You are on page 1of 32

HUMAN MUTATION 28(2), 99^130, 2007

REVIEW ARTICLE

Understanding the Recent Evolution


of the Human Genome: Insights from
HumanChimpanzee Genome Comparisons
Hildegard Kehrer-Sawatzki1 and David N. Cooper2
1
Department of Human Genetics, University of Ulm, Ulm, Germany; 2Institute of Medical Genetics, Cardiff University, Heath Park,
Cardiff, United Kingdom

Communicated by Peter Oefner


The sequencing of the chimpanzee genome and the comparison with its human counterpart have begun to
reveal the spectrum of genetic changes that has accompanied human evolution. In addition to gross karyotypic
rearrangements such as the fusion that formed human chromosome 2 and the human-specific pericentric
inversions of chromosomes 1 and 18, there is considerable submicroscopic structural variation involving
deletions, duplications, and inversions. Lineage-specific segmental duplications, detected by array comparative
genomic hybridization and direct sequence comparison, have made a very significant contribution to this
structural divergence, which is at least three-fold greater than that due to nucleotide substitutions. Since
structural genomic changes may have given rise to irreversible functional differences between the diverging
species, their detailed analysis could help to identify the biological processes that have accompanied speciation.
To this end, interspecies comparisons have revealed numerous human-specific gains and losses of genes as well
as changes in gene expression. The very considerable structural diversity (polymorphism) evident within both
lineages has, however, hampered the analysis of the structural divergence between the human and chimpanzee
genomes. The concomitant evaluation of genetic divergence and diversity at the nucleotide level has
nevertheless served to identify many genes that have evolved under positive selection and may thus have
been involved in the development of human lineage-specific traits. Genes that display signs of weak negative
selection have also been identified and could represent candidate loci for complex genomic disorders. Here,
we review recent progress in comparing the human and chimpanzee genomes and discuss how the differences
detected have improved our understanding of the evolution of the human genome. Hum Mutat 28(2), 99130,
2007. r 2006 Wiley-Liss, Inc.

KEY WORDS: human genome evolution; chimpanzee vs. human genome comparisons; sequence and expression divergence

INTRODUCTION references therein]. That the speciation process of humans and


chimpanzees may have been rather complex, has been suggested
Irrespective of our anthropocentric viewpoint, hominid evolu-
by DNA sequence divergence determinations using a large
tion and human speciation are among the most fascinating topics
genomic dataset (comprising about 87 Mb) in five primate
in evolutionary biology. As opined by McConkey and Goodman
species including human and chimpanzee [Patterson et al.,
[1997]: Comparative analysis of human and ape genomes is far
2006]. It was inferred that the process of human-chimpanzee
more than an excursion into natural history at the molecular level.
speciation began o6.3 Mya. However, after the initial divergence
Until we have a detailed understanding of the genetic differences
of the two lineages, further interlineage hybridizations appear to
between ourselves and our closest evolutionary relatives, we
have occurred prior to the permanent separation of both species
cannot really know what we are (p. 351).
[Osada and Wu, 2005; Patterson et al., 2006].
The phenotypic divergence evident between humans and
chimpanzees is assumed to have been strongly influenced by
lineage- and species-specific genomic changes that have given rise
to differences in gene expression as well as gains, losses, and The Supplementary Material referred to in this article can be
changes in protein function [King and Wilson, 1975; reviewed in accessed at http://www.interscience.wiley.com/jpages/1059 -7794/
Carroll, 2003; Olson and Varki, 2003; Enard and Paabo, 2004; suppmat.
Carroll, 2005; Goodman et al., 2005; Varki and Altheide, 2005]. Received 4 May 2006; accepted revised manuscript 10 August
2006.
Comparative analyses of the genomes and transcriptomes of Correspondence to: Dr. Hildegard Kehrer-Sawatzki, Department
humans and chimpanzees are therefore proving extremely useful in of Human Genetics, University of Ulm, Albert-Einstein-Allee 11,
our attempt to build a picture of the events that have shaped our 89081 Ulm, Germany. E-mail: hildegard.kehrer-sawatzki@uni-ulm.de
respective genomes after their evolutionary divergence, estimated DOI 10.1002/humu.20420
to have occurred between 5 and 7 million years ago (Mya) [Leakey Published online 5 October 2006 in Wiley InterScience (www.
et al., 1998; Glazko and Nei, 2003; Kumar et al., 2005; and interscience.wiley.com).

r 2006 WILEY-LISS, INC.


100 HUMAN MUTATION 28(2), 99^130, 2007

The availability of the initial sequence of the chimpanzee intraspecies polymorphism, which has been estimated to account
genome has allowed comparison with its human counterpart, for some 11 to 22% of the observed divergence between humans
thereby revealing differences and similarities as well as common and chimpanzees [Chen and Li, 2001; Ebersberger et al., 2002;
patterns of evolution (The Chimpanzee Sequencing and Analysis Elango et al., 2006]. If this polymorphic variation is subtracted
Consortium [CSAC]) [Mikkelsen et al., 2005]. The chimpanzee from the overall divergence, the rate of fixed (nonpolymorphic)
genome sequence was derived from a single individual of the sequence divergence between aligned human and chimpanzee
Pan troglodytes verus subspecies and represents a draft version DNA sequence becomes even lower (1%).
covering 94% of the genome sequence; however, several The extent of DNA sequence variation in chimpanzees is some
segments are not yet anchored within large contigs. This three-fold higher than that in humans, as determined by the
notwithstanding, the accuracy of the nucleotide sequence is high: analysis of a 10-kb region on the X chromosome [Kaessmann
thus, 98% of the assembled sequence has an estimated error rate of et al., 2001], and 1.5-fold higher according to estimates of
less than 104. This draft sequence therefore provides the means variation across more extensive regions of human, bonobo,
to analyze both structural and functional aspects of human/simian and chimpanzee (eastern, western, and central African) genomes
genome evolution and the underlying mutational processes [Yu et al., 2003]. The lower sequence diversity in humans is
that have molded it. The completion of the chimpanzee genome probably due to a combination of the historically lower effective
sequence and the availability of high quality sequence data population size and the recent demographic expansion in the
promises not only to help to refine these analyses by excluding human lineage.
many sequencing errors [Taudien et al., 2006] but should also Humans have a longer generation time than hominoids
potentiate the comparison of highly complex genomic regions such (gibbons, orangutan, gorilla, chimpanzees). According to the
as the pericentromeric and subtelomeric regions. generation-time effect hypothesis [Goodman, 1962; Li et al.,
The main focus of most human/chimpanzee genome compar- 1996], most germline mutations originate from errors in DNA
isons has been the search for lineage-specific differences. Other replication. The longer the generation time, the lower the number
primate species have frequently been included in these analyses as of germ cell replications per unit time, which is in turn reflected in
outgroups in order to determine whether the detected differences a smaller number of fixed substitutions. Thus, species with longer
occurred in the human or the chimpanzee lineage. As emphasized generation times are expected to exhibit slower rates of molecular
by Goodman et al. [2005], comparative sequence data from evolution than those with shorter generation times. Despite the
other primate species are an invaluable aid to the interpretation of relatively low degree of sequence divergence between humans and
results obtained from human/chimpanzee genome comparisons, chimpanzees, humans exhibit a significant slowdown of molecular
particularly with respect to the determination of the lineage evolution as determined by studies of smaller genomic regions
specificity of observed differences. The identification of the (e.g. [Li et al., 1987; Bailey et al., 1991; Jaruzelska et al., 1999;
genomic differences between humans and chimpanzees, together Kaessmann et al., 1999; Shi et al., 2003a; Ebersberger and Meyer,
with the discovery of the molecular signatures of deviations from 2005]) and confirmed genomewide employing baboons or rhesus
neutral evolution, promise to reveal those proteins and the nature macaques as outgroups [Elango et al., 2006]. The apparently high
of the functional networks that have evolved under lineage- degree of nucleotide sequence conservation manifested by the
specific adaptive constraints. chimpanzee and human genomes, a level of conservation which is
We attempt here to summarize recent findings, derived from the particularly pronounced in protein coding regions, is consistent
direct comparison of the human and chimpanzee genomes and with the view that the relatively low level of protein divergence
transcriptomes, that shed new light on the evolution of the human is on its own insufficient to account for the anatomical and
genome (Table 1). We explore how human/chimpanzee genome behavioral differences evident between humans and chimpanzees.
comparisons (as well as comparative studies involving the other Thus, it has been proposed that regulatory changes leading to gene
great apes and Old World monkeys) have contributed to our expression differences are likely to have strongly influenced human
understanding of structural (interspecies) divergence and (in- lineage-specific evolution [King and Wilson, 1975; Carroll, 2005].
traspecies) diversity. These studies are likely to have a very The extensive nucleotide sequence similarity evident between
considerable impact not only on our understanding of primate the human and chimpanzee genomes simply represents an overall
evolution but also on studies of human disease susceptibility/ average value that conceals important local differences. Certain
complex trait development. gene regions manifest considerably more divergence than others
(summarized in Supplementary Table S1; available online at http://
www.interscience.wiley.com/jpages/1059-7794/suppmat). The low-
NUCLEOTIDE SEQUENCE DIVERGENCE
est extent of sequence divergence is observed in coding regions, a
Genomewide nucleotide sequence data have allowed the finding that reflects negative selection against deleterious muta-
comparison of 2,400 megabases (Mb) of aligned DNA sequence tions. 30 untranslated regions (UTRs) have diverged at twice the
between humans and chimpanzees and have confirmed initial rate of coding sequences (Supplementary Table S1). This difference
assessments of a rather low overall degree of divergence (inter- would appear to be unrelated to the number of hypermutable
species differences) amounting to only about 1.23% [Mikkelsen CpG sites [Shen et al., 1994a] since 30 UTRs contain on average
et al., 2005]. Similar rates of divergence have been observed in about one-half as many CpG sites as coding regions [Hellmann
previous studies performed with more limited sequence datasets et al., 2003a]. Obviously, different functional constraints have
[Kumar and Hedges, 1998; Chen and Li, 2001; Ebersberger shaped the evolution of particular genic regions. The degree of
et al., 2002; Fujiyama et al., 2002; Wildman et al., 2003; Elango divergence evident in the 50 UTR is even higher than in the 30 UTR
et al., 2006]. (Supplementary Table S1). Taking intraspecies diversity into
For closely related species such as humans and chimpanzees, the account, the observed divergence between humans and chimpan-
level of nucleotide sequence divergence is strongly influenced zees in 50 UTRs is higher than expected and could underlie some
by the degree of polymorphism within the species. Thus, the of the gene expression differences noted between humans and
observed number of nucleotide differences has to be corrected for chimpanzees [Enard et al., 2002; Caceres et al., 2003; Karaman

