AND SCHELLER RH. Phosphorylation of synaptic vesicle proteins: modulation of the aSNAP interaction with the core complex. Proc Natl Acad Sci USA 93: 1194511949, 1996. HIRT RP, HUGHES GJ, FRUTIGER S, MICHETTI P, PERREGAUX C, POULAINGODEFROY O, JEANGUENAT N, NEUTRA MR, AND KRAEHENBUHL JP. Transcytosis of the polymeric Ig receptor requires phosphorylation of serine 664 in the absence but not the presence of dimeric IgA. Cell 74: 245255, 1993. HOCINI H AND BOMSEL M. Infectious human immunodeciency virus can rapidly penetrate a tight human epithelial barrier by transcytosis in a process impaired by mucosal immunoglobulins. J Infect Dis 179: S448 S453, 1999. HOEKSTRA D AND KOK JW. Trafcking of glycosphingolipids in eukaryotic cells: sorting and recycling of lipids. Biochim Biophys Acta 1113: 277294, 1992. HOEKSTRA D AND VAN IJZENDOORN SC. Lipid trafcking and sorting: how cholesterol is lling gaps. Curr Opin Cell Biol 12: 496 502, 2000. HOEKSTRA D, ZEGERS MM, AND VAN IJZENDOORN SC. Membrane ow, lipid sorting and cell polarity in HepG2 cells: role of a subapical compartment. Biochem Soc Trans 27: 422 428, 1999. HOLASH JA, HARIK SI, PERRY G, AND STEWART PA. Barrier properties of testis microvessels. Proc Natl Acad Sci USA 90: 11069 11073, 1993. HOLTHUIS JC, POMORSKI T, RAGGERS RJ, SPRONG H, AND VAN MEER G. The organizing potential of sphingolipids in intracellular membrane transport. Physiol Rev 81: 1689 1723, 2001. Physiol Rev VOL

923

215.

216. 217.

218.

219. 220.

221.

222.

223. 224.

225.

226. 227. 228.

229.

230.

231.

232.

233.

234.

235.

236.

237.

238. HOPPE CA, CONNOLLY TP, AND HUBBARD AL. Transcellular transport of polymeric IgA in the rat hepatocyte: biochemical and morphological characterization of the transport pathway. J Cell Biol 101: 21132123, 1985. 239. HSU SC, TING AE, HAZUKA CD, DAVANGER S, KENNY JW, AND KEE Y. The mammalian brain rsec6/8 complex. Neuron 17: 1209 1219, 1996. 240. HUBER LA, PIMPLIKAR S, PARTON RG, VIRTA H, ZERIAL M, AND SIMONS K. Rab8, a small GTPase involved in vesicular trafc between the TGN and the basolateral plasma membrane. J Cell Biol 123: 35 45, 1993. 241. HUGHSON EJ AND HIRT RP. Assessment of cell polarity. In: Epithelial Cell Culture: A Practical Approach, edited by Shaw AJ. Oxford, UK: Oxford Univ. Press, 1996, p. 37 66. 242. HUIJBREGTS RP, TOPALOF L, AND BANKAITIS VA. Lipid metabolism and regulation of membrane trafcking. Trafc 1: 195202, 2000. 243. HULL RN, CHERRY WR, AND WEAVER GW. The origin and characteristics of a pig kidney cell strain, LLC-PK. In Vitro 12: 670 677, 1976. 244. HUNZIKER W. The calmodulin antagonist W-7 affects transcytosis, lysosomal transport, and recycling but not endocytosis. J Biol Chem 269: 2900329009, 1994. 245. HUNZIKER W AND KRAEHENBUHL J-P. Epithelial transcytosis of immunoglobulins. J Mammary Gland Biol Neoplasia 3: 287302, 1998. 246. HUNZIKER W, MALE P, AND MELLMAN I. Differential microtubule requirements for transcytosis in MDCK cells. EMBO J 9: 35153525, 1990. 247. HUNZIKER W AND PETERS PJ. Rab17 localizes to recycling endosomes and regulates receptor-mediated transcytosis in epithelial cells. J Biol Chem 273: 15734 15741, 1998. 248. HUWYLER J AND PARDRIDGE WM. Examination of blood-brain barrier transferrin receptor by confocal uorescent microscopy of unxed isolated rat brain capillaries. J Neurochem 70: 883 886, 1998. 249. IDRISS JM AND JONAS AJ. Vitamin B12 transport by rat liver lysosomal membrane vesicles. J Biol Chem 266: 9438 9441, 1991. 250. IHRKE G, MARTIN GV, SHANKS MR, SCHRADER M, SCHROER TA, AND HUBBARD AL. Apical plasma membrane proteins and endolyn-78 travel through a subapical compartment in polarized WIF-B hepatocytes. J Cell Biol 141: 115133, 1998. 251. IHRKE G, NEUFELD EB, MEADS T, SHANKS MR, CASSIO D, LAURENT M, SCHROER TA, PAGANO RE, AND HUBBARD AL. WIF-B cells: an in vitro model for studies of hepatocyte polarity. J Cell Biol 123: 17611775, 1993. 252. IKONEN E. Roles of lipid rafts in membrane transport. Curr Opin Cell Biol 13: 470 477, 2001. 253. IKONEN E, TAGAYA M, ULLRICH O, MONTECUCCO C, AND SIMONS K. Different requirements for NSF, SNAP, and Rab proteins in apical and basolateral transport in MDCK cells. Cell 81: 571580, 1995. 254. IMADA M, TATSUMI H, AND FUJITA H. Scanning electron microscopy of vascular architecture in the gastric mucosa of the golden hamster. Cell Tissue Res 250: 287293, 1987. 255. INOUE T, NIELSEN S, MANDON B, TERRIS J, KISHORE BK, AND KNEPPER MA. SNAP-23 in rat kidney: colocalization with aquaporin-2 in collecting duct vesicles. Am J Physiol Renal Physiol 275: F752 F760, 1998. 256. IOANNOU YA. Multidrug permeases and subcellular cholesterol transport. Nat Rev Mol Cell Biol 2: 657 668, 2001. 257. ISBERG RR AND BARNES P. Subversion of integrins by enteropathogenic Yersinia. J Cell Sci 114: 2128, 2001. 258. ISBERG RR, HAMBURGER Z, AND DERSCH P. Signaling and invasinpromoted uptake via integrin receptors. Microbes Infect 2: 793 801, 2000. 259. ISBERG RR AND LEONG JM. Multiple beta 1 chain integrins are receptors for invasin, a protein that promotes bacterial penetration into mammalian cells. Cell 60: 861 871, 1990. 260. JACKLE S, BEISIEGEL U, RINNINGER F, BUCK F, GRIGOLEIT A, BLOCK A, GROGER I, GRETEN H, AND WINDLER E. Annexin VI, a marker protein of hepatocytic endosomes. J Biol Chem 269: 1026 1032, 1994. 261. JACKMAN MR, SHURETY W, ELLIS JA, AND LUZIO JP. Inhibition of apical but not basolateral endocytosis of ricin and folate in Caco-2 cells by cytochalasin D. J Cell Sci 107: 25472556, 1994. 262. JAHN R AND SUDHOF TC. Membrane fusion and exocytosis. Annu Rev Biochem 68: 863911, 1999. 263. JANZER RC AND RAFF MC. Astrocytes induce blood-brain barrier properties in endothelial cells. Nature 325: 253257, 1987. www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

924

PAMELA L. TUMA AND ANN L. HUBBARD lated myelin protein in oligodendrocytes. J Neurosci Res 42: 413 422, 1995. KLIMAN HJ, NESTLER JE, SERMASI E, SANGER JM, AND STRAUSS JF III. Purication, characterization, and in vitro differentiation of cytotrophoblasts from human term placentae. Endocrinology 118: 15671582, 1986. KNIESEL U AND WOLBURG H. Tight junctions of the blood-brain barrier. Cell Mol Neurobiol 20: 5776, 2000. KNIGHT A, HUGHSON E, HOPKINS CR, AND CUTLER DF. Membrane protein trafcking through the common apical endosome compartment of polarized Caco-2 cells. Mol Biol Cell 6: 597 610, 1995. KOBAYASHI S, KONDO S, AND JUNI K. Permeability of peptides and proteins in human cultured alveolar A549 cell monolayer. Pharm Res 12: 11151119, 1995. KOJIMA K, UTSUMI H, OGAWA H, AND MATSUMOTO I. Highly polarized expression of carbohydrate-binding protein p33/41 (annexin IV) on the apical plasma membrane of epithelial cells in renal proximal tubules. FEBS Lett 342: 313318, 1994. KOYAMA H, GOODPASTURE C, MILLER MM, TEPLITZ RL, AND RIGGS AD. Establishement and characterization of a cell line from the American opossum (Didelphys virginia). In Vitro 14: 239 246, 1978. KOZYRAKI R. Cubilin, a multifunctional epithelial receptor: an overview. J Mol Med 79: 161167, 2001. KREMMER T, WISHER MH, AND EVANS WH. The lipid composition of plasma membrane subfractions originating from the three major functional domains of the rat hepatocyte cell surface. Biochim Biophys Acta 455: 655 664, 1976. KUME N, ARAI H, KAWAI C, AND KITA T. Receptors for modied low-density lipoproteins on human endothelial cells: different recognition for acetylated low-density lipoprotein and oxidized lowdensity lipoprotein. Biochim Biophys Acta 1091: 63 67, 1991. KURZCHALIA TV AND PARTON RG. Membrane microdomains and caveolae. Curr Opin Cell Biol 11: 424 431, 1999. LAFONT F, BURKHARDT JK, AND SIMONS K. Involvement of microtubule motors in basolateral and apical transport in kidney cells. Nature 372: 801 803, 1994. LAFONT F, LECAT S, VERKADE P, AND SIMONS K. Annexin XIIIb associates with lipid microdomains to function in apical delivery. J Cell Biol 142: 14131427, 1998. LAFONT F, VERKADE P, GALLI T, WIMMER C, LOUVARD D, AND SIMONS K. Raft association of SNAP receptors acting in apical trafcking in Madin-Darby canine kidney cells. Proc Natl Acad Sci USA 96: 3734 3738, 1999. LAMM ME. Interaction of antigens and antibodies at mucosal surfaces. Annu Rev Microbiol 51: 311340, 1997. LAMM ME. Current concepts in mucosal immunity. IV. How epithelial transport of IgA antibodies relates to host defense. Am J Physiol Gastrointest Liver Physiol 274: G614 G617, 1998. LANG T, BRUNS D, WENZEL D, RIEDEL D, HOLROYD P, THIELE C, AND JAHN R. SNAREs are concentrated in cholesterol-dependent clusters that dene docking and fusion sites for exocytosis. EMBO J 20: 22022213, 2001. LAPIERRE LA, KUMAR R, HALES CM, NAVARRE J, BHARTUR SG, BURNETTE JO, PROVANCE DW JR, MERCER JA, BAHLER M, AND GOLDENRING JR. Myosin vb is associated with plasma membrane recycling systems. Mol Biol Cell 12: 18431857, 2000. LARKIN JM, OSWALD BJ, BALAN VJ, LARUSSO NF, AND MCNIVEN MA. RAB3D, a small GTP binding protein implicated in regulated secretion, is expressed in rat hepatocytes (Abstract). Mol Biol Cell 5: 448a, 1994. LARKIN JM AND PALADE GE. Transcytotic vesicular carriers for polymeric IgA receptors accumulate in rat hepatocytes after bile duct ligation. J Cell Sci 98: 205216, 1991. LARSEN JE, AVVAKUMOV GV, HAMMOND GL, AND VOGEL LK. N-glycans are not the signal for apical sorting of corticosteroid binding globulin in MDCK cells. FEBS Lett 451: 19 22, 1999. LEBIVIC A, QUARONI A, NICHOLS B, AND RODRIGUEZ-BOULAN E. Biogenetic pathways of plasma membrane proteins in Caco-2, a human intestinal epithelial cell line. J Cell Biol 111: 13511361, 1990. LECAT S, VERKADE P, THIELE C, FIEDLER K, SIMONS K, AND LAFONT F. Different properties of two isoforms of annexin XIII in MDCK cells. J Cell Sci 113: 26072618, 2000. LEE EY, BARCELLOS-HOFF MH, CHEN LH, PARRY G, AND BISSELL MJ. www.prv.org