Human Mutation DOI 10.1002/humu


TABLE 1. Summary of the Main Findings of the Genomewide Human vs. Chimpanzee Genome Comparisons

Category Results

Nucleotide divergence Number of xed nucleotide substitutions between PTR and HSA:1%.
Regional variation in divergence; substitution rates in subtelomeric regions are elevated in hominids as compared to murids.
Higher divergence in 5and 3 UTRs than in protein coding regions of genes.
Substitution rate at silent sites in exons is lower than at intronic sites, suggesting weak purifying selection operating on silent sites.
Chromosomal variation with highest sequence divergence found on theYchromosome (1.74%) and lowest on the X chromosome (0.94%).
Recombination pattern Recombination hotspots appear not be conserved between PTR and HSA.
divergence Genomewide correlation between sequence divergence and recombination rates.
Divergence/evolution of 70^80% of proteins are nonidentical between PTR and HSA, although they dier on average by only two amino acid residues.
proteins Average KA/KS ratio is 0.23; the proportion of advantageous mutations in the human lineage is lower than previously estimated but twice that estimated from
the mouse-rat comparison (0.13).
5% of proteins evolved under positive selection specically in the human lineage.
Proteins that have experienced adaptive evolution have been identied in many functional categories.Those associated with brain function/ development may
have been intimately involved in the evolution of human-specic traits.
Evolution of theY The absence of any loss or decay of X degenerate genes in HSA as compared to PTR does not favor theimpending demise hypothesis
chromosome for the humanYchromosome.
Human lineage-specic gene Gene loss or inactivation has not been a rare event during human evolution after the separation from PTR; 90 examples so far known.
inactivation Adaptive inactivation which has not yet been completely xed in the human population has been demonstrated in only one case (CASP12 gene)
Divergence in cancer-related High degree of overall conservation but a total of 1,542 amino acid changes were identied that could contribute to dierential cancer susceptibility
genes in PTR and HSA.
Expression divergence The highest degree of expression divergence has been observed in the testis, while the lowest expression divergence has been observed in the brain.
The majority of genes with expression divergence in the brain exhibit a human specic upregulation (increased expression).This human specic upregulation
has not been observed in the liver.
Parallel patterns of gene expression dierences and protein divergence have been detected, suggesting that both types of changes have evolved in concert.
Genes expressed in testis, and especially those located on the X chromosome, display high expression and sequence divergence but reduced diversity in humans,
indicating that genes involved in reproduction evolved under positive selection.
Evolution of transcriptional Adaptive evolution of human upstream regulatory regions l has been proven for the prodynorphin (PDYN) and the factorVII (F7) genes.
cis-regulation
Chromosomal dierences Nine cytogenetically detectable pericentric inversions and one fusion that gave rise to human chromosome 2 serve to distinguish PTR and HSA; no genes were
disrupted at the breakpoints.Two inversions (chromosomes 1 and 18) were xed in the human lineage.
Insertions/deletions Insertions of 10 to 15 kb have given rise to a total of 32 Mb of human-specic DNA sequence.
Insertions of 415 kb gave rise to 8 Mb of human-specic sequences.
Genomewide comparisons have indicated that some 40^45 Mb of lineage-specic sequence results from insertions/deletions resulting in copy number
dierences between HSA and PTR.
The number of Alu sequence insertions in HSA was 3.4-fold higher than in PTR. A dierent set of Alu sequences has been amplied in both species, while the
rate of L1 element retrotranspositional activity has been relatively similar in HSA and PTR.
Direct genome comparisons revealed 1,000 SVA retrotransposons specic to the human lineage and a comparable number of chimpanzee
lineage-specic integrations.
The large number of lineage-specic transposon insertions in PTR and HSA could have had functional consequences with respect
to gene expression or inactivation.
45 chimpanzee-specic endogenous retroviral element insertions and 73 human-specic insertions have been identied.
Microsatellite/RY-tract/ Microsatellites, triplet-repeats, and RY-tracts tend to be longer in HSA than in PTR suggesting that expansion-type mutations have occurred disproportionately
triplet repeat evolution in the human lineage.
Submicroscopic structural 33% of segmental duplications (26.5 Mb) are duplicated only in HSA; they map to at least 500 distinct regions of average length 55 kb and are distributed
divergence/segmental throughout the genome.
duplications There are many structural dierences in subtelomeric regions of HSA compared to PTR.
Microinversions of 41kb up to several Mb contribute signicantly to genome divergence and (intraspecies) diversity.
Human-specic genes/ At least 180 genes and partial gene sequences have become duplicated specically in the human genome.
HUMAN MUTATION 28(2), 99^130, 2007

pseudogenes Humans manifest a larger number of lineage-specic gene duplications than the great apes;134 gene families have been identied with human lineage-specic
gains including a number of genes with brain-related functions.
163 human-specic retrotransposed gene copies have been identied.