264. JAREB M AND BANKER G. The polarized sorting of membrane proteins expressed in cultured hippocampal neurons using viral vectors. Neuron 20: 855 867, 1998. 265. JIN M, SAUCAN L, FARQUHAR MG, AND PALADE GE. Rab1a and multiple other Rab proteins are associated with the transcytotic pathway in rat liver. J Biol Chem 271: 3010530113, 1996. 266. JINGA VV, GAFENCU A, ANTOHE F, CONSTANTINESCU E, HELTIANU C, RAICU M, MANOLESCU I, HUNZIKER W, AND SIMIONESCU M. Establishment of a pure vascular endothelial cell line from human placenta. Placenta 21: 325336, 2000. 267. JOHN TA, VOGEL SM, MINSHALL RD, RIDGE K, TIRUPPATHI C, AND MALIK AB. Evidence for the role of alveolar epithelial gp60 in active transalveolar albumin transport in the rat lung. J Physiol 533: 547559, 2001. 268. JOHN TA, VOGEL SM, TIRUPPATHI C, MALIK AB, AND MINSHALL RD. Quantitative analysis of albumin uptake and transport in the rat microvessel endothelial monolayer. Am J Physiol Lung Cell Mol Physiol 284: L187L196, 2003. 269. JOHNSON LG, CHENG PW, AND BOUCHER RC. Albumin absorption by canine bronchial epithelium. J Appl Physiol 66: 27722777, 1989. 270. JOU TS, LEUNG SM, FUNG LM, RUIZ WG, NELSON WJ, AND APODACA G. Selective alterations in biosynthetic and endocytic protein trafc in Madin-Darby canine kidney epithelial cells expressing mutants of the small GTPase Rac1. Mol Biol Cell 11: 287304, 2000. 271. JUILLERAT-JEANNERET L, AGUZZI A, WIESTLER OD, DAREKAR P, AND JANZER RC. Dexamethasone selectively regulates the activity of enzymatic markers of cerebral endothelial cell lines. In Vitro Cell Dev Biol 28: 537543, 1992. 272. JUVET LK, BERG T, AND GJOEN T. The expression of endosomal rab proteins correlates with endocytic rate in rat liver cells. Hepatology 25: 1204 1212, 1997. 273. KAETZEL CS. Polymeric Ig receptor: defender of the fort or Trojan horse? Curr Biol 11: R35R38, 2001. 274. KAETZEL MA, PULA G, CAMPOS B, UHRIN P, HORSEMAN N, AND DEDMAN JR. Annexin VI isoforms are differentially expressed in mammalian tissues. Biochim Biophys Acta 1223: 368 374, 1994. 275. KAISER CA AND SCHEKMAN R. Distinct sets of SEC genes govern transport vesicle formation and fusion early in the secretory pathway. Cell 61: 723733, 1990. 276. KAPADIA CR. Vitamin B12 in health and disease: part Iinherited disorders of function, absorption, and transport. Gastroenterologist 3: 329 344, 1995. 277. KAPLAN J. Mechanisms of cellular iron acquisition. Another iron in the re. Cell 111: 603 606, 2002. 278. KAPLAN J AND OHALLORAN TV. Iron metabolism in eukaryotes: Mars and Venus at it again. Science 271: 1510 1512, 1996. 279. KARKI S AND HOLZBAUR ELF. Cytoplasmic dynein and dynactin in cell division and intracellular transport. Curr Opin Cell Biol 11: 4553, 1999. 280. KAUFMAN SS, TUMA DJ, PARK JH, AND GRANDJEAN CJ. Effects of cytochalasin B on the synthesis and secretion of plasma proteins by developing rat liver. J Pediatr Gastroenterol Nutr 7: 107114, 1988. 281. KAWAI K, FUJITA M, AND NAKAO M. Lipid componenets of two different regions of an intestinal epithelial cell membrane of mouse. Biochim Biophys Acta 369: 222233, 1974. 282. KEE Y, YOO JS, HAZUKA CD, PETERSON KE, HSU SC, AND SCHELLER RH. Subunit structure of the mammalian exocyst complex. Proc Natl Acad Sci USA 94: 14438 14443, 1997. 283. KENNEDY ML, DOUGLAS GC, AND KING BF. Effect of iron on transferrin receptor expression by human placental syncytiotrophoblast cells. Reprod Fertil Dev 9: 609 616, 1997. 284. KERNEIS S, BOGDANOVA A, KRAEHENBUHL JP, AND PRINGAULT E. Conversion by Peyers patch lymphoctyes of human enterocytes into M cells that transport bacteria. Science 277: 949 952, 1997. 285. KERNEIS S, CALIOT E, STUBBE H, BOGDANOVA A, KRAEHENBUHL J, AND PRINGAULT E. Molecular studies of the intestinal mucosal barrier physiopathology using cocultures of epithelial and immune cells: a technical update. Microbes Infect 2: 1119 1124, 2000. 286. KIM KS. Escherichia coli translocation at the blood-brain barrier. Infect Immun 69: 52175222, 2001. 287. KIM T, FIEDLER K, MADISON DL, KRUEGER WH, AND PFEIFFER SE. Cloning and characterization of MVP17: a developmentally reguPhysiol Rev VOL

288.

289. 290.

291.

292.

293.

294. 295.

Downloaded from physrev.physiology.org on November 4, 2009

296.

297. 298.

299.

300.

301. 302.

303.

304.

305.

306.

307.

308.

309.

310.

83 JULY 2003

TRANSCYTOSIS Transferrin is a major mouse milk protein and is synthesized by mammary epithelial cells. In Vitro Cell Dev Biol 23: 221226, 1987. LEHESTE JR, ROLINSKI B, VORUM H, HILPERT J, NYKJAER A, JACOBSEN C, AUCOUTURIER P, MOSKAUG JO, OTTO A, CHRISTENSEN EI, AND WILLNOW TE. Megalin knockout mice as an animal model of low molecular weight proteinuria. Am J Pathol 155: 13611370, 1999. LELOUP C, ARLUISON M, KASSIS N, LEPETIT N, CARTIER N, FERRE P, AND PENICAUD L. Discrete brain areas express the insulin-responsive glucose transporter GLUT4. Brain Res 38: 4553, 1996. LENCER WI, MOE S, RUFO PA, AND MADARA JL. Transcytosis of cholera toxin subunits across model human intestinal epithelia. Proc Natl Acad Sci USA 92: 10094 10098, 1995. LEUNG SM, CHEN D, DASGUPTA BR, WHITEHEART SW, AND APODACA G. SNAP-23 requirement for transferrin recycling in streptolysin-Opermeabilized Madin-Darby canine kidney cells. J Biol Chem 273: 1773217741, 1998. LEUNG SM, ROJAS R, MAPLES C, FLYNN C, RUIZ WG, JOU TS, AND APODACA G. Modulation of endocytic trafc in polarized MadinDarby canine kidney cells by the small GTPase RhoA. Mol Biol Cell 10: 4369 4384, 1999. LEUNG SM, RUIZ WG, AND APODACA G. Sorting of membrane and uid at the apical pole of polarized Madin-Darby canine kidney cells. Mol Biol Cell 11: 21312150, 2000. LEVINE JS, NAKANE PK, AND ALLEN RH. Human intrinsic factor secretion: immunocytochemical demonstration of membrane-associated vesicular transport in parietal cells. J Cell Biol 90: 644 655, 1981. LI S, SEITZ R, AND LISANTI MP. Phosphorylation of caveolin by Src tyrosine kinases. J Biol Chem 271: 38633868, 1996. LIEBNER S, KNIESEL U, KALBACHER H, AND WOLBURG H. Correlation of tight junction morphology with the expression of tight junction proteins in blood-brain barrier endothelial cells. Eur J Cell Biol 79: 707717, 2000. LIN SX, FERRO KL, AND COLLINS CA. Cytoplasmic dynein undergoes intracellular redistribution concomitant with phosphorylation of the heavy chain in response to serum starvation and okadaic acid. J Cell Biol 127: 1009 1019, 1994. LIONNE C, BUSS F, HODGE T, IHRKE G, AND KENDRICK-JONES J. Localization of myosin Va is dependent on the cytoskeletal organization in the cell. Biochem Cell Biol 79: 93106, 2001. LIPARDI C, MORA R, COLOMER V, PALADINO S, NITSCH L, RODRIGUEZBOULAN E, AND ZURZOLO C. Caveolin transfection results in caveolae formation but not apical sorting of glycosylphosphatidylinositol (GPI)-anchored proteins in epithelial cells. J Cell Biol 140: 617 626, 1998. LIPPOLDT A, LIEBNER S, ANDBJER B, KALBACHER H, WOLBURG H, HALLER H, AND FUXE K. Organization of choroid plexus epithelial and endothelial cell tight junctions and regulation of claudin-1, -2 and -5 expression by protein kinase C. Neuroreport 11: 14271431, 2000. LIPSCHUTZ JH, GUO W, LE OB, NGUYEN YH, NOVICK P, AND MOSTOV KE. Exocyst is involved in cystogenesis and tubulogenesis and acts by modulating synthesis and delivery of basolateral plasma membrane and secretory proteins. Mol Biol Cell 11: 4259 4275, 2000. LISANTI MP, LEBIVIC A, SARGIACOMO M, AND RODRIGUEZ-BOULAN E. Steady-state distribution and biogenesis of endogenous MadinDarby canine kidney glycoproteins: evidence for intracellular sorting and polarized cell surface delivery. J Cell Biol 109: 21172127, 1989. LISANTI MP, SCHERER PE, VIDUGIRIENE J, TANG Z, HERMANOWSKI-VOSATKA A, TU YH, COOK RF, AND SARGIACOMO M. Characterization of caveolin-rich membrane domains isolated from an endothelial-rich source: implications for human disease. J Cell Biol 126: 111126, 1994. LIU F, SOARES MJ, AND AUDUS KL. Permeability properties of monolayers of the human trophoblast cell line BeWo. Am J Physiol Cell Physiol 273: C1596 C1604, 1997. LIU X, BROUWER KL, GAN LS, BROUWER KR, STIEGER B, MEIER PJ, AUDUS KL, AND LECLUYSE EL. Partial maintenance of taurocholate uptake by adult rat hepatocytes cultured in a collagen sandwich conguration. Pharm Res 15: 15331539, 1998. LLORENTE A, GARRED O, HOLM PK, EKER P, JACOBSEN J, VAN DEURS B, AND SANDVIG K. Effect of calmodulin antagonists on endocytosis and Physiol Rev VOL

925

311.

330.

312.

331.

313.

332.

314.

333.

315.

334.

316.

335. 336. 337.

317.

318. 319.

338.

320.

339.

340.

321.

322.

341.

342.

323.

343.

324.

325.

344.

345.

326.

346.

327.

347.

328.

348.