Human Mutation DOI 10.1002/humu


HSA, Homo sapiens; PTR, Pan troglodytes.
101
102 HUMAN MUTATION 28(2), 99^130, 2007

et al., 2003; Marvanova et al., 2003; Uddin et al., 2004; whole-chromosome comparison of human chromosome 21 and
Khaitovich et al., 2005a]. its chimpanzee homologue has indicated considerably higher
The nucleotide substitution rate in nondegenerate sites within divergence (1.44%) than the overall value for the genome
coding sequences is substantially lower than expected, probably a [Watanabe et al., 2004]. By contrast, the X chromosome exhibits
reflection of the action of purifying selection. Similarly, divergence the lowest sequence divergence for any chromosome (0.94%)
at silent sites (in particular four-fold degenerate sites where whereas the Y chromosome displays the highest (1.78% divergence
substitutions do not affect the encoded protein) is often lower as compared to the overall value of 1.23%) [Hughes et al.,
than at nearby intronic locations or within intergenic regions, a 2005; Kuroki et al., 2006; Patterson et al., 2006]. This is probably
finding that is also consistent with the action of purifying selection a consequence of the elevated germline mutation rate in males
[Hellmann et al., 2003a; Mikkelsen et al., 2005]. Even if CpG [Li et al., 2002; Makova and Li, 2002; Mikkelsen et al., 2005;
sites are disregarded, the divergence at four-fold degenerate sites is Taylor et al., 2006].
39% lower than in intergenic regions [Hellmann et al., 2003a].
This is in accord with the study of Bustamante et al. [2002], who
RECOMBINATION PATTERN DIVERGENCE
compared synonymous sites between functional human genes
BETWEEN HUMANS AND CHIMPANZEES
and pseudogenes. The reasons why purifying selection may have
acted on four-fold degenerate sites in humans may include codon Comparative analyses of recombination patterns in humans
usage bias [Lander et al., 2001], isoacceptor tRNA preference and chimpanzees may shed light on the evolution of a key
[Sharp and Li, 1986] and an influence on mRNA splicing. In mechanism which underlies the generation of genetic diversity
humans, codon usage bias is positively correlated with the breadth (reviewed by Nishant and Rao [2006]). Comparison of linkage
of the genes tissue expression profile [Urrutia and Hurst, 2001], disequilibrium landscapes in humans and chimpanzees has
conservation of mRNA secondary structure, and regulation of revealed major divergence in haplotype block structures and
mRNA splicing, as discussed by Hellmann et al. [2003a]. hence, by inference, in the positions of recombination hotspots
Human/chimpanzee genome comparisons have revealed regio- [Wall et al., 2003; Ptak et al., 2004, 2005; Winckler et al., 2005].
nal differences in sequence divergence such as dark G-bands In view of the low degree of sequence divergence between humans
on chromosomes. These bands are both G1C-poor and gene-poor, and chimpanzees, changes in local DNA sequence motifs are
exhibit a low recombination frequency [Holmquist, 1992], and unlikely to be the sole determinants of the differential recombina-
display a degree of divergence that is 10% higher than the tion hotspot distribution.
genome average. By contrast, greater divergence is evident Despite differences in the crossover hotspot patterns in humans
within the gene-rich and G1C rich regions at the ends of those and chimpanzees, a common gradient of increasing recombination
chromosomes that exhibit high recombination rates [Hellmann rate is apparent in the pseudoautosomal region on the short arm
et al., 2003b]. The variable degree of divergence in different (p-PAR) of the X chromosome in both species [Filatov, 2004]. The
chromosomal regions is probably influenced by a complex interplay mutagenic influence of recombination may make a significant
between different parameters such as gene content and recombi- contribution to the gradual increase in silent substitution rates
nation frequency, together with other factors (e.g., variation in the that extends from the proximal to the distal portion of the p-PAR.
mutation rate, biased gene conversion, and the DNA sequence Furthermore, the between-species silent divergence in the
context) [Lercher and Hurst, 2002; Meunier and Duret, 2004]. p-PAR also forms a gradient that increases towards the telomere
Remarkably, neither the sequence divergence nor the recombina- [Filatov, 2004; Bussell et al., 2006]. The correlation between
tion rate in distal chromosomal regions is increased in mouse or rat human-chimpanzee sequence divergence and recombination
[Jensen-Seaman et al., 2004], in contrast to the situation in rates is not restricted to the p-PAR but rather is observed
hominids (humans and great apes). Remarkably, the sequence genomewide, suggesting that mutation and recombination
divergence in the most distal 10 Mb of hominid chromosomes is are intimately associated processes [Lercher and Hurst, 2002;
10% higher than in rest of the genome [Mikkelsen et al., 2005]. Hellmann et al., 2003b].
By contrast, in the nondistal regions of chromosomes, divergence
is strongly correlated between hominids and murids. Accordingly,
TRANSSPECIES OR SHARED POLYMORPHISMS
a lineage-specific increase in sequence divergence may be noted
in hominids in the gene-rich distal chromosomal regions At the nucleotide sequence level, shared polymorphisms among
[Mikkelsen et al., 2005]. higher primate species are likely to be rare. They do however exist,
Considerable regional differences in sequence evolution are as is to be expected in cases of independently occurring mutations
evident on the Y chromosome [Ebersberger and Meyer, 2005]. In a or where positive selection for diversity has had a major influence
detailed comparison of a 90-kb region from the nonrecombining on allele frequencies, as with the human leucocyte antigen (HLA)
portion of the Y chromosome between humans and chimpanzees genes [Watkins, 1995]. Only a few shared human/chimpanzee
(using gorilla as an outgroup), significantly different region-specific RFLPs [Mountain and Cavalli-Sforza, 1994] or SNPs [Martinko
substitution rates have been observed. These differences were et al., 1993; Deeb et al., 1994; Jaeger et al., 1998; Hacia et al.,
found to be associated with a change in GC content that is 1999] have been identified to date. This may be in part because,
confined to the human lineage (and is not evident in the with the sole exception of the study of Asthana et al. [2005], no
chimpanzee). Recent species-specific changes in the regions systematic attempt has yet been made to search for shared
mutation pattern (as well as selective effects) may thus have polymorphisms between humans and chimpanzees.
driven the sequence divergence in this chromosomal region in Consistent both with theoretical expectations [Kimura, 1983]
both humans and chimpanzees [Ebersberger and Meyer, 2005]. and empirical observations [Kennedy et al., 2003], the vast
Nucleotide sequence divergence varies not only within but majority of identified human SNPs have had a relatively recent
also between chromosomes [Chen et al., 2001; Ebersberger evolutionary origin and are not shared by other primate species.
et al., 2002; Fujiyama et al., 2002; Smith et al., 2002; Mikkelsen Thus, the probability that a polymorphism originally present
et al., 2005; Hughes et al., 2005; Kuroki et al., 2006]. The in the common ancestor of chimpanzees and humans would have

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 103

survived for 4.6 million years (Myrs) appears to be on the order of In order to factor some of the above considerations into tests for
2  106 [Asthana et al., 2005]. It therefore follows that most deviation from neutrality based on KA/KS ratios, different methods
polymorphisms shared by humans and chimpanzees are more likely have been developed to search for positive selection acting on
to be the result of independent mutational events in both lineages given branches or on a subset of sites (summarized in Zhang
rather than being truly identical-by-descent. This notwithstand- et al. [2005a]). The use of different methods may explain certain
ing, the probable extreme rarity of shared identical-by-descent discrepancies between studies designed to identify genes under
polymorphisms renders their occasional identification all the positive selection in the human lineage [Clark et al., 2003; Dorus
more meaningful since they could at least in principle provide et al., 2004; Mikkelsen et al., 2005; Nielsen et al., 2005; Arbiza
information on relative rates of mutation, recombination, gene et al., 2006]. In a set of 13,454 human/chimpanzee orthologous
conversion, and even more importantly, evidence of positive gene pairs, the CSAC identified 585 genes exhibiting KA/KS ratios
selection due to heterozygote advantage. 41 in the human lineage as being potential candidates for positive
Plausible human examples of balanced functional polymorph- selection [Mikkelsen et al., 2005]. This proportion was lower
isms fulfilling a role in defense against pathogens are to be found in than estimates obtained from two other genomewide screens
the chemokine receptor [Tang et al., 2002], the HLA gene clusters for positively selected genes in human/chimpanzee comparisons
[Gao et al., 2001], as well as the cytokine network [Shin [Clark et al., 2003; Nielsen et al., 2005]. In addition, only a subset
et al., 2000]. Although these examples have originated relatively of genes was identified as having evolved under positive selection
recently in human evolution, the maintenance of variants in both common to all three studies.
species is indicative of rather more ancient selective pressures. The More recently, Arbiza et al. [2006] employed a two branch-site
identification of shared identical-by-descent polymorphism in maximum likelihood (ML) method corrected for multiple testing
humans and chimpanzees could serve to pinpoint genomic regions on a set of 13,198 genes with human, chimpanzee, mouse, rat,
harboring loci that manifest heterozygote advantage. However, and dog orthologs. According to their findings, some 5% of
humanchimpanzee genome comparisons have also proven useful human genes and 10% of chimpanzee genes have evolved under
in excluding the possibility of shared heterozygote advantage, as in positive selection. The higher number of positively selected
the case of the variants that impart bitter taste sensitivity; genes in chimpanzees as compared to humans may have been a
variation at the TAS2R38 locus, the molecular basis of taste consequence of the smaller effective population sizes during
sensitivity, has been shown to have arisen independently in the human evolution [Chen and Li, 2001].
two primate species [Wooding et al., 2006]. Arbiza et al. [2006] also observed that many functional
categories were overrepresented among the genes found to have
evolved under positive selection, for example nociception/sensory
perception, brain-related functions and development, pathogen
ADAPTIVE EVOLUTION OF PROTEINS
resistance, immunity, reproduction, dietary adaptation, and trans-
Differences in the evolutionary rates of genes indicate that both criptional regulation. These observations are in accord with the
positive and negative selection have operated extensively on the results of Clark et al. [2003] as well as findings from studies that
human and chimpanzee genomes [Lu and Wu, 2005]. Genes that employed more limited datasets and those that focused upon
have experienced positive selection specifically in the human individual genes [Yang, 1998; Wyckoff et al., 2000; Enard et al.,
lineage, promise by virtue of their function to identify the 2002; Swanson and Vacquier, 2002; Choi and Lahn, 2003; Gilad
molecular mechanisms responsible for the development of human et al., 2003a; Maier et al., 2003; Wang et al., 2003; Bersaglieri
specializations [Sabeti et al., 2006]. et al., 2004; Dorus et al., 2004; Kitano et al., 2004; reviewed in
An estimated 70 to 80% of protein sequences are nonidentical Vallender and Lahn, 2004a; Bustamante et al., 2005; Clark and
between humans and chimpanzees, although these proteins differ Swanson, 2005; Nielsen et al., 2005; Dorus et al., 2006; Verrelli
by on average only two amino acid residues [Glazko et al., 2005; et al., 2006; Voight et al., 2006]. Several of the above-mentioned
Mikkelsen et al., 2005]. To establish whether the fixation of biological processes, in particular pathogen resistance and
amino acid replacements has been a consequence of positive reproduction, are likely to have been influenced by selection not
selection (and hence may be regarded as adaptive changes), the only in humans but also in chimpanzees [Arbiza et al., 2006]. By
KA/KS ratios (representing the relative ratio of nonsynonymous/ contrast, adaptive evolution associated with brain function/
synonymous substitutions) are determined as pairwise values development may have been intimately related to the emergence
between the human and chimpanzee sequences [Kimura, 1968; of human-specific traits.
Eyre-Walker and Keightley, 1999]. If KA/KS41, then the gene in Of especial interest are those genes that have not only evolved
question may be considered to have evolved under directional under positive selection in early primate evolution but which
positive selection [Yang and Bielawski, 2000]. Neutrally also still display signatures of positive selection postdating
evolving genes generally have KA/KS values around 1, whereas the emergence of anatomically modern humans, e.g., micro-
most proteins have KA/KS values r1 and hence may be cephalin (MCPH1) and abnormal spindle-like microcephaly-associated
considered to be under purifying or negative selection. The power (ASPM). Both these genes may influence brain size since muta-
to detect genes that have evolved under positive selection by KA/ tions in them cause primary microcephaly, a neurodevelopmental
KS determination using the complete coding sequence is, however, disorder characterized by a global reduction in cerebral cortical
often quite low [Kitano et al., 2004; Ellegren, 2005; Nielsen et al., volume but with no major abnormalities in cortical architecture
2005] and even genes with KA/KS values o1 may have evolved [Bond et al., 2002; Jackson et al., 2002; Roberts et al., 2002].
under positive selection [Dorus et al., 2004]. Furthermore, the Remarkably, both genes display evidence of strong selection in the
significance of single adaptive amino acid changes within primate lineage leading to humans. The ASPM gene experienced
crucial functional domains may not be immediately apparent pronounced positive selection in the African hominoid clade
[Andres et al., 2004a], particularly if negative selection has [Evans et al., 2004a; Kouprina et al., 2004a], whereas the MCPH1
acted upon other sites in the protein in question and the overall gene did so in an earlier phase of primate evolution. Strong
KA/KS value is less than 1. positive selection has impacted upon the MCPH1 gene in New

Human Mutation DOI 10.1002/humu


104 HUMAN MUTATION 28(2), 99^130, 2007

World monkeys, Old World monkeys, gibbons, and orangutan is very different from that expected in the absence of selection
[Evans et al., 2004b]. Intriguingly, both brain-expressed genes [Maynard Smith and Haigh, 1974; Fay and Wu, 2000; Bamshad
appear to have been continuing their trend of adaptive evolution and Wooding, 2003; Enard and Paabo, 2004]. The selective sweep
since certain variants or haplotypes have been reported to increase in the FOXP2 region appears to have occurred during the last
in frequency in human populations more rapidly than would be 100,000 years [Zhang et al., 2002b], a time period that overlaps
predicted by random drift alone [Evans et al., 2005; Mekel-Bobrov the emergence of modern humans and the acquisition of their
et al., 2005]. The biological basis for this selection (for example, language capability. Although FOXP2 may indeed be causatively
whether it is associated with the proliferation of neural progenitor associated with this most characteristic of human features, it is
cells) is unclear, but its elucidation promises to yield new not necessarily going to be unique in this regard. Moreover, as a
insights into basic mechanisms of human brain evolution [Stern transcription factor whose activity is not restricted to the brain,
and Woods, 2006]. FOXP2 clearly does not control language capability directly but
Interestingly, MCPH1 is located in a very rapidly evolving rather may either play a more indirect role in a neuronal pathway
15-Mb region on chromosome 8p; this region displays an extremely that regulates language acquisition or instead regulate anatomical
high degree of nucleotide sequence divergence between human requirements for speech development outside the brain. Human
and chimpanzee, the highest so far reported for the autosomes language ought not to be viewed as an innovation that occurred
[Nusbaum et al., 2006]. Apart from the MCPH1 gene, this region specifically (and in its entirety) in the human lineage but rather
also contains other genes that appear to have evolved under perhaps as a complex reconfiguration of preexisting systems that
positive selection in primates, such as the major defensin gene evolved coordinately (reviewed by Fisher and Marcus [2006]).
cluster [Boniotto et al., 2003; Maxwell et al., 2003; Semple et al., Thus, the identification of genes that evolved under recent
2003, 2005; Xiao et al., 2004]. The defensin genes encode a family positive selection may help us to identify the mechanistic origins of
of peptides that are involved in the immune response [Lehrer, human speech development.
2004] and may play a role in conferring differential disease Regions in the human genome where diversity is lower than
susceptibility in humans. Genes and segmental duplications within expected (as deduced from the divergence rate with the
the defensin gene cluster exhibit considerable copy number chimpanzee) can help to pinpoint regions that have evolved
variation between human populations [Mars et al., 1995; Hollox under positive selection in the human lineage. Mikkelsen et al.
et al., 2003; Aldred et al., 2005] as well as haplotype diversity [2005] identified six such regions of reduced diversity that serve as
[Taudien et al., 2004], suggesting persistent adaptive evolution in indicators of strong selective sweeps that have acted during the
humans and chimpanzees. It is possible that MCPH1 variants last 250,000 years of human evolution (Supplementary Fig. S1).
could have become fixed by hitchhiking along with new defensin Some of these genomic regions, such as that on chromosome 22,
variants as they themselves became fixed. are quite gene-rich, whereas others, such as that on chromosome
The adaptive evolution of brain-expressed genes in primates is 4, do not contain any genes. Detailed analyses of the genetic
unlikely to be restricted to ASPM and MCPH1. Indeed, 17 out of variation patterns of these regions can help to identify genes or
24 genes with marked KA/KS disparities between primates and regulatory elements that have been the target of selection.
rodents are linked to brain development and physiology [Dorus Some major differences between humans and chimpanzees
et al., 2004]. However, the issue of lineage specificity remains with respect to disease (in particular, cancer) susceptibility have
questionable, since the accelerated evolution of these genes been noted (summarized by Varki and Altheide [2005]). Cancer-
has also been observed in the dog lineage as compared to mouse related genes may have experienced greater selective pressure than
[Lindblad-Toh et al., 2005]. When human and chimpanzee are other disease genes [Thomas et al., 2003]; for example, adaptive
compared, genes with maximal expression in the brain exhibited evolution of the breast cancer (BRCA1) and angiogenin (ANG)
little or no evidence of positive selection in the human lineage genes has been evident during primate evolution [Huttley et al.,
[Nielsen et al., 2005]. Further, of 2,633 human brain-expressed 2000; Zhang and Rosenberg, 2002; Pavlicek et al., 2004]. Puente
genes, only 47 displayed evidence of positive selection in the et al. [2006] identified a total of 1,542 amino acid changes in 333
human lineage but not in chimpanzees [Yu et al., 2006]. cancer-related genes that could, at least in principle, contribute to
These findings support the view that cognitive differences might a possible differential cancer susceptibility between humans and
be related to adaptive changes in only a few genes with low chimpanzees. In addition, 20 cancer-related genes were identified
or regionally restricted expression in the brain and/or changes in that contain codon insertions or deletions in their protein
gene expression. coding regions when humans and chimpanzees are compared;
The insufficient sensitivity of whole-genome analyses based on 70% of these occurred within trinucleotide repeats, underlining
relative ratio tests to detect all genes under positive selection is the genetic instability of these sequences (summarized in
evidenced by examples such as FOXP2, which was not identified Supplementary Table S2).
as being positively selected by Nielsen et al. [2005] and Arbiza
et al. [2006]. FOXP2 is an attractive candidate for involvement in
NEGATIVE SELECTION
human speech development. Carriers of FOXP2 mutations
IN THE HUMAN LINEAGE
display impairment in writing, verbal articulation (developmental
verbal dyspraxia [DVD]), and grammatical usage [Lai et al., 2001; Genes with signatures of negative selection (KA/KSr1) are
Bishop, 2002]. Although the FOXP2 gene has been very highly also potentially important in the context of medical
conserved in mammals, two amino acid changes occurred genetics. Genes that have evolved under negative or balancing
specifically in the human lineage; this was accompanied by the selection attract attention by virtue of the minimal amino acid
loss of local DNA sequence diversity in the FOXP2 gene region sequence divergence between humans and chimpanzees since
[Enard et al., 2002; Zhang et al., 2002a]. Such a selective sweep those amino acid substitutions that have occurred are likely to be
generally occurs when an advantageous allele arises and spreads deleterious to differing extents and hence are less likely to
rapidly through the population. Concomitantly, adjacent regions become fixed. Such a pattern was evident in 13.5% of 6,000
hitchhike with the advantageous site and display a genealogy that genes analyzed by Bustamante et al. [2005]. Those human genes