329.

intracellular transport of Ricin in polarized MDCK cells. Exp Cell Res 227: 298 308, 1996. LOMAN S, RADL J, JANSEN HM, OUT TA, AND LUTTER R. Vectorial transcytosis of dimeric IgA by the Calu-3 human lung epithelial cell line: upregulation by IFN-. Am J Physiol Lung Cell Mol Physiol 272: L951L958, 1997. LOW SH, CHAPIN SJ, WEIMBS T, KOMUVES LG, BENNETT MK, AND MOSTOV KE. Differential localization of syntaxin isoforms in polarized Madin-Darby canine kidney cells. Mol Biol Cell 7: 20072018, 1996. LOW SH, CHAPIN SJ, WIMMER C, WHITEHEART SW, KOMUVES LG, MOSTOV KE, AND WEIMBS T. The SNARE machinery is involved in apical plasma membrane trafcking in MDCK cells. J Cell Biol 141: 15031513, 1998. LOW SH, ROCHE PA, ANDERSON HA, VAN IJZENDOORN SCD, ZHANG M, MOSTOV KE, AND WEIMBS T. Targeting of SNAP-23 and SNAP-25 in polarized epithelial cells. J Biol Chem 273: 34223430, 1998. LOW SH, WONG SH, TANG BL, AND HONG W. Involvement of both vectorial and transcytotic pathways in the preferential apical cell surface localization of rat dipeptidyl peptidase IV in transfected LLC-PK1 cells. J Biol Chem 266: 19710 19716, 1991. LUDGER J AND GALLI T. Exocytosis: SNAREs drum up! Eur J Neurosci 10: 415 422, 1998. LUM H AND MALIK AB. Mechanisms of increased endothelial permeability. Can J Physiol Pharmacol 74: 787 800, 1996. LUTCKE A, JANSSON S, PARTON RG, CHAVRIER P, VALENCIA A, HUBER LA, LEHTONEN E, AND ZERIAL M. Rab17, a novel small GTPase, is specic for epithelial cells and is induced during cell polarization. J Cell Biol 121: 553564, 1993. LUTCKE A, PARTON RG, MURPHY C, OLKKONEN VM, DUPREE P, VALENCIA A, SIMONS K, AND ZERIAL M. Cloning and subcellular localization of novel rab proteins reveals polarized and cell type-specic expression. J Cell Sci 107: 34373448, 1994. LUTON F, CARDONE MH, ZHANG M, AND MOSTOV KE. Role of tyrosine phosphorylation in ligand-induced regulation of transcytosis of the polymeric Ig receptor. Mol Biol Cell 9: 17871802, 1998. LUTON F AND MOSTOV KE. Transduction of basolateral-to-apical signals across epithelial cells: ligand-stimulated transcytosis of the polymeric immunoglobulin receptor requires two signals. Mol Biol Cell 10: 1409 1427, 1999. LUTON F, VERGES M, VAERMAN J-P, SUDOL M, AND MOSTOV KE. The SRC family protein tyrosine kinase p62yes controls polymeric IgA transcytosis in vivo. Mol Cell 4: 627 632, 1999. MA Y, SPECIAN RD, YEH KY, YEH M, RODRIGUEZ-PARIS J, AND GLASS J. The transcytosis of divalent metal transporter 1 and apo-transferrin during iron uptake in intestinal epithelium. Am J Physiol Gastrointest Liver Physiol 283: G965G974, 2002. MACKIC JB, STINS M, MCCOMB JG, CALERO M, GHISO J, KIM KS, YAN SD, STERN D, SCHMIDT AM, FRANGIONE B, AND ZLOKOVIC BV. Human blood-brain barrier receptors for Alzheimers amyloid-beta 1 40. Asymmetrical binding, endocytosis, and transcytosis at the apical side of brain microvascular endothelial cell monolayer. J Clin Invest 102: 734 743, 1998. MAKSYMOWYCH AB AND SIMPSON LL. Binding and transcytosis of botulinum neurotoxin by polarized human colon carcinoma cells. J Biol Chem 273: 21950 21957, 1998. MANDON B, CHOU CL, NIELSEN S, AND KNEPPER MA. Syntaxin-4 is localized to the apical plasma membrane of rat renal collecting duct cells: possible role in aquaporin-2 trafcking. J Clin Invest 98: 906 913, 1996. MAPLES CJ, RUIZ WG, AND APODACA G. Both microtubules and actin laments are required for efcient postendocytic trafc of the polymeric immunoglobulin receptor in polarized Madin-Darby canine kidney cells. J Biol Chem 272: 6741 6751, 1997. MARINO M, ANDREWS D, BROWN D, AND MCCLUSKEY RT. Transcytosis of retinol-binding protein across renal proximal tubule cells after megalin (gp 330)-mediated endocytosis. J Am Soc Nephrol 12: 637 648, 2001. MARINO M, CHIOVATO L, MITSIADES N, LATROFA F, ANDREWS D, TSELENIBALAFOUTA S, COLLINS AB, PINCHERA A, AND MCCLUSKEY RT. Circulating thyroglobulin transcytosed by thyroid cells is complexed with secretory components of its endocytic receptor megalin. J Clin Endocrinol Metab 85: 3458 3467, 2000. www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

926

PAMELA L. TUMA AND ANN L. HUBBARD MW, AND SIMPSON RJ. A novel duodenal iron-regulated transporter, IREG1, implicated in the basolateral transfer of iron to the circulation. Mol Cell 5: 299 309, 2000. MCNIVEN MA AND MARLOWE KJ. Contributions of molecular motor enzymes to vesicle-based protein transport in gastrointestinal epithelial cells. Gastroenterology 116: 438 451, 1999. MEADS T AND SCHROER TA. Polarity and nucleation of microtubules in polarized epithelial cells. Cell Motil Cytoskeleton 32: 273288, 1995. MEERSON NR, BELLO V, DELAUNAY JL, SLIMANE TA, DELAUTIER D, LENOIR C, TRUGNAN G, AND MAURICE M. Intracellular trafc of the ecto-nucleotide pyrophosphatase/phosphodiesterase NPP3 to the apical plasma membrane of MDCK and Caco-2 cells: apical targeting occurs in the absence of N-glycosylation. J Cell Sci 113: 4193 4202, 2000. MERESSE S, DELBART C, FRUCHART J-C, AND CECCHELLI R. Low-density lipoprotein receptor on endothelium of brain capillaries. J Neurochem 53: 340 345, 1989. MICHEL CC AND CURRY FE. Microvascular permeability. Physiol Rev 79: 703761, 1999. MILICI AJ, WATROUS NE, STUKENBROK H, AND PALADE GE. Transcytosis of albumin in capillary endothelium. J Cell Biol 105: 26032612, 1987. MINSHALL RD, TIRUPPATHI C, VOGEL SM, NILES WD, GILCHRIST A, HAMM HE, AND MALIK AB. Endothelial cell-surface gp60 activates vesicle formation and trafcking via G(i)-coupled Src kinase signaling pathway. J Cell Biol 150: 10571070, 2000. MOE AJ. Placental amino acid transport. Am J Physiol Cell Physiol 268: C1321C1331, 1995. MOESTRUP SK, BIRN H, FISCHER PB, PETERSON CM, VERROUST PJ, SIM RS, CHRISTENSEN EI, AND NEXO E. Megalin-mediated endocytosis of transcobalamin-vitamin12 complexes suggests a role of the receptor in vitamin-B12 homeostasis. Proc Natl Acad Sci USA 93: 8612 8617, 1996. MONKS JA AND NEVILLE MC. Transcytosis of proteins across the mammary epithelium into milk. J Womens Cancer 2: 193200, 2000. MOORE KL AND PERSAUD TVN. The Developing Human. Philadelphia, PA: Saunders, 1993. MOOS T AND MORGAN EH. Transferrin and transferrin receptor function in brain barrier systems. Cell Mol Neurobiol 20: 7795, 2000. MORITA K, SASAKI H, FURUSE M, AND TSUKITA S. Endothelial claudin: claudin-5/TMVCF constitutes tight junction strands in endothelial cells. J Cell Biol 147: 185194, 1999. MORROW JS, CIANCI CD, ARDITO T, MANN AS, AND KASHGARIAN M. Ankyrin links fodrin to the alpha subunit of Na,K-ATPase in MadinDarby canine kidney cells and in intact renal tubule cells. J Cell Biol 108: 455 465, 1989. MOSS SE. Annexins. Trends Cell Biol 7: 87 89, 1997. MOSTOV K AND KAETZEL CS. Immunoglobulin Transport and the Polymeric Immunoglobulin Receptor. San Diego, CA: Academic, 1999. MOSTOV KE. Transepithelial transport of immunoglobulins. Annu Rev Immunol 12: 63 84, 1994. MOSTOV KE, FRIEDLANDER M, AND BLOBEL G. The receptor for transepithelial transport of IgA and IgM contains multiple immunoglobulin-like domains. Nature 308: 37 43, 1984. MOSTOV KE, KRAEHENBUHL JP, AND BLOBEL G. Receptor-mediated transcellular transport of immunoglobulin: synthesis of secretory component as multiple and larger transmembrane forms. Proc Natl Acad Sci USA 77: 72577261, 1980. MOSTOV KE, TER BEEST MBA, AND CHAPIN SJ. Catch the mu1B train to the basolateral surface. Cell 99: 121122, 1999. MOSTOV KE, VERGES M, AND ALTSCHULER Y. Membrane trafc in polarized epithelial cells. Curr Opin Cell Biol 12: 483 490, 2000. MOSTOV KM AND SIMISTER NE. Transcytosis. Cell 45: 389 390, 1985. MULCAHY RT, ROSENKRANS WA JR, PENNEY DP, AND COOPER RA. The growth and morphology of FRTL-5 thyroid epithelial cells grown as multicellular spheroids in vitro. In Vitro Cell Dev Biol 21: 513520, 1985. NABI IR, MATHEWS AP, COHEN-GOULD L, GUNDERSEN D, AND RODRIGUEZ-BOULAN E. Immortalization of polarized rat retinal pigment epithelium. J Cell Sci 104: 37 49, 1993. www.prv.org

349. MARINO M AND MCCLUSKEY RT. Megalin-mediated transcytosis of thyroglobulin by thyroid cells is a calmodulin-dependent process. Thyroid 10: 461 469, 2000. 350. MARINO M AND MCCLUSKEY RT. Role of thyroglobulin endocytic pathways in the control of thyroid hormone release. Am J Physiol Cell Physiol 279: C1295C1306, 2000. 351. MARINO M, PINCHERA A, MCCLUSKEY RT, AND CHIOVATO L. Megalin in thyroid physiology and pathology. Thyroid 11: 4756, 2001. 352. MARINO M, ZHENG G, CHIOVATO L, PINCHERA A, BROWN D, ANDREWS D, AND MCCLUSKEY RT. Role of megalin (gp330) in transcytosis of thyroglobulin by thyroid cells. A novel function in the control of thyroid hormone release. J Biol Chem 275: 71257137, 2000. 353. MARTIN TF. PI(4,5)P(2) regulation of surface membrane trafc. Curr Opin Cell Biol 13: 493 499, 2001. 354. MARTINEZ O AND GOUD B. Rab proteins. Biochim Biophys Acta 1404: 101112, 1998. 355. MASSEY D, FERACCI H, GORVEL JP, RIGAL A, SOULIE JM, AND MAROUX S. Evidence for the transit of aminopeptidase N through the basolateral membrane before it reaches the brush border of enterocytes. J Membr Biol 96: 19 25, 1987. 356. MASSEY D, TRAVERSO V, RIGAL A, AND MAROUX S. Cellular and subcellular localization of annexin IV in rabbit intestinal epithelium, pancreas and liver. Biol Cell 73: 151156, 1991. 357. MASSEY-HARROCHE D, MAYRAN N, AND MAROUX S. Polarized localizations of annexins I, II, VI and XIII in epithelial cells of intestinal, hepatic and pancreatic tissues. J Cell Sci 111: 30073015, 1998. 358. MASTICK CC, BRADY MJ, AND SALTIEL AR. Insulin stimulates the tyrosine phosphorylation of caveolin. J Cell Biol 129: 15231531, 1995. 359. MASTICK CC AND FALICK AL. Association of N-ethylmaleimide sensitive fusion (NSF) protein and soluble NSF attachment proteinsalpha and -gamma with glucose transporter-4-containing vesicles in primary rat adipocytes. Endocrinology 138: 23912397, 1997. 360. MATLIN KS AND SIMONS K. Sorting of an apical plasma membrane glycoprotein occurs before it reaches the cell surface in cultured epithelial cells. J Cell Biol 99: 21312139, 1984. 361. MATTER K. Epithelial polarity: sorting out the sorters. Curr Biol 10: R39 R42, 2000. 362. MATTER K, BUCHER K, AND HAURI HP. Microtubule perturbation retards both the direct and the indirect apical pathway but does not affect sorting of plasma membrane proteins in intestinal epithelial cells (Caco-2). EMBO J 9: 31633170, 1990. 363. MAUCHAMP J, CHAMBARD M, GABRION J, AND VERRIER B. Polarized multicellular structures designed for the in vitro study of thyroid cell function and polarization. Methods Enzymol 98: 477 486, 1983. 364. MAXFIELD FR AND WUSTNER D. Intracellular cholesterol transport. J Clin Invest 110: 891 898, 2002. 365. MAYRAN N, TRAVERSO V, MAROUX S, AND MASSEY-HARROCHE D. Cellular and subcellular localizations of annexins, I, IV, and VI in lung epithelia. Am J Physiol Lung Cell Mol Physiol 270: L863L871, 1996. 366. MAYS RW, BECK KA, AND NELSON WJ. Organization and function of the cytoskeleton in polarized epithelial cells: a component of the protein sorting machinery. Curr Opin Cell Biol 6: 16 24, 1994. 367. MAYS RW, SIEMERS KA, FRITZ BA, LOWE AW, VAN MEER G, AND NELSON WJ. Hierarchy of mechanisms involved in generating Na/K-ATPase polarity in MDCK epithelial cells. J Cell Biol 130: 11051115, 1995. 368. MCBRIDE HM, RYBIN V, MURPHY C, GINER A, TEASDALE R, AND ZERIAL M. Oligomeric complexes link Rab5 effectors with NSF and drive membrane fusion via interactions between EEA1 and syntaxin 13. Cell 98: 377386, 1999. 369. MCCALL AL, VAN BUEREN AM, HUANG L, STENBIT A, CELNIK E, AND CHARRON MJ. Forebrain endothelium expresses GLUT4, the insulinresponsive glucose transporter. Brain Res 744: 318 326, 1997. 370. MCCARTHY KM, YOONG Y, AND SIMISTER NE. Bidirectional transcytosis of IgG by the rat neonatal Fc receptor expressed in a rat kidney cell line: a system to study protein transport across epithelia. J Cell Sci 113: 12771285, 2000. 371. MCINTOSH DP AND SCHNITZER JE. Caveolae require intact VAMP for targeted transport in vascular endothelium. Am J Physiol Heart Circ Physiol 277: H2222H2232, 1999. 372. MCKIE AT, MARCIANI P, ROLFS A, BRENNAN K, WEHR K, BARROW D, MIRET S, BOMFORD A, PETERS TJ, FARZANEH F, HEDIGER MA, HENTZE Physiol Rev VOL

373.

374.

375.

376.

377. 378.

Downloaded from physrev.physiology.org on November 4, 2009

379.

380. 381.

382.

383. 384. 385.

386.

387. 388.

389. 390.

391.

392. 393. 394. 395.

396.

83 JULY 2003

TRANSCYTOSIS 397. NATVIG IB, JOHANSEN FE, NORDENG TW, HARALDSEN G, AND BRANDTZAEG P. Mechanism for enhanced external transfer of dimeric IgA over pentameric IgM: studies of diffusion, binding to the human polymeric Ig receptor, and epithelial transcytosis. J Immunol 159: 4330 4340, 1997. 398. NESBITT SA AND HORTON MA. Trafcking of matrix collagens through bone-resorbing osteoclasts. Science 276: 266 269, 1997. 399. NETLAND PA, ZETTER BR, VIA DP, AND VOYTA JC. In situ labelling of vascular endothelium with uorescent acetylated low density lipoprotein. Histochem J 17: 1309 1320, 1985. 400. NEUTRA MR. Interactions of viruses and microparticles with apical plasma membranes of M cells: implications for human immunodeciency virus transmission. J Infect Dis 179: S441S443, 1999. 401. NEUTRA MR, FREY A, AND KRAEHENBUHL JP. Epithelial M cells: gateways for mucosal infection and immunization. Cell 86: 345348, 1996. 402. NEUTRA MR, GIANNASCA PJ, GIANNASCA KT, AND KRAEHENBUHL J-P. M cells and microbial pathogens. In: Infections of the Gastrointestinal Tract, edited by Blaser MJ, Ravdin JI, Greenberg HB, and Guerrant RL. New York: Raven, 1995, p. 163178. 403. NEUTRA MR, MANTIS NJ, FREY A, AND GIANNASCA PJ. The composition and function of M cell apical membranes: implications for microbial pathogenesis. Semin Immunol 11: 171181, 1999. 404. NEUTRA MR, PHILLIPS TL, MAYER EL, AND FISHKIND DJ. Transport of membrane-bound macromolecules by M cells in follicle-associated epithelium of rabbit Peyers patch. Cell Tissue Res 247: 537546, 1987. 405. NIELSON M, HUSMARK J, NILSSON B, TISELL LE, AND ERICSON LE. Primary culture of human thyrocytes in Transwell bicameral chamber: thyrotropin promotes polarization and epithelial barrier function. Eur J Endocrinol 135: 469 480, 1996. 406. NITSCH L, TRAMONTANO D, AMBESI-IMPIOMBATO FS, QUARTO N, AND BONATTI S. Morphological and functional polarity of an epithelial thyroid cell line. Eur J Cell Biol 38: 57 66, 1985. 407. NIZET V, KIM KS, STINS M, JONAS M, CHI EY, NGUYEN D, AND RUBENS CE. Invasion of brain microvascular endothelial cells by group B streptococci. Infect Immun 65: 5074 5081, 1997. 408. NOVICK P, FERRO S, AND SCHEKMAN R. Order of events in the yeast secretory pathway. Cell 25: 461 469, 1981. 409. NOVICK P, FIELD C, AND SCHEKMAN R. Identication of 23 complementation groups required for posttranslational events in the yeast secretory pathway. Cell 21: 205215, 1980. 410. NOVIKOFF PM, CAMMER M, TAO L, ODA H, STOCKERT RJ, AND WOLKOFF AW. Three-dimensional organization of rat hepatocyte cytoskeleton: relation to the asialoglycoprotein endocytosis pathway. J Cell Sci 109: 2132, 1996. 411. NUMATA S, SHIRATAKI H, HAGI S, YAMAMOTO T, AND TAKAI Y. Phosphorylation of Rabphilin-3A, a putative target protein for Rab3A, by cyclic AMP-dependent protein kinase. Biochem Biophys Res Commun 203: 19271934, 1994. 412. NURNBERG B AND AHNERT-HILGER G. Potential roles of heterotrimeric G proteins of the endomembrane system. FEBS Lett 389: 61 65, 1996. 413. NYKJAER A, DRAGUN D, WALTHER D, VORUM H, JACOBSEN C, HERZ J, MELSEN F, CHRISTENSEN EI, AND WILLNOW TE. An endocytic pathway essential for renal uptake and activation of the steroid 25-(OH) vitamin D3. Cell 96: 507515, 1999. 414. OBUNIKE JC, LUTZ EP, LI Z, PAKA L, KATOPODIS T, STRICKLAND DK, KOZARSKY KF, PILLARISETTI S, AND GOLDBERG IJ. Transcytosis of lipoprotein lipase across cultured endothelial cells requires both heparan sulfate proteoglycans and the very low density lipoprotein receptor. J Biol Chem 276: 8934 8941, 2001. 415. ODA H, STOCKERT RJ, COLLINS C, WANG H, NOVIKOFF PM, SATIR P, AND WOLKOFF AW. Interaction of the microtubule cytoskeleton with endocytic vesicles and cytoplasmic dynein in cultured rat hepatocytes. J Biol Chem 270: 1524215249, 1995. 416. OH P, MCINTOSH DP, AND SCHNITZER JE. Dynamin at the neck of caveolae mediates their budding to form transport vesicles by GTP-driven ssion from the plasma membrane of endothelium. J Cell Biol 141: 101114, 1998. 417. OHKURA K AND TERADA H. Polarity of cultured newborn rat skin basal cell layer. Biol Pharm Bull 19: 10131017, 1996. 418. OHKURA K AND TERADA H. Transcellular transport of low density Physiol Rev VOL

927

lipoprotein through cultured newborn rat skin epidermal cell monolayer. Chem Pharm Bull 39: 437 440, 1991. 419. OJAKIAN GK AND SCHWIMMER R. The polarized distribution of an apical cell surface glycoprotein is maintained by interactions with the cytoskeleton of Madin-Darby canine kidney cells. J Cell Biol 107: 23772387, 1988. 420. OJAKIAN GK AND SCHWIMMER R. Antimicrotubule drugs inhibit the polarized insertion of an intracellular glycoprotein pool into the apical membrane of Madin-Darby canine kidney (MDCK) cells. J Cell Sci 103: 677 687, 1992. 421. OKAMOTO CT, SHIA SP, BIRD C, MOSTOV KE, AND ROTH MG. The cytoplasmic domain of the polymeric immunoglobulin receptor contains two internalization signals that are distinct from its basolateral sorting signal. J Biol Chem 267: 99259932, 1992. 422. OKAMOTO CT, SONG W, BOMSEL M, AND MOSTOV KE. Rapid internalization of the polymeric immunoglobulin receptor requires phosphorylated serine 726. J Biol Chem 269: 15676 15682, 1994. 423. ORLANS E, PEPPARD J, FRY JF, HINTON RH, AND MULLOCK BM. Secretory component as the receptor for polymeric IgA on rat hepatocytes. J Exp Med 150: 15771581, 1979. 424. ORTEGA D, POL A, BIERMER M, JACKLE S, AND ENRICH C. Annexin VI denes an apical endocytic compartment in rat liver hepatocytes. J Cell Sci 111: 261269, 1998. 425. OWEN RL. Sequential uptake of horseradish peroxidase by lymphoid follicle epithelium of Peyers patches in the normal unobstructed mouse intestine: an ultrastructural study. Gastroenterology 72: 440 451, 1977. 425a.PAGEOT LP, PERRAULT N, BASORA N, FRANCOEUR C, MAGNY P, AND BEAULIEU JF. Human cell models to study small intestinal functions: recapitulation of the crypt-villus axis. Microsc Res Tech 49: 394 406, 2000. 426. PALADE GE. The ne structure of blood capillaries. J Appl Physiol 24: 1424, 1953. 427. PANARETOU C, DOMAIN J, COCKCROFT S, AND WATERFIELD MD. Characterization of p150, an adaptor protein for the human phosphatidylinositol (PtdIns) 3-kinase. J Biol Chem 272: 24772485, 1997. 428. PAPPENHEIMER JR, RENKIN EM, AND BORRERO LM. Filtration, diffusion and molecular sieving through peripheral capillary membranes. 167: 13 46, 1951. 429. PARDRIDGE WM. Blood-brain barrier biology and methodology. J Neurovirol 5: 556 569, 1999. 430. PARDRIDGE WM, EISENBERG J, AND YANG J. Human blood-brain barrier transferrin receptor. Metabolism 36: 892 895, 1987. 431. PARDRIDGE WM, KANG YS, BUCIAK JL, AND YANG J. Human insulin receptor monoclonal antibody undergoes high afnity binding to human brain capillaries in vitro and rapid transcytosis through the blood-brain barrier in vivo in the primate. Pharm Res 12: 807 816, 1995. 432. PARKKILA S, WAHEED A, BRITTON RS, BACON BR, ZHOU XY, TOMATSU S, FLEMING RE, AND SLY WS. Association of the transferrin receptor in human placenta with HFE, the protein defective in hereditary hemochromatosis. Proc Natl Acad Sci USA 94: 13198 13202, 1997. 433. PARMLEY RT, BARTON JC, AND CONRAD ME. Ultrastructural localization of transferrin, transferrin receptor, and iron-binding sites on human placental and duodenal microvilli. Br J Haematol 60: 81 89, 1985. 434. PARTON RG, DOTTI CG, BACALLAO R, KURTZ I, SIMONS K, AND PRYDZ K. pH-induced microtubule-dependent redistribution of late endosomes in neuronal and epithelial cells. J Cell Biol 113: 261274, 1991. 435. PARTON RG, JOGGERST B, AND SIMONS K. Regulated internalization of caveolae. J Cell Biol 127: 1199 1215, 1994. 436. PATTILLO RA AND GEY GO. The establishment of a cell line of human hormone-synthesizing trophoblastic cells in vitro. Cancer Res 28: 12311236, 1968. 437. PATTON JS. Mechanisms of macromolecule absorption by the lungs. Adv Drug Delivery Rev 19: 336, 1996. 438. PENG XR, YAO X, CHOW D-C, FORTE JG, AND BENNETT MK. Association of Syntaxin 3 and vesicle-associated membrane protein (VAMP) with H/K-ATPase-containing tubulovesicles in gastric parietal cells. Mol Biol Cell 8: 399 407, 1997. 439. PEPPARD JV AND RUSSELL MW. Phylogenetic Development and Comparative Physiology of IgA. San Diego, CA: Academic, 1999. www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