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 105

that manifest low sequence divergence but a high degree of interspecies homoplasy makes genegene direct comparisons
intraspecific diversity are particularly worthy of note since these difficult between human and chimpanzee mitochondrial sequences
genes could harbor an excess of mildly deleterious variants. [Kivisild et al., 2006], mitochondrial genome diversity does appear
Interestingly, cytoskeletal proteins and proteins involved in both to be markedly higher in chimpanzees than in humans, possibly as
ectoderm development and neurogenesis display signatures of a result of recent human population bottlenecks [Wise et al.,
negative selection with an excess of amino acid polymorphism 1997].
relative to divergence. A substantial proportion of genes with Sequence transfer from the mitochondrial genome to the
signatures of negative selection are known to be associated with nuclear genome has been an ongoing process during mammalian
Mendelian disorders (including Duchenne muscular dystrophy, evolution. Although the precise mechanism of sequence transfer
Huntington disease and Usher syndrome among others [Busta- is unclear, more than 500 of these nuclear DNA sequences of
mante et al., 2005]). It follows that weak negative selection mitochondrial origin (NUMTs) have been identified in the human
signatures could prove useful in identifying genes associated with genome. Further, a total of 27 NUMTs have been identified as
complex disorders. being human-specific, having been inserted into the human
nuclear genome over the last 5 to 7 Myrs [Ricchetti et al., 2004.
EVOLUTION OF THE HUMAN Y CHROMOSOME
HUMAN LINEAGE-SPECIFIC GENE INACTIVATION
The Y chromosome has a number of unique features; i.e., it is
haploid and male-specific. Owing to its lower effective population The less is more hypothesis has emphasized the importance of
size as compared to other chromosomes, selection acting on the Y loss-of-function mutations during human genome evolution
chromosome is necessarily weaker. On the other hand, the Y [Olson, 1999; Olson and Varki, 2003]. In this context, gene loss
chromosome evolves more rapidly than other chromosomes due to or pseudogenization could have had a much greater impact than
the higher mutation rate in the male germline [reviewed in Jobling most amino acid replacements due to the immediate loss of gene
and Tyler-Smith, 2003; Tyler-Smith et al., 2006]. X and Y function [Wang et al., 2006]. A good example of this principle is
chromosomes coevolved from an ancestral pair of autosomes that provided by MYH16, a myosin heavy chain gene expressed in the
existed 300 Mya [Lahn and Page, 1999]. Since then, the Y jaw musculature and inactivated by a human-specific truncating
chromosome has degenerated gradually both in terms of gene mutation that is thought to have originated 2.4 Mya [Stedman
content and size, because over most of its length it does not et al., 2004]. This inactivation event therefore predates the
recombine during meiosis [Charlesworth and Charlesworth, 2000]. emigration of Homo from Africa and could represent a molecular
The considerable degree of evolutionary decline has suggested to adaptation that enabled remodeling of the hominid cranium by
some that the Y chromosome could become devoid of any reducing masticatory musculature.
functional gene within 10 Myrs [Aitken and Marshall Graves, Numerous other examples of human-specific gene inactivation
2002]. As a means to investigate this impending demise events have been identified (listed in Table 2). These inactivations
hypothesis, which implies that the Y chromosome is continually were either caused by nonsense/frameshift mutations or by the loss
losing genes shared with the X, the comparison of Y-linked genes of paralogous gene copies that were duplicated during hominoid
in humans and chimpanzees has proven very informative. Hughes evolution. One gene family that has been subject to extensive
et al. [2005] compared 16 Y chromosomal genes and 11 gene loss in the human and primate lineages (as compared to
pseudogenes located in the so called X degenerate regions of rodents) is that of the olfactory receptor (OR) genes [Rouquier
humans and chimpanzees. These genes have diverged substantially et al., 1998; Sharon et al., 1999; Young et al., 2005], one of the
during the period of separated evolution of the X and Y largest mammalian gene superfamilies, comprising 41,000 genes
chromosomes and are not involved in recombination or gene [Young and Trask, 2002; Grus et al., 2005]. Remarkably, humans
conversion as are genes located in the palindromic sequences that have accumulated mutations that have disrupted OR gene coding
are abundant on the Y chromosome [Rozen et al., 2003; Skaletsky regions roughly four-fold faster than the great apes or macaques.
et al., 2003]. These comparisons indicated no significant loss or Of 50 human OR genes comparatively investigated in humans and
decay of these genes in the human lineage [Hughes et al., 2005] other primates, 54% were found to contain inactivating mutations
and the results of these analyses were confirmed by comparison and thus represent pseudogenes [Gilad et al., 2003b]. Although
with another chimpanzee individual [Kuroki et al., 2006]. This the pseudogenization of OR genes has also been noted in
observation is clearly inconsistent with the impending demise nonhuman primates, a much higher attrition rate in terms of
hypothesis for the human Y chromosome since no gene loss or functional OR genes has occurred in the human lineage, possibly
decay has occurred during the last 5 to 7 million years of human due to reduced chemosensory dependence as compared to the
evolution. Furthermore, human/chimpanzee sequence compari- great apes [Gilad et al., 2003b].
sons revealed pronounced coding sequence conservation in the X Another very interesting example of gene inactivation,
degenerate genes, indicating that their function has been occurring specifically in the human lineage, was mediated by an
maintained by purifying selection [Hughes et al., 2005]. Alu insertion into the CMP-Neu5Ac hydroxylase (CMAH) gene,
an event that occurred after the separation of humans and
chimpanzees. Owing to this deficiency, N-acetylneuraminic acid
COMPARATIVE EVOLUTION OF THE HUMAN
(Neu5Ac) cannot be transformed into N-glycolylneuraminic acid
AND CHIMPANZEE MITOCHONDRIAL GENOMES
(Neu5Gc), which is consequently absent in humans but present in
Comparison of the complete sequences of the human and the great apes [Chou et al., 1998; Irie et al., 1998; Muchmore
chimpanzee mitochondrial genomes has yielded a value of 8.9% for et al., 1998; Hayakawa et al., 2001]. Neu5Gc and Neu5Ac are the
the nucleotide sequence divergence between the two primate major sialic acids that function as cell-surface glycoconjugates and
species [Arnason et al., 1996]. Corrected for the actual mutation appear to be responsible for the observed humanchimpanzee
rates, however, divergence estimates have sometimes been differences in susceptibility to malaria [Martin et al., 2005].
reported to be twice as high. Although the high level of Interestingly, loss of Neu5Gc would appear to have initiated other