928

PAMELA L. TUMA AND ANN L. HUBBARD 462. RAMANUJAM KS, SEETHARAM S, DAHMS NM, AND SEETHARAM B. Effect of processing inhibitors on cobalamin (vitamin B12) transcytosis in polarized opossum kidney cells. Arch Biochem Biophys 315: 8 15, 1994. 463. RAMASAMY M, ALPERS DH, TIRUPPATHI C, AND SEETHARAM B. Cobalamin release from intrinsic factor and transfer to transcobalamin II within the rat enterocyte. Am J Physiol Gastrointest Liver Physiol 256: G791G797, 1989. 464. RAUPACH B, MECSAS J, HECZKO U, FALKOW S, AND FINLAY BB. Bacterial epithelial cell cross talk. Curr Top Microbiol Immunol 236: 137 161, 1999. 465. RAZANI B, ENGELMAN JA, WANG XB, SCHUBERT W, ZHANG XL, MARKS CB, MACALUSO F, RUSSELL RG, LI M, PESTELL RG, DI VIZIO D, HOU H JR, KNIETZ B, LAGAUD G, CHRIST GJ, EDELMANN W, AND LISANTI MP. Caveolin-1 null mice are viable, but show evidence of hyper-proliferative and vascular abnormalities. J Biol Chem 276: 3812138138, 2001. 466. REN M, XU G, ZENG J, DE LEMOS-CHIARANDINI C, ADESNIK M, AND SABATINI DD. Hydrolysis of GTP on rab11 is required for the direct delivery of transferrin from the pericentriolar recycling compartment to the cell surface but not from sorting endosomes. Proc Natl Acad Sci USA 95: 6187 6192, 1998. 467. RENKIN EM. Cellular and intercellular transport pathways in exchange vessels. Am Rev Respir Dis 146: S28 S31, 1992. 468. RENSTON RH, JONES AL, CHRISTIANSEN WD, AND HRADEK GT. Evidence for a vesicular transport mechanism in hepatocytes for biliary secretion of immunoglobulin A. Science 208: 1276 1278, 1980. 469. RICHARDSON J, BOUCHARD T, AND FERGUSON CC. Uptake and transport of exogenous proteins by respiratory epithelium. Lab Invest 35: 307314, 1976. 470. RICHARDSON JC, SCALERA V, AND SIMMONS NL. Identication of two strains of MDCK cells which resemble separate nephron tubule segments. Biochim Biophys Acta 673: 26 36, 1981. 471. RIENTO K, GALLI T, JANSSON S, EHNHOLM C, LEHTONEN E, AND OLKKONEN VM. Interaction of Munc-18 2 with syntaxin 3 controls the association of apical SNAREs in epithelial cells. J Cell Sci 111: 26812688, 1998. 472. RIENTO K, JANTTI J, JANSSON S, HIELM S, LEHTONEN E, EHNHOLM C, KERANEN S, AND OLKKONEN VM. A Sec1-related vesicle-transport protein that is expressed predominantly in epithelial cells. Eur J Biochem 239: 638 646, 1996. 473. RING A, WEISER JN, AND TUOMANEN EI. Pneumococcal trafcking across the blood-brain barrier. Molecular analysis of a novel bidirectional pathway. J Clin Invest 102: 347360, 1998. 474. RISINGER C AND BENNETT MK. Differential phosphorylation of syntaxin and synaptosome-associated protein of 25 kDa (SNAP-25) isoforms. J Neurochem 72: 614 624, 1999. 475. RIZZOLO LJ AND JOSHI HC. Apical orientation of the microtubule organizing center and associated gamma-tubulin during the polarization of the retinal pigment epithelium in vivo. Dev Biol 157: 147156, 1993. 476. ROBERTS R, SANDRA A, SIEK GC, LUCAS JJ, AND FINE RE. Studies of the mechanism of iron transport across the blood-brain barrier. Ann Neurol 32: S43S50, 1992. 477. ROBERTS WG AND PALADE GE. Increased microvascular permeability and endothelial fenestration induced by vascular endothelial growth factor. J Cell Sci 108: 2369 2379, 1995. 478. RODMAN JS AND WANDINGER-NESS A. Rab GTPases coordinate endocytosis. J Cell Sci 113: 183192, 2000. 479. RODRIGUEZ-BOULAN E AND PENDERGAST M. Polarized distribution of viral envelope proteins in the plasma membrane of infected epithelial cells. Cell 20: 4554, 1980. 480. RODRIGUEZ-BOULAN E AND SABATINI DD. Asymmetric budding of viruses in epithelial monolayers: a model system for study of epithelial polarity. Proc Natl Acad Sci USA 75: 50715075, 1978. 481. ROGERS KA AND KALNINS VI. A method for examining the endothelial cytoskeleton in situ using immunouorescence. J Histochem Cytochem 31: 13171320, 1983. 482. ROPER K, CORBEIL D, AND HUTTNER WB. Retention of prominin in microvilli reveals distinct cholesterol-based lipid micro-domains in the apical plasma membrane. Nat Cell Biol 2: 582592, 2000. 483. ROUSSET B AND MORNEX R. The thyroid hormone secretory pathwww.prv.org

440. PETERSON MD AND MOOSEKER MS. Characterization of the enterocyte-like brush border cytoskeleton of the C2BBe clones of the human intestinal cell line, Caco-2. J Cell Sci 102: 581 600, 1992. 441. PETERSON WD JR, STULBERG CS, AND SIMPSON WF. A permanent heteroploid human cell line with type B glucose-6-phosphate dehydrogenase. Proc Soc Exp Biol Med 136: 11871191, 1971. 442. PEVSNER J. The role of Sec1p-related proteins in vesicle trafcking in the nerve terminal. J Neurol Res 45: 89 95, 1996. 443. PFEFFER SR. Transport vesicle docking: SNAREs and associates. Annu Rev Cell Dev Biol 12: 441 461, 1996. 444. PILETTE C, OUADRHIRI Y, GODDING V, VAERMAN JP, AND SIBILLE Y. Lung mucosal immunity: immunoglobulin-A revisited. Eur Respir J 18: 571588, 2001. 445. PIMPLIKAR SW AND SIMONS K. Role of heterotrimeric G proteins in polarized membrane transport. J Cell Sci 17: 2732, 1993. 446. PIMPLIKAR SW AND SIMONS K. Regulation of apical transport in epithelial cells by a Gs class of heterotrimeric G proteins. Nature 362: 456 458, 1993. 447. PIMPLIKAR SW AND SIMONS K. Activators of protein kinase A stimulate apical but not basolateral transport in epithelial Madin-Darby canine kidney cells. J Biol Chem 269: 19054 19059, 1994. 448. PLATICA O, JANECZKO R, QUADROS EV, REGEC A, ROMAIN R, AND ROTHENBERG SP. The cDNA sequence and the deduced amino acid sequence of human transcobalamin II show homology with rat intrinsic factor and human transcobalamin I. J Biol Chem 266: 7860 7863, 1991. 449. POLLARD TD, DOBERSTEIN SK, AND ZOT HG. Myosin I. Annu Rev Physiol 53: 653 681, 1991. 450. POLLIOTTI BM, PANIGEL M, AND MILLER RK. Free vitamin B12 and transcobalamin II-vitamin B12 complex uptake by the visceral yolk sac of the Sprague-Dawley rat: efffect on inhibitors. Reprod Toxicol 4: 617 627, 1997. 451. PONS L, GUY M, LAMBERT D, HATIER R, AND GUEANT J. Transcytosis and coenzymatic conversion of [(57)Co]cobalamin bound to either endogenous transcobalamin II or exogenous intrinsic factor in caco-2 cells. Cell Physiol Biochem 10: 135148, 2000. 452. POUCELL-HATTON S, PERKINS PS, DEERINCK TJ, ELLISMAN MH, HARDISON WG, AND PANDOL SJ. Myosin I is associated with zymogen granule membranes in the rat pancreatic acinar cell. Gastroenterology 113: 649 658, 1997. 453. POUS C, CHABIN K, DRECHOU A, BARBOT L, PHUNG-KOSKAS T, SETTEGRANA C, BOURGUET-KONDRACKI ML, MAURICE M, CASSIO D, GUYOT M, AND DURAND G. Functional specialization of stable and dynamic microtubules in protein trafc in WIF-B cells. J Cell Biol 142: 153165, 1998. 454. PRAETOR A, ELLINGER I, AND HUNZIKER W. Intracellular trafc of the MHC class I-like IgG Fc receptor, FcRn, expressed in epithelial MDCK cells. J Cell Sci 112: 22912299, 1999. 455. PREDESCU D, HORVAT R, PREDESCU S, AND PALADE GE. Transcytosis in the continuous endothelium of the myocardial microvasculature is inhibited by N-ethylmaleimide. Proc Natl Acad Sci USA 91: 3014 3018, 1994. 456. PREDESCU D, PREDESCU S, MCQUISTAN T, AND PALADE GE. Transcytosis of alpha1-acidic glycoprotein in the continuous microvascular endothelium. Proc Natl Acad Sci USA 95: 6175 6180, 1998. 457. PREDESCU SA, PREDESCU DN, AND PALADE GE. Endothelial transcytotic machinery involves supramolecular protein-lipid complexes. Mol Biol Cell 12: 1019 1033, 2001. 458. PREKERIS R, KLUMPERMAN J, AND SCHELLER RH. A Rab11/Rip11 protein complex regulates apical membrane trafcking via recycling endosomes. Mol Cell 6: 14371448, 2000. 459. PUERTOLLANO R, MARTIN-BELMONTE F, MILLAN J, DE MARCO MC, ALBAR JP, KREMER L, AND ALONSO MA. The MAL proteolipid is necessary for normal apical transport and accurate sorting of the inuenza virus hemagglutinin in Madin-Darby canine kidney cells. J Cell Biol 145: 141151, 1999. 460. QUADROS EV, REGIC AL, KHAN KMF, QUADROS E, AND ROTHENBERG SP. Transcobalamin II synthesized in the intestinal villi facilitiates transfer of cobalamin to the portal blood. Am J Physiology Gastrointest Liver Physiol 277: G161G166, 1999. 461. RADER DJ AND DUGI KA. The endothelium and lipoproteins: insights from recent cell biology and animal studies. Semin Thromb Hemost 26: 521528, 2000. Physiol Rev VOL