Human Mutation DOI 10.1002/humu


TABLE 2. Genes Inactivated in the Human Lineage but Functional in the Chimpanzee
106

Chromosomal
Gene Pseudogene description Functional context (type of inactivating mutation) localization References

BASE Breast cancer and salivary gland Unknown (frameshift 1-bp deletion) 20 Hahn and Lee [2005]; Wang et al. [2006]
expression
DNAJB3 DnaJ (Hsp40) homolog, subfamily B, Heat shock protein binding (frameshift 1-bp insertion) 2 Hahn and Lee [2005]; Wang et al. [2006]
member 3
FLJ33674 Hypothetical protein FLJ33674 Unknown (frameshift deletion) 3 Hahn and Lee [2005]; Wang et al. [2006]
HEJ1 Transcribed processed pseudogene Unknown (insertion of 2 nucleotides) 1 Hahn and Lee [2005]
of DNAJA1
NTSR2 Neurotensin receptor 2 Levocabastine-sensitive G protein-coupled neurotensin 2 Hahn and Lee [2005]

Human Mutation DOI 10.1002/humu


receptor 2 (frameshift deletion)
RPL13AP Transcribed processed pseudogene Unknown (insertion of 2 nucleotides) 14 Hahn and Lee [2005]
of RPL13A
SCGB1D4 Secretoglobin family 1D member 4 Expression is inducible by interferon-g; involved in 11 Hahn and Lee [2005]
chemotactic migration and in invasion of lymphoblast
cells (frameshift deletion)
WBSCR27 Williams Beuren syndrome chromosome Unknown (frameshift 11-bp insertion) 7q11.23 Hahn and Lee [2005]; Wang et al. [2006]
region 27
ZCCHC13 Zinc nger, CCHC domain containing 13 DNA binding (1-bp insertion) X Hahn and Lee [2005]
HUMAN MUTATION 28(2), 99^130, 2007

CML2 Putative N-acetyltransferase Homology to bacterial acetyltransferases involved in drug 2p13.1 Hahn and Lee [2006]
Camello-like 2 resistance (nonsense mutation)
FLJ14640 Hypothetical protein ATPase domain; function unknown (nonsense mutation) 19q13.11 Hahn and Lee [2006]
MT1L Metallothionein 1L Heavy metal binding activity (nonsense mutation) 16q13 Hahn and Lee [2006]
NPPA Natriuretic peptide precursorA Ligand- and retinoid X receptor-dependent, hormonal 1p36.21 Hahn and Lee [2006]
activity (nonsense mutation)
PDE3B Phosphodiesterase 3B, cGMP-inhibited 3 0, 50 -cyclic-nucleotide phosphodiesterase activity; 11p15.1 Hahn and Lee [2006]
hydrolase activity (nonsense mutation)
SERPINA13 Serine peptidase inhibitor, clade A, Serine-type endopeptidase inhibitor activity 14q32.13 Hahn and Lee [2006]
member 13 (nonsense mutation)
TAP2 Transporter 2, ATP-binding cassette, Membrane-associated, involved in multidrug resistance, 6p21.3 Hahn and Lee [2006]
sub-family B heterodimerization withTAP1 (nonsense mutation)
UIP1 26S proteasome-associated UCH Interaction with ubiquitin C-terminal hydrolase Xq28 Hahn and Lee [2006]
interacting protein 1 37 (nonsense mutation)
ZNF277 Zinc nger protein (C2H2 type) 277 Zinc ion binding, transcription factor activity (nonsense 7q31.1 Hahn and Lee [2006]
mutation)
MYH16 Myosin,heavy polypeptide 16 Sarcomeric myosin heavy chain expressed in non-human 7q22.1 Stedman et al. [2004]; Wang et al. [2006]
primate masticatory muscles (frameshift mutation)
CASP12P1 Caspase-12 pseudogene Cysteine protease that cleaves C-terminal 11q22.3 Fischer et al. [2002]; Saleh et al. [2004];
aspartic acid residues on their substrate molecules Wang et al. [2006]
(nonsense mutation)
FMO2 Flavin containing monooxygenase 2 NADPH-dependent enzyme that catalyzes the oxidation of 1q23^q25 Dolphin et al. [1998]
many drugs and xenobiotics (nonsense mutation)
TCRGV10 Tcell receptor gamma variable 10 (point mutation in theV10 leader donor splice site) 7p15 Zhang et al. [1996]
OR 36 olfactory receptor (OR) pseudogenes Interaction with odorant molecules to initiate a neuronal Gilad et al. [2003b]; Wang et al. [2006]
response and perception of smell (nonsense mutations)
ELA1 Pancreatic elastase 1 Serine protease (inactivating substitutions in enhancer and 12q13 Rose and MacDonald [1997]
promoter)
KRTHAP1 Keratin, hair, acidic pseudogene 1 Intermediate lament (nonsense mutation) 17q12^q21 Winter et al. [2001]
TAS2R62P, Bitter taste receptor pseudogenes Bitter taste reception (nonsense mutation) 7q35,7p21.3, Conte et al. [2002]; Shi et al. [2003b];
TAS2R2, 12p13.2 Wang et al. [2004, 2006]; Go et al.
TAS2R64P [2005]
NAPSB Napsin B peptidase pseudogene Napsin B aspartic peptidase (nonsense mutation) 19q13.33 Puente et al. [2005]
HUMAN MUTATION 28(2), 99^130, 2007 107

adaptive changes in human sialic acid biology affecting Siglec-9

Hamann et al. [2003]; Wang et al. [2006]


and Siglec L1 (Siglec XII), sialic acid-binding receptors of innate

Muchmore et al. [1998]; Hayakawa


immune cells [Angata et al., 2001; Sonnenburg et al., 2004].

Chou et al. [1998]; Irie et al. [1998];

et al. [2001]; Wang et al. [2006]


Further, the human-specific loss of Siglec expression on T cells is
associated with Tcell hyperactivity, which might itself be related to
the striking differences between humans and chimpanzees in terms
McEvoy and Maeda [1988]

of both the prevalence and severity of T-cell mediated diseases,


such as acquired immunodeficiency syndrome (AIDS) [Nguyen
Angata et al. [2004]

et al., 2006a].
Jury et al. [1998]

In addition to the human-specific gene inactivation events


listed in Table 2, Wang et al. [2006] identified a further 67
pseudogenes that have been generated specifically in the human
lineage after the split from the chimpanzee lineage. This serves
to indicate that gene losses restricted to the human lineage are by
no means rare occurrences. Wang et al. [2006] allocated these
67 pseudogenes, together with 13 previously described human-
12q24.12^q24.13

specific pseudogenes, to functional categories. An overrepresenta-


6p21.32
19p13.3

tion of functional categories such as chemoreception, immune


16q22

response, and calcitonin receptor activity was noted, which might


be associated with species-specific physiological differences in
olfaction [Glusman et al., 2001; Shepherd, 2004] or pathogen
susceptibility [Escalante et al., 1995; Novembre et al., 1997]. The
genes involved in the latter two processes are known to have
Metallopeptidase domain 1 (small insertions, deletions and
Present in hominoids, but deleted in humans. Hominoids

evolved rapidly [Grus et al., 2005; Kelley et al., 2005] but the
G-protein coupled receptor activity, calcium ion binding,

contribution of adaptive gene loss within these functional


N-glycolylneuraminic acid (Alu repeat insertion)

categories remains to be clarified [Wang et al., 2006].


Immunoglobulin superfamily member lectins that

Although the importance of the aforementioned gene inactiva-


Converts sialic acid N-acetylneuraminic acid to
selectively recognize sialic acids (gene loss)
integral to membrane (nonsense mutation)
have 3 haptoglobin genes, humans have 2

tion events for human evolution remains to be established, the


idea encapsulated in the gene loss hypothesis is highly attractive
since it provides a mechanism whereby trait evolution can occur
rapidly without being dependent upon the occurrence of several
genetic alterations acting additively, each of which would
individually be subject to the vagaries of genetic drift [Olson
and Varki, 2003]. The above notwithstanding, many of the
(gene loss in humans)

nonsense mutations)

published examples of gene loss in the human lineage may well


have been neutral with respect to fitness, with the null allele
having simply become fixed through genetic drift.
One good example of adaptive pseudogenization is provided by
the human-specific inactivation of the caspase-12 (CASP12) gene.
The CASP12 gene has been specifically inactivated in humans by
a protein truncating transition but remains functional in other
mammals [Saleh et al., 2004]. Caspase 12 is thought to play an
important role in apoptosis and the processing of inflammatory
cytokines, including the interleukin-1 (IL-1) and nuclear factor
EGF-like module containing, mucin-like,

acetylneuraminic acid hydroxylase

(NF)-kB pathways [Nakagawa et al., 2000; Lamkanfi et al., 2002;


A disintegrin and metalloproteinase
domain 1 (fertilin a) pseudogene

Saleh et al., 2004]. The human-specific inactivating mutation has


Sialic acid binding Ig-like lectin 13

been identified in all Caucasians and Asians but has not yet been
Cytidine monophosphate-N-

fixed as a null-allele in humans since it is present in only 80% of


hormone receptor-like 4

Africans [Fischer et al., 2002; Saleh et al., 2004]. In about 20%


of people of African descent, the functional caspase 12 polypeptide
is still expressed, as it is in New World and Old World monkeys
and rodents [Saleh et al., 2004]. The full-length caspase-12
isoform encoded by the functional gene confers endotoxin
Haptoglobin

hyporesponsiveness, which appears to manifest in the clinic as


an increased susceptibility to severe life-threatening sepsis. By
contrast, possession of the truncated caspase-12 isoform is
associated with a reduced incidence of severe sepsis and
consequent mortality [Saleh et al., 2004]. By means of diversity
analyses of the region around the inactivating mutation, performed
to search for selective sweeps and to make coalescent simulations,
SIGLEC13
ADAM1

the drift towards the fixation of the CASP12 null allele has been
CMAH
EMR4

driven by positive selection [Wang et al., 2006; Xue et al., 2006].


HPP

The pseudogenization of the CASP12 gene appears to have

Human Mutation DOI 10.1002/humu


108 HUMAN MUTATION 28(2), 99^130, 2007

occurred prior to the out-of-Africa migration of modern humans genomewide changes in gene expression associated with aging in
because the CASP12 T alleles of Africans and non-Africans share different species [Fraser et al., 2005].
the same origin [Wang et al., 2006]. Although CASP12 represents The highest number of genes displaying differences in expres-
the first reported case of adaptive gene loss in humans, the high sion between human and chimpanzee was noted in the testis
frequency of pseudogenization and examples of adaptive gene loss [Khaitovich et al., 2005a]. By contrast, the lowest degree of gene
in Drosophila [Takahashi et al., 2001; Greenberg et al., 2003, 2006] expression divergence between these species was observed in
suggest that this process may have generally played a key role the brain, probably due to the significant functional constraints
in evolution. enforced upon this organ in higher primates [Enard et al.,
It is of course not only human-specific gene losses that will have 2002; Khaitovich et al., 2005a]. Remarkably, most of the genes
contributed to the genomic differences between humans and differentially expressed in the brain of humans as compared to
chimpanzees; the inactivation or loss of genes in the chimpanzee chimpanzees exhibit increased expression in humans [Enard et al.,
lineage is also likely to have played a role. Indeed, Mikkelsen 2002; Caceres et al., 2003; Gu and Gu, 2003; Hsieh et al., 2003].
et al. [2005] identified a total of 53 human genes that were The upregulation or induced expression in the human brain as
either partially (17) or entirely (36) deleted in the chimpanzee. compared to chimpanzees would appear, however, to be brain-
According to the preliminary results of Wang et al. [2006], the specific, since in the liver no systematic increase or decrease in
chimpanzee genome may contain even more pseudogenes than the regulation of differentially-expressed genes in either humans or
that of humans, although the availability of a more refined chimpanzees was observed [Gilad et al., 2006]. In contrast to
sequence of the chimpanzee genome will be required to draw firm earlier studies, these authors used species-specific microarrays for
conclusions. It may be that some of the gene inactivation events five primate species including human and chimpanzee cDNAs to
that originally occurred in incipient chimpanzee populations avoid errors due to sequence divergence. They observed that most
were important in bringing about the permanent separation of genes are under selective pressure to maintain a constant level of
this lineage from that of early ancestral humans. Further in- expression and that only a few genes (110/1,056) are differentially
depth analyses of these gene losses in the chimpanzee are expressed in humans as compared to chimpanzees. Among the
needed to evaluate whether they occurred specifically in the genes manifesting a higher expression level in humans, transcrip-
chimpanzee lineage or whether they actually represent tion factors are themselves overrepresented, in contrast to
human-specific gains. chimpanzees. Since transcription factors regulate gene expression,
Interestingly, X-degenerated genes on the Y chromosome have these findings support the view that many differences between
exhibited a significant level of gene decay during chimpanzee humans and chimpanzees may be due to changes in gene
evolution although not in the human lineage [Hughes et al., regulation [King and Wilson, 1975]. Remarkably, transcriptional
2005]. The causes and consequences of this lineage-specific repressors of the KRAB-ZNF gene family are also characterized by
decay process are currently unknown but it has probably been rapid expansion and lineage-specific diversification during primate
influenced by strong positive selection focused on other regions of evolution [Hamilton et al., 2006].
the chimpanzee Y chromosome [Hughes et al., 2005]. Since the Y It is always possible that measured mRNA levels may not reflect
chromosome does not for the most part participate in recombina- the actual levels of functionally active proteins synthesized by
tion, selection must act on the chromosome as a unit. The genes the respective genes [Gygi et al., 1999; Preuss et al., 2004].
under positive selection on the chimpanzee Y chromosome could Interestingly, parallel patterns of gene expression differences and
be those specifically expressed in the testis and involved in protein divergence have been observed in comparisons of human
improving sperm competition [Rice, 1987]. It is not unreasonable and chimpanzee transcriptomes and proteomes [Bustamante
to assume that these genes could have evolved under powerful et al., 2005; Khaitovich et al., 2005a; Nielsen, 2006; Gilad
selective pressure in view of the fairly promiscuous mating et al., 2006]. These parallel patterns indicate that significant gene
behavior of extant chimpanzees [Gagneux et al., 1999]. expression differences correlate with extensive divergence of the
encoded proteins. This suggests that both types of change have
evolved in concert and that similar factors may have influenced
EXPRESSION REGULATION AND
protein and gene expression divergence [Khaitovich et al., 2005a].
TRANSCRIPTOME EVOLUTION
Genes with tissue-specific expression patterns tend to display
Gene Expression Divergence Between Humans
the highest degree of divergence at both RNA and protein levels,
and Chimpanzees
whereas genes that are expressed in several tissues tend to be the
Changes in gene expression have had a major impact on the least divergent between the species and to show the most minimal
evolution of eukaryotes [Wray et al., 2003] and hence probably expression differences [Khaitovich et al., 2005a]. This probably
also on human gene evolution [King and Wilson, 1975; Carroll, reflects the greater selective constraints acting on genes that are
2005]. With the advent of DNA microarray technologies, it has expressed in multiple tissues.
become possible to perform genomewide interhominoid trans- If these comparative analyses of gene expression and
criptome comparisons [Enard et al., 2002; Caceres et al., 2003; protein divergence are complemented with data on intraspecies
Karaman et al., 2003; Marvanova et al., 2003; Uddin et al., 2004; variation (diversity) patterns, it should be possible to identify
Khaitovich et al., 2005a; Gilad et al., 2006]. These analyses genes that have evolved under positive selection. For example,
have revealed many differences in the species- and tissue-specific genes expressed in testis, and especially those located on
expression patterns of both humans and chimpanzees. Further, the X chromosome, display high expression and sequence
microarray analyses have detected considerable diversity within divergence but reduced diversity as compared to other tissues
the human brain with respect to age-related changes in gene such as liver, heart, and kidney [Khaitovich et al., 2005a]. This is
expression. In the chimpanzee cortex, age-related changes in gene compatible with previous observations that genes encoding
expression have also been observed, but these patterns were proteins involved in reproduction tend to have evolved
significantly different from those exhibited by the human cortex. under positive selection [Wyckoff et al., 2000; Swanson and
These findings imply the potential for the rapid evolution of Vacquier, 2002].