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

TRANSCYTOSIS way current dogmas and alternative hypotheses. Mol Cell Endocrinol 78: C89 C93, 1991. ROUSSET M. The human colon carcinoma cell lines HT-29 and Caco-2: two in vitro models for the study of intestinal differentiation. Biochimie 68: 10351040, 1986. ROY CN AND ENNS CA. Iron homeostasis: new tales from the crypt. Blood 96: 4020 4027, 2000. RUNNEGAR MT, WEI X, AND HAMM-ALVAREZ SF. Increased protein phosphorylation of cytoplasmic dynein results in impaired motor function. Biochem J 342: 1 6, 1999. RUSSELL-JONES GJ. Use of vitamin B12 conjugates to deliver protein drugs by the oral route. Crit Rev Ther Drug Carrier Syst 15: 557586, 1998. RUSSELL-JONES GJ AND ALPERS DH. Vitamin B12 transporters. Pharm Biotechnol 12: 493520, 1999. SAKISAKA S, NG OC, AND BOYER JL. Tubulovesicular transcytotic pathway in isolated rat hepatocyte couplets in culture. Effect of colchicine and taurocholate. Gastroenterology 95: 793 804, 1988. SALO J, LEHENKARI P, MULARI M, METSIKKO K, AND VAANANEN HK. Removal of osteoclast bone resorption products by transcytosis. Science 276: 270 272, 1997. SALVI S AND HOLGATE ST. Could the airway epithelium play an important role in mucosal immunoglobulin A production? Clin Exp Allergy 29: 15971605, 1999. SANSONETTI PJ AND PHALIPON A. M cells as ports of entry for enteroinvasive pathogens: mechanisms of interaction, consequences for the disease process. Semin Immunol 11: 193203, 1999. SARNATARO D, NITSCH L, HUNZIKER W, AND ZURZOLO C. Detergent insoluble microdomains are not involved in transcytosis of polymeric Ig receptor in FRT and MDCK cells. Trafc 1: 794 802, 2000. SATO-YOSHITAKE R, YORIFUJI H, IMAGAKI M, AND HIROKAWA N. The phosphorylation of kinesin regulates its binding to synaptic vesicles. J Biol Chem 267: 23930 23936, 1992. SAVAGE CR JR AND GREEN PD. Biosynthesis of transcobalamin II by adult rat liver parenchymal cells in culture. Arch Biochem Biophys 173: 691702, 1976. SAWAMURA T, KUME N, AOYAMA T, MORIWAKI H, HOSHIKAWA H, AIBA Y, TANAKA T, MIWA S, KATSURA Y, KITA T, AND MASAKI T. An endothelial receptor for oxidized low-density lipoprotein. Nature 386: 7377, 1997. SCHAERER E, VERREY F, RACINE L, TALLICHET C, REINHARDT M, AND KRAEHENBUHL JP. Polarized transport of the polymeric immunoglobulin receptor in transfected rabbit mammary epithelial cells. J Cell Biol 110: 987998, 1990. SCHEIFFELE P, VERKADE P, FRA AM, VIRTA H, SIMONS K, AND IKONEN E. Caveolin-1 and -2 in the exocytic pathway of MDCK cells. J Cell Biol 140: 795 806, 1998. SCHELL MJ, MAURICE M, STIEGER B, AND HUBBARD AL. 5-Nucleotidase is sorted to the apical domain of hepatocytes via an indirect. J Cell Biol 119: 11731182, 1992. SCHIMMOLLER F, SIMON I, AND PFEFFER SR. Rab GTPases, directors of vesicle docking. J Biol Chem 273: 2216122164, 1998. SCHLACHETZKI F, ZHU C, AND PARDRIDGE WM. Expression of the neonatal Fc receptor (FcRn) at the blood-brain barrier. J Neurochem 81: 203206, 2002. SCHMID SL, MCNIVEN MA, AND DECAMILLI P. Dynamin and its partners: a progress report. Curr Opin Cell Biol 10: 504 512, 1998. SCHNEEBERGER EE AND HAMELIN M. Interaction of serum proteins with lung endothelial glycocalyx: its effect on endothelial permeability. Am J Physiol Heart Circ Physiol 247: H206 H217, 1984. SCHNITZER JE. gp60 is an albumin-binding glycoprotein expressed by continous endothelium involved in albumin transcytosis. Am J Physiol Heart Circ Physiol 262: H246 H254, 1992. SCHNITZER JE, ALLARD J, AND OH P. NEM inhibits transcytosis, endocytosis, and capillary permeability: implication of caveolae fusion in endothelia. Am J Physiol Heart Circ Physiol 268: H48 H55, 1995. SCHNITZER JE AND BRAVO J. High afnity binding, endocytosis, and degradation of conformationally modied albumins. J Biol Chem 268: 75627570, 1993. SCHNITZER JE, CARLEY WW, AND PALADE GE. Albumin interacts specically with a 60-kDa microvascular endothelial glycoprotein. Proc Natl Acad Sci USA 85: 6773 6777, 1988. Physiol Rev VOL

929

484.

485. 486.

487.

488. 489.

490.

491.

492.

493.

494.

495.

496.

497.

498.

499.

500. 501.

502. 503.

504.

505.

506.

507.

508. SCHNITZER JE, CARLEY WW, AND PALADE GE. Specic albumin binding to microvascular endothelium in culture. Am J Physiol Heart Circ Physiol 254: H425H437, 1988. 509. SCHNITZER JE, LIU J, AND OH P. Endothelial caveolae have the molecular transport machinery for vesicle budding, docking, and fusion including VAMP. NSF, SNAP, annexins, and GTPases. J Biol Chem 270: 14399 14404, 1995. 511. SCHNITZER JE, OH P, PINNEY E, AND ALLARD J. Filipin-sensitive caveolae-mediated transport in endothelium: reduced transcytosis, scavenger endocytosis, and capillary permeability of select macromolecules. J Cell Biol 127: 12171232, 1994. 512. SCHUBERT W, FRANK PG, RAZANI B, PARK DS, CHOW CW, AND LISANTI MP. Caveolae-decient endothelial cells show defects in the uptake and transport of albumin in vivo. J Biol Chem 276: 48619 48622, 2001. 513. SCHULTE R, KERNEIS S, KLINKE S, BARTELS H, PREGER S, KRAEHENBUHL JP, PRINGAULT E, AND AUTENRIETH IB. Translocation of Yersinia entrocolitica across reconstituted intestinal epithelial monolayers is triggered by Yersinia invasin binding to beta1 integrins apically expressed on M-like cells. Cell Microbiol 2: 173185, 2000. 514. SEETHARAM B. Receptor-mediated endocytosis of cobalamin (vitamin B12). Annu Rev Nutr 19: 173195, 1999. 515. SEETHARAM B, ALPERS DH, AND ALLEN RH. Isolation and characterization of the ileal receptor for intrinsic factor-cobalamin. J Biol Chem 256: 37853790, 1981. 516. SEETHARAM B, CHRISTENSEN EI, MOESTRUO SK, HAMMOND TG, AND VERROUST P. Identication of rat yolk sac target protein of teratogenic antibodies, gp280, as intrinsic factor-cobalamin receptor. J Clin Invest 99: 23172322, 1997. 517. SEETHARAM B, LEVINE JS, RAMASAMY M, AND ALPERS DH. Purication, properties, and immunochemical localization of a receptor for intrinsic factor-cobalamin complex in the rat kidney. J Biol Chem 263: 4443 4449, 1988. 518. SEETHARAM B AND LI N. Transcobalamin II and its cell surface receptor. Vitam Horm 59: 337366, 2000. 519. SEETHARAM B, PRESTI M, FRANK B, TIRUPPATHI C, AND ALPERS DH. Intestinal uptake and release of cobalamin complexed with rat intrinsic factor. Am J Physiol Gastrointest Liver Physiol 248: G326 G331, 1985. 520. SHAH D AND SHEN WC. Transcellular delivery of an insulin-transferrin conjugate in enterocyte-like Caco-2 cells. J Pharm Sci 85: 1306 1311, 1996. 521. SHANKS MS, CASSIO D, LECOQ O, AND HUBBARD AH. An improved rat hepatoma hybrid cell line. Generation and comparison with its hepatoma relatives and hepatocytes in vivo. J Cell Sci 107: 813 825, 1994. 522. SHEN BQ, FINKBEINER WE, WINE JJ, MRSNY RJ, AND WIDDICOMBE JH. Calu-3: a human airway epithelial cell line that shows cAMP-dependent Cl secretion. Am J Physiol Lung Cell Mol Physiol 266: L493L501, 1994. 523. SHENNAN DB AND PEAKER M. Transport of milk constituents by the mammary gland. Physiol Rev 80: 925951, 2000. 524. SHEPHERD PR, REAVES BJ, AND DAVIDSON HW. Phosphoinositide 3-kinases and membrane trafc. Trends Cell Biol 6: 9297, 1996. 525. SHETH S AND BRITTENHAM GM. Genetic disorders affecting proteins of iron metabolism: clinical implications. Annu Rev Med 51: 443 464, 2000. 526. SHIMIZU M, TSUNOGAI M, AND ARAI S. Transepithelial transport of oligopeptides in the human intestinal cell, Caco-2. Peptides 18: 681 687, 1997. 527. SHUANG R, ZHANG L, FLETCHER A, GROBLEWSKI GE, PEVSNER J, AND STUENKEL EL. Regulation of Munc-18/syntaxin 1A interaction by cyclin-dependent kinase 5 in nerve endings. J Biol Chem 273: 4957 4966, 1998. 528. SHURETY W, BRIGHT NA, AND LUZIO JP. The effects of cytochalasin D and phorbol myristate acetate on the apical endocytosis of ricin in polarised Caco-2 cells. J Cell Sci 109: 29272935, 1996. 529. SIDDHANTA U, MCILROY J, SHAH A, ZHANG Y, AND BACKER JM. Distinct roles for p110a and hVPS34 phosphatidylinositol 3-kinases in vesicular trafcking, regulation of the actin cytoskeleton, and mitogenesis. J Cell Biol 143: 16471659, 1998. 530. SIFLINGER-BIRNBOIM CJA, DEL VECCHIO PJ, LUM H, AND MALIK AB. www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