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 109

Possible Causes of Gene Expression Divergence accelerated evolution of transcription factors is indicative of how
a relatively small number of genetic changes in key locations may
The central question remains as to the molecular basis (and its
pleiotropically affect the expression patterns of a myriad of
ultimate causes) of the observed gene expression divergence
different genes by influencing their transcription factor binding
between humans and chimpanzees. The processes involved are
capacity. We may therefore surmise that the accelerated evolution
likely to include the sequence divergence of regulatory
of transcription factors in the human lineage has in all likelihood
regions, differences in the control of transcriptional initiation,
had a major impact on gene expression divergence between
RNA processing and translation, the modification of chromatin
human and chimpanzee.
structure, and potentially also differences in DNA methylation.
When humans and chimpanzees have been compared with
Sequence variation in gene promoter regions has certainly
respect to DNA methylation patterns, the degree of CpG
been shown to influence transcription levels in individual
methylation has generally been found to be higher in the human
humans [Hoogendoorn et al., 2003; Bray et al., 2003; Trinklein
brain as compared to chimpanzee brain. Higher levels of CpG
et al., 2003; Buckland, 2004], probably due to the consequent
methylation are also apparent in other human tissues compara-
differential binding of cis- and trans-acting regulatory factors
tively investigated, such as liver and lymphocytes, but are not
[Pilpel et al., 2001]. In the same vein, nucleotide sequence
as pronounced as those noted for the brain. There is, however,
divergence is also likely to be responsible for differentially modulat-
no evidence as yet that these changes in methylation status
ing promoter activity in humans as compared to chimpanzees,
are associated with the observed expression divergence [Enard
one example being the apolipoprotein(a) (LPA) gene promoter
et al., 2004].
[Huby et al., 2001].
A general tendency has been noted for the 50 upstream
noncoding regions of genes to exhibit higher rates of nucleotide Adaptive Evolution of Transcriptional Regulation
sequence divergence between humans and chimpanzees than in Although our knowledge of the evolution of transcriptional
coding sequences and 30 UTRs [Chen and Li, 2001; Hellmann cis-regulation is still rather limited and will probably be very
et al., 2003a; Shi et al., 2003a; Keightley et al., 2005]. However, difficult to investigate on a large scale [Huby et al., 2001; Heissig
the determination of the overall divergence of promoter regions is et al., 2005], the population genetic and phylogenetic evidence
probably always going to be of limited utility since comparatively for positive selection acting on regulatory mutations at the human
few nucleotide changes are likely to exert a strong influence on F7 [Hahn et al., 2004] and PDYN [Rockman et al., 2005] loci
gene expression, irrespective of the overall level of sequence imply that positive selection can operate on regulatory mutations
divergence. In support of this view, the comparative analysis of that have occurred specifically in the human lineage. One example
16-bp blocks from promoter regions of 5,547 human-chimpanzee of this phenomenon is provided by the PDYN gene, which encodes
orthologous gene pairs has indicated that small sequence windows prodynorphin, a precursor molecule for a series of endogenous
have evolved under selective constraints and that purifying opioids and neuropeptides with critical roles in the anticipation
selection has indeed been operating within hominid noncoding and experience of pain perception, behavior including social
sequences [Bush and Lahn, 2005]. Further, human/chimpanzee/ attachment and bonding, as well as learning and memory [Rodgers
mouse comparisons of candidate transcription factor binding sites and Cooper, 1988; Wagner et al., 1993; Cheng et al., 2002; Moles
has revealed that the loss of such sites in the human lineage has et al., 2004]. A 68-bp tandem repeat polymorphism in the human
not simply been random and that these changes in regulatory PDYN gene promoter, 1,250 bp upstream from the transcriptional
motifs have affected the expression of specific functional categories start site, influences the inducibility of the gene [Zimprich et al.,
of genes, involving principally the olfactory system [Donaldson 2000]. During human evolution, a duplication of this 68-bp
and Gottgens, 2006]. regulatory element occurred generating four different genotypes
The unambiguous identification of relevant regulatory elements containing either one, two, three, or four copies of the repeat. By
could facilitate the design of functional assays that might then contrast, great apes and Old World monkeys have only one copy
be used to identify the molecular basis of expression divergence of the 68-bp element. Interestingly, this element has evolved
between humans and chimpanzees. In practice, however, the rapidly in the human lineage, as indicated by the five nucleotide
design of such assays has proved to be very difficult. Indeed, substitutions that occurred after the separation of humans and
comparative in vitro expression studies using promoter sequences chimpanzees, but prior to the human-specific duplication of the
from genes that are differentially expressed in these species suggest element. This number of nucleotide substitutions is somewhat
that there is no simple relationship between in vitro promoter higher than that expected for a neutrally evolving segment and
activity in cell lines and expression divergence in tissues in vivo suggests that the rapid evolution of the human 68-bp repeat
[Heissig et al., 2005]. It necessarily follows that expression occurred under positive selection [Rockman et al., 2005]. The
divergence will be difficult to predict by in vitro expression/ combined effect of these fixed cis-regulatory mutations would
DNA sequence analysis, probably as a consequence of the tissue- appear to be the upregulation of PDYN gene expression upon
and cell linespecificity of the transcription factors involved and induction by intracellular calcium release. An excess of high-
their potentially limited availability in vitro [Heissig et al., 2005]. frequency derived mutations in the flanking regions of the 68-bp
Just as gene expression diversity between individual humans is segments suggests that the entire region, including the regulatory
known to be influenced by multiple cis- and trans-acting factors element, evolved under positive selection. Further, the pattern of
[Morley et al., 2004; Stranger et al., 2005], so it is likely that microsatellite variation within and between modern human
multiple genetic differences will be found to influence expression populations implies that recent selection, subsequent to the
divergence between humans and chimpanzees. Particularly inter- fixation of the human-specific mutations, has favored different
esting in the context of the expression divergence between these PDYN cis-regulatory alleles in different parts of the world. Thus,
higher primates is the finding that transcription factors have selection, both ancient and recent, has served to highlight the
evolved rather rapidly in the human lineage by comparison significance of PDYN gene expression to human biology. Although
with other human proteins [Bustamante et al., 2005]. The the direct target of this selection is unclear, it appears to involve

Human Mutation DOI 10.1002/humu


110 HUMAN MUTATION 28(2), 99^130, 2007

changes in the inducibility of prodynorphin, an endogenous opioid the finding that alternatively spliced transcripts, conserved
precursor that is a prime candidate for involvement in conferring between human and mouse, have evolved under strong selection
human-specific traits due to its role in perception, emotion, [Modrek and Lee, 2003]. Further, it suggests that alternative
nociception, and learning [Rodgers and Cooper, 1988; Wagner splicing may have played a major role in genome evolution because
et al., 1993; Cheng et al., 2002; Moles et al., 2004; Balter, 2005]. new exons will have been able to evolve with fewer constraints.
In accordance with the predictions of the regulatory hypothesis Alternative splicing patterns observed in multiple species may
put forward by King and Wilson [1975], the evolutionary history be considered to be ancestral and such cases usually display
of the PDYN gene suggests that it may represent a good example of evidence of strong functional constraints. Frequently, these
a gene in which regulatory changes have played a significant role exons are modular in that their lengths are multiples of three
in human evolution. It remains to be seen how frequently nucleotides [Thanaraj et al., 2003; Resch et al., 2004] thereby
regulatory changes fixed in the human lineage have been adaptive, allowing their alternative inclusion or exclusion without disrupting
having evolved under selective pressure as opposed to being fixed the reading frame which would lead and leading to premature
simply as a consequence of stochastic processes. protein truncation.
Khaitovich et al. [2004, 2005b] have presented a model, which When human and chimpanzee exon flanking regions were
proposes that the majority of expression differences observed compared, a significant reduction in the nucleotide substitution
between species are selectively neutral and lack any functional rate of the flanking regions of alternatively spliced exons was
significance. Consistent with this postulate, only a very small observed, independent of the site-by-site variation in mutability
proportion of the gene expression changes in human relative to due to different CpG contexts. Thus, increased purifying selection
chimpanzee appear to be due to diversifying selection [Hsieh