930

PAMELA L. TUMA AND ANN L. HUBBARD Selectivity of the endothelial monolayer: effects of increased permeability. Microvasc Res 36: 216 227, 1988. SIFLINGER-BIRNBOIM ADVP, COOPER JA, BLUMENSTOCK FA, SHEPARD JM, AND MALIK AB. Molecular sieving characteristics of the cultured endothelial monolayer. J Cell Physiol 132: 111117, 1987. SILVER DL, WANG N, XIAO X, AND TALL AR. High density lipoprotein (HDL) particle uptake mediated by scavenger receptor class B type 1 results in selective sorting of HDL cholesterol from protein and polarized cholesterol secretion. J Biol Chem 276: 2528725293, 2001. SILVERMAN MA, KAECH S, JAREB M, BURACK MA, VOGT L, SONDEREGGER P, AND BANKER G. Sorting and directed transport of membrane proteins during development of hippocampal neurons in culture. Proc Natl Acad Sci USA 98: 70517057, 2001. SIMIONESCU M. Receptor-mediated transcytosis of plasma molecules by vascular endothelium. In: Endothelia Cell Biology in Health and Disease, edited by Simionescu SA. New York: Plenum, 1988, p. 69 104. SIMIONESCU M AND SIMIONESCU N. Ultrastructure of the microvascular wall: functional correlations. In: Handbook of Physiology. The Cardiovascular System. Microcirculation. Bethesda, MD: Am Physiol Soc, 1984, sect. 2, vol. IV, pt. 1, chapt. 3, p. 41101. SIMIONESCU M AND SIMIONESCU N. Endothelial transport of macromolecules: transcytosis and endocytosis. Cell Biol Rev 25: 1 80, 1991. SIMIONESCU M, SIMIONESCU N, AND PALADE GE. Differentiated microdomains on the luminal surface of capillary endothelium: distribution of lectin receptors. J Cell Biol 94: 406 413, 1982. SIMIONESCU N. The microvascular endothelium: segmental differentiations; transcytosis, selective distribution of anionic sites. In: Advances in Inammation Research, edited by Weissman G, Samuelson B, and Paoletti R. New York: Raven, 1979, p. 6170. SIMIONESCU N. Cellular aspects of transcapillary exhcange. Physiol Rev 63: 1536 1579, 1983. SIMIONESCU N, SIMIONESCU M, AND PALADE GE. Differentiated microdomains on the luminal surface of the capillary endothelium. I. Preferential distribution of anionic sites. J Cell Biol 90: 605 613, 1981. SIMISTER NE, JACOBOWITZ ISRAEL E, AHOUSE JC, AND STORY CM. New functions of the MHC class I-related Fc receptor, FcRn. Biochem Soc Trans 25: 481 486, 1997. SIMISTER NE AND MOSTOV KE. An Fc receptor structurally related to MHC class I antigens. Nature 337: 184 187, 1989. SIMISTER NE, STORY CM, CEN H-L, AND HUNT JS. An IgG-transporting Fc receptor expressed in the syncytiotrophoblast of human placenta. Eur J Immunol 26: 15271531, 1996. SIMONS K, DUPREE P, FIEDLER K, HUBER LA, KOBAYASHI T, KURZCHALIA T, OLKKONEN V, PIMPLIKAR S, PARTON R, AND DOTTI C. Biogenesis of cell-surface polarity in epithelial cells and neurons. Cold Spring Harb Symp Quant Biol 57: 611 619, 1992. SKARLATOS S, YOSHIKAWA T, AND PARDRIDGE WM. Transport of [125I]transferrin through the rat blood-brain barrier. Brain Res 683: 164 171, 1995. SMART EJ, GRAF GA, MCNIVEN MA, SESSA WC, ENGELMAN JA, SCHERER PE, OKAMOTO T, AND LISANTI MP. Caveolins, liquid-ordered domains, and signal transduction. Mol Cell Biol 19: 7289 7304, 1999. SOLLNER T, BENNETT MK, WHITEHEART SW, SCHELLER RH, AND ROTHMAN JE. A protein assembly-disassembly pathway in vitro that may correspond to sequential steps of synaptic vesicle docking, activation and fusion. Cell 75: 409 418, 1993. SONG W, BOMSEL M, CASANOVA J, VAERMAN JP, AND MOSTOV K. Stimulation of transcytosis of the polymeric immunoglobulin receptor by dimeric IgA. Proc Natl Acad Sci USA 91: 163166, 1994. SOORANNA SR AND CONTRACTOR SF. Vectorial transcytosis of immunoglobulin g by human term trophoblast cells in culture. Exp Cell Res 192: 41 45, 1991. SOORANNA SR, MOSS J, AND CONTRACTOR SF. Comparison of the intracellular pathways of immunoglobulin-G and low density lipoproteinin cultured human term trophoblast cells. Cell Tissue Res 273: 619 625, 1993. SOTGIA F, RAZANI B, BONUCCELLI G, SCHUBERT W, BATTISTA M, LEE H, CAPOZZA F, SCHUBERT AL, MINETTI C, BUCKLEY JT, AND LISANTI MP. Physiol Rev VOL Intracellular retention of glycosylphosphatidyl inositol-linked proteins in caveolin-decient cells. Mol Cell Biol 22: 39053926, 2002. SPECTOR R AND JOHANSON CE. The mammalian choroid plexis. Sci Am 68 74, 1989. SPIEKERMANN GM, FINN PW, WARD ES, DUMONT J, DICKINSON BL, BLUMBERG RS, AND LENCER WI. Receptor-mediated immunoglobulin G transport across mucosal barriers in adult life: functional expression of FcRn in the mammalian lung. J Exp Med 196: 303310, 2002. STAN RV. Structure and function of endothelial caveolae. Microsc Res Tech 57: 350 364, 2002. STEEGMAIER M, LEE KC, PREKERIS R, AND SCHELLER RH. SNARE protein trafcking in polarized MDCK cells. Trafc 1: 553560, 2000. STEELE-MORTIMER O, GRUENBERG J, AND CLAGUE MJ. Phosphorylation of GDI and membrane cycling of rab proteins. FEBS Lett 329: 313318, 1993. STEFANER I, PRAETOR A, AND HUNZIKER W. Nonvectorial surface transport, endocytosis via a Di-leucine-based motif, and bidirectional transcytosis of chimera encoding the cytosolic tail of rat FcRn expressed in Madin-Darby canine kidney cells. J Biol Chem 274: 8998 9005, 1999. STEFANER I, STEFANESCU A, HUNZIKER W, AND FUCHS R. Expression of placental alkaline phosphatase does not correlate with IgG binding, internalization and transcytosis. Biochem J 327: 585592, 1997. STEINBRECHER UP. Receptors for oxidized low density lipoprotein. Biochim Biophys Acta 1436: 279 298, 1999. STENMARK H AND AASLAND R. FYVE-nger proteins effectors of an inositol lipid. J Cell Sci 112: 4175 4183, 1999. STINS MF, PRASADARAO NV, ZHOU J, ARDITI M, AND KIM KS. Bovine brain microvascular endothelial cells transfected with SV40-large T antigen: development of an immortalized cell line to study pathophysiology of CNS disease. In Vitro Cell Dev Biol Anim 33: 243 247, 1997. STOW JL. Regulation of vesicular transport by GTP-binding proteins. Curr Opin Nephrol Hypertens 4: 421 425, 1995. SUBRAMANIAM VN, LOH E, HORSTMANN H, HABERMANN A, XU Y, COE J, GRIFFITHS G, AND HONG W. Preferential association of syntaxin 8 with the early endosome. J Cell Sci 113: 9971008, 2000. SWAIRJO MA AND SEATON BA. Annexin structure and membrane interactions: a molecular perspective. Annu Rev Biophys Biomol Struct 23: 193213, 1994. SZTUL E, COLOMBO M, STAHL P, AND SAMANTA R. Control of protein trafc between distinct plasma membrane domains. J Biol Chem 268: 1876 1885, 1993. SZTUL ES, HOWELL KE, AND PALADE GE. Biogenesis of the polymeric IgA receptor in rat hepatocytes. I. Kinetic studies of its intracellular forms. J Cell Biol 100: 1248 1454, 1985. TAGOE CE, BOUSTEAD CM, HIGGINS SJ, AND WALKER JH. Characterization and immunolocalization of rat liver annexin VI. Biochim Biophys Acta 1192: 272280, 1994. TAI AW, CHUANG JZ, AND SUNG CH. Cytoplasmic dynein regulation by subunit heterogeneity and its role in apical transport. J Cell Biol 153: 1499 1509, 2001. TAKEI K, MCPHERSON PS, SCHMID SL, AND DECAMILLI P. Tubular membrane invaginations coated by dynamin rings are induced by GTPgamma-S in nerve terminals. Nature 374: 186 190, 1995. TANG SS, JUNG F, DIAMANT D, BROWN D, BACHINSKY D, HELLMAN P, AND INGELFINGER JR. Temperature-sensitive SV40 immortalized rat proximal tubule cell line has functional renin-angiotensin system. Am J Physiol Renal Fluid Electrolyte Physiol 268: F435F446, 1995. TAVASSOLI M, KISHIMOTO T, AND KATAOKA M. Liver endothelium mediates the hepatocytes uptake of ceruloplasmin. J Cell Biol 102: 1298 1303, 1986. TAYLOR A AND GRANGER DN. Exchange of macromolecules across the microcirculation. In: Handbook of Physiology. The Cardiovascular System. Microcirculation. Bethesda, MD: Am Physiol Soc, 1984, sect. 2, vol. IV, pt. 1, chapt. 11, p. 467520. TEITELBAUM SL. Bone resorption by osteoclasts. Science 289: 1504 1508, 2000. TERBUSH DR AND NOVICK P. Sec6, Sec8, and Sec15 are components of a multisubunit complex which localizes to small bud tips in Saccharomyces cerevisiae. J Cell Biol 130: 299 312, 1995. TING AE, HAZUKA CD, HSU SC, KIRK MD, BEAN AJ, AND SCHELLER RH. www.prv.org

531.

552. 553.

532.

553. 554.

533.

555.

534.

556.

535.

557.

Downloaded from physrev.physiology.org on November 4, 2009

536.

558. 559. 560.

537.

538.

539. 540.

561. 562.

541.

563.

542. 543.

564.

565.

544.

566.

545.

567.

546.

568.

569.

547.

570.

548.

571.

549.

550.

572. 573.

551.

574.

83 JULY 2003

TRANSCYTOSIS rSec6 and rSec8, mammalian homologs of yeast proteins essential for secretion. Proc Natl Acad Sci USA 92: 96139617, 1995. 575. TIRUPPATHI C, FINNEGAN A, AND MALIK AB. Isolation and characterization of a cell surface albumin-binding protein from vascular endothelial cells. Proc Natl Acad USA 93: 250 254, 1996. 576. TIRUPPATHI C, SONG W, BERGENFELDT SASS MP, AND MALIK AB. Gp60 activation mediates albumin transcytosis in endothelial cells by tyrosine kinase-dependent pathway. J Biol Chem 272: 25968 25975, 1997. 577. TOMASI TB JR, TAN EM, SOLOMON A, AND PRENDERGAST RA. Characteristics of an immune system common to certain external secretions. J Exp Med 121: 101124, 1965. 578. TOMASI TBJ AND ZIGELBAUM S. The selective occurrence of g1A Globulins in certain body uids. J Clin Invest 42: 15521560, 1963. 579. TRIER JS. Structure and function of intestinal M cells. Gastroenterol Clin North Am 20: 531547, 1991. 580. TSUKITA S AND FURUSE M. The structure and function of claudins, cell adhesion molecules at tight junctions. Ann NY Acad Sci 915: 129 135, 2000. 581. TUCKER SP, MELSEN LR, AND COMPANS RW. Migration of polarized epithelial cells through permeable membrane substrates of dened pore size. Eur J Cell Biol 58: 280 290, 1992. 582. TUMA PL, FINNEGAN CM, YI JH, AND HUBBARD AL. Evidence for apical endocytosis in polarized hepatic cells: phosphoinositide 3-kinase inhibitors lead to the lysosomal accumulation of resident apical plasma membrane proteins. J Cell Biol 145: 1089 1102, 1999. 583. TUMA PL, NYASAE LK, BACKER JM, AND HUBBARD AL. Vps34p differentially regulates endocytosis from the apical and basolateral domains in polarized hepatic cells. J Cell Biol 154: 113, 2001. 584. UI M, OKADA T, HAZEKI K, AND HAZEKI O. Wortmannin as a unique probe for an intracellular signalling protein, phosphoinositide 3-kinase. Trends Biochem Sci 20: 303307, 1995. 585. VAANANEN HK, ZHAO H, MULARI M, AND HALLEEN JM. The cell biology of osteoclast function. J Cell Sci 113: 377381, 2000. 586. VAN DER BLIEK AM. Is dynamin a regular motor or a master regulator? Trends Cell Biol 9: 253254, 1999. 587. VAN DER ENDE A, DU MAINE A, SCHWARTZ AL, AND STROUS GJ. Modulation of transferrin-receptor activity and recycling after induced differentiation of BeWo choriocarcinoma cells. Biochem J 270: 451 457, 1990. 588. VAN DER ENDE A, DU MAINE A, SCHWARTZ A, AND STROUS G. Iron metabolism in BeWo chorion cells. J Biol Chem 262: 8910 1987, 1987. 589. VAN DEURS B. Horseradish peroxidase uptake into the rat choroid plexus epithelium, with special reference to the lysosomal system. J Ultrastruct Res 62: 155167, 1978. 590. VAN DEURS B. Structural aspects of brain barriers, with special reference to the permeability of the crebral endothelium and choroidal epithelium. Int Rev Cytol 65: 117191, 1980. 591. VAN GELDER W, HUIJSKES-HEINS MI, CLETON-SOETEMAN MI, VAN DIJK JP, AND VAN EIJK HG. Iron uptake in blood-brain barrier endothelial cells cultured in iron-depleted and iron-enriched media. J Neurochem 71: 1134 1140, 1998. 591a.VANHAESEBROECK B, LEEVERS SJ, PANAYOTOU G, AND WATERFIELD MD. Phosphoinositide 3-kinases: a conserved family of signal transducers. Trends Biochem Sci 22: 267272, 1997. 592. VAN HOUTEN M AND POSNER BI. Insulin binds to brain blood vessels in vivo. Nature 282: 623 625, 1979. 593. VAN IJZENDOORN SC AND HOEKSTRA D. (Glyco)sphingolipids are sorted in sub-apical compartments in HepG2 cells: a role for nonGolgi-related intracellular sites in the polarized distribution of (glyco)sphingolipids. J Cell Biol 142: 683 696, 1998. 594. VAN IJZENDOORN SC AND HOEKSTRA D. Polarized sphingolipid transport from the subapical compartment: evidence for distinct sphingolipid domains. Mol Biol Cell 10: 3449 3461, 1999. 595. VAN IJZENDOORN SC, TUVIM MJ, WEIMBS T, DICKEY BF, AND MOSTOV KE. Direct interaction between Rab3b and the polymeric immunoglobulin receptor controls ligand-stimulated transcytosis in epithelial cells. Dev Cell 2: 219 228, 2002. 596. VAN IJZENDOORN SC, ZEGERS MM, KOK JW, AND HOEKSTRA D. Segregation of glucosylceramide and sphingomyelin occurs in the apical to basolateral transcytotic route in HepG2 cells. J Cell Biol 137: 347357, 1997. Physiol Rev VOL

931

597. VAN MEER G AND SIMONS K. Lipid polarity and sorting in epithelial cells. J Cell Biochem 36: 5158, 1988. 598. VAN ZEIJL MJAH AND MATLIN KS. Microtubule perturbation inhibits intracellular transport of an apical membrane glycoprotein in a substrate-dependent manner in polarized Madin-Darby canine kidney epithelial cells. Cell Regul 1: 921936, 1990. 600. VASILE E, SIMIONESCU M, AND SIMIONESCU N. Visualization of the binding, endocytosis, and transcytosis of low-density lipoprotein in the arterial endothelium in situ. J Cell Biol 96: 16771689, 1983. 601. VILLARS F, CONRAD V, ROUAIS F, LEFEBVRE F, AMEDEE J, AND BORDENAVE L. Ability of various inserts to promote endothelium cell culture for the establishment of coculture models. Cell Biol Toxicol 12: 207214, 1996. 602. VOGEL SM, MINSHALL RD, PILIPOVIC M, TIRUPPATHI C, AND MALIK AB. Albumin uptake and transcytosis in endothelial cells in vivo induced by albumin-binding protein. Am J Physiol Lung Cell Mol Physiol 281: L1512L1522, 2001. 603. VOGEL U, SANDVIG K, AND VAN DEURS B. Expression of caveolin-1 and polarized formation of invaginated caveolae in Caco-2 and MDCK II cells. J Cell Sci 111: 825 832, 1998. 604. VOLINIA S, DHAND R, VANHAESEBROECK B, MACDOUGALL LK, STEIN R, ZVELEBIL MJ, DOMIN J, PANARETOU C, AND WATERFIELD MD. A human phosphatidylinositol 3-kinase complex related to yeast Vps34pVps15p protein sorting system. EMBO J 14: 3339 3348, 1995. 605. VON BONSDORFF CH, FULLER SD, AND SIMONS K. Apical and basolateral endocytosis in Madin-Darby canine kidney (MDCK) cells grown on nitrocellulose lters. EMBO J 4: 27812792, 1985. 606. VOYTA JC, VIA DP, BUTTERFIELD CE, AND ZETTER BR. Identication and isolation of endothelial cells based on their increased uptake of acetylated-low density lipoprotein. J Cell Biol 99: 2034 2040, 1984. 607. VULPE CD, KUO YM, MURPHY TL, COWLEY L, ASKWITH C, LIBINA N, GITSCHIER J, AND ANDERSON GJ. Hephaestin, a ceruloplasmin homologue implicated in intestinal iron transport, is defective in the sla mouse. Nat Genet 21: 195199, 1999. 608. WAGNER RC AND CASLEY-SMITH JR. Endothelial vesicles. Microvasc Res 21: 267298, 1981. 609. WANG E, BROWN PS, AROETI B, CHAPIN S, MOSTOV K, AND DUNN KW. Apical and basolateral endocytic pathways of MDCK cells meet in acidic common endosomes distinct from a nearly-neutral apical recycling endosome. Trafc 1: 480 493, 2000. 610. WANG X, KUMAR R, NAVARRE J, CASANOVA JE, AND GOLDENRING JR. Regulation of vesicle trafcking in MDCK cells by Rab11a and Rab25. J Biol Chem 275: 29138 29146, 2000. 611. WEBER E, BERTA G, TOUSSON A, ST. JOHN P, GREEN MW, GOPALOKRISHNAN U, JILLING T, SORSCHER EJ, ELTON TS, ABRAHAMSON DR, AND KIRK KL. Expression and polarized targeting of a Rab3 isoform in epithelial cells. J Cell Biol 125: 583594, 1994. 612. WEIMBS T, LOW SH, CHAPIN SJ, AND MOSTOV KE. Apical targeting in polarized epithelial cells: theres more aoat than rafts. Trends Cell Biol 7: 393399, 1997. 613. WEIMBS T, LOW SH, CHAPIN SJ, MOSTOV KE, BUCHER P, AND HOFMANN K. A conserved domain is present in different families of vesicular fusion proteins: a new superfamily. Proc Natl Acad Sci USA 94: 3046 3051, 1997. 614. WEINBAUM SACE. Modeling the structural pathways for transcapillary exchange. Soc Exp Biol: 323345, 1995. 615. WEINMAN JS, FEINBERG JM, RAINTEAU DP, GASPERS BD, AND WEINMAN SJ. Annexins in rat enterocyte and hepatocyte: an immunogold electron-microscope study. Cell Tissue Rev 278: 389 397, 1994. 616. WELTZIN R, LUCIA-JANDRIS P, MICHETTI P, FIELDS BN, KRAEHENBUHL JP, AND NEUTRA MR. Binding and transepithelial transport of immunoglobulins by intestinal M cells: demonstration using monoclonal IgA antibodies against enteric viral proteins. J Cell Biol 108: 1673 1685, 1989. 617. WHITEHEART SW AND KUBALEK EW. SNAPs and NSF: general members of the fusion apparatus. Trends Cell Biol 5: 64 68, 1995. 618. WICE BM AND GORDON JI. A strategy for isolation of cDNAs encoding proteins affecting human intestinal epithelial cell growth and differentiation: characterization of a novel gut-specic N-myristoylated annexin. J Cell Biol 116: 405 422, 1992. 619. WILLNOW TE. The low-density lipoprotein receptor gene family: multiple roles in lipid metabolism. J Mol Med 77: 306 315, 1999. 620. WILTON JC, MATTHEWS GM, BURGOYNE RD, MILLS CO, CHIPMAN JK, www.prv.org

Downloaded from physrev.physiology.org on November 4, 2009

83 JULY 2003

932
AND

PAMELA L. TUMA AND ANN L. HUBBARD COLEMAN R. Fluorescent choleretic and cholestatic bile salts take different paths across the hepatocyte: transcytosis of glycolithocholate leads to an extensive redistribution of annexin II. J Cell Biol 127: 401 410, 1994. WOLF JL, RUBIN DH, FINBERG R, KAUFFMAN RS, SHARPE AH, TRIER JS, AND FIELDS BN. Intestinal M cells: a pathway for entry of reovirus into the host. Science 212: 471 472, 1981. WONG SH, ZHANG T, XU Y, SUBRAMANIAM VN, GRIFFITHS G, AND HONG W. Endobrevin, a novel synaptobrevin/VAMP-like protein preferentially associated with the early endosome. Mol Biol Cell 9: 1549 1563, 1998. WOOD RJ AND HAN O. Recently identied molecular aspects of intestinal iron absorption. J Nutr 128: 18411844, 1998. WOOD WG, SCHROEDER F, AVDULOV NA, CHOCHINA SV, AND IGBAVBOA U. Recent advances in brain cholesterol dynamics: transport, domains, and Alzheimers disease. Lipids 34: 225234, 1999. WURMSER AE, GARY JD, AND EMR SD. Phosphoinositide 3-kinases and their FYVE domain-containing effectors as regulators of vacuolar/lysosomal membrane trafcking pathway. J Biol Chem 274: 9129 9132, 1999. WUSTNER D, HERRMANN A, HAO M, AND MAXFIELD FR. Rapid nonvesicular transport of sterol between the plasma membrane domains of polarized hepatic cells. J Biol Chem 277: 3032530336, 2002. WUSTNER D, MUKHERJEE S, MAXFIELD FR, MULLER P, AND HERRMANN A. Vesicular and nonvesicular transport of phosphatidylcholine in polarized HepG2 cells. Trafc 2: 277296, 2001. XIA CQ AND SHEN WC. Tyrphostin-8 enhances transferrin receptormediated transcytosis in Caco-2 cells and increases hypoglycemic effect of orally administered insulin-transferrin conjugate in diabetic rats. Pharm Res 18: 191195, 2001. YAMAUCHI K, TOZUKA M, HIDAKA H, HIDAKA E, KONDO Y, AND KATSUYAMA T. Characterization of apolipoprotein E-containing lipoproteins in cerebrospinal uid: effect of phenotype on the distribution of apolipoprotein E. Clin Chem 45: 14311438, 1999. YU LC AND PERDUE MH. Immunologically mediated transport of ions and macromolecules. Ann NY Acad Sci 915: 247259, 2000. ZACCHI P, STENMARK H, PARTON RG, ORIOLI D, LIM F, GINER A, MELLMAN I, ZERIAL M, AND MURPHY C. Rab17 regulates membrane trafcking through apical recycling endosomes in polarized epithelial cells. J Cell Biol 140: 1039 1053, 1998. ZAHRAOUI A, JOBERTY G, ARPIN M, FONTAINE JJ, HELLIO R, TAVITIAN A, AND LOUVARD D. A small rab GTPase is distributed in cytoplasmic vesicles in non polarized cells but colocalizes with the tight junction marker ZO-1 in polarized epithelial cells. J Cell Biol 124: 101115, 1994. ZHANG JR, MOSTOV KE, LAMM ME, NANNO M, SHIMIDA S, OHWAKI M, AND TUOMANEN E. The polymeric immunoglobulin receptor translocates pneumococci across human nasopharyngeal epithelial cells. Cell 102: 827 837, 2000. ZHANG Y AND PARDRIDGE WM. Mediated efux of IgG molecules from brain to blood across the blood-brain barrier. J Neuroimmunol 114: 168 172, 2001. ZHANG Y AND PARDRIDGE WM. Rapid transferrin efux from brain to blood across the blood-brain barrier. J Neurochem 76: 15971600, 2001. ZURZOLO C, LE BIVIC A, QUARONI A, NITSCH L, AND RODRIGUEZ-BOULAN E. Modulation of transcytotic and direct targeting pathways in a polarized thyroid cell line. EMBO J 11: 23372344, 1992. ZWEIBAUM A, PINTO M, CHEVALIER G, DUSSAULX E, TRIADOU N, LACROIX B, HAFFEN K, BRUN J, AND ROUSSET M. Enterocytic differentiation of a subpopulation of the human colon tumor cell line HT-29 selected for growth in sugar-free medium and its inhibition by glucose. J Cell Physiol 122: 2129, 1985.

630. 631.

621.

622.

632.

623. 624.

633.

625.

634.

626.

635.

Downloaded from physrev.physiology.org on November 4, 2009

627.

636.

628.

637.

629.

Physiol Rev VOL

83 JULY 2003

www.prv.org

You might also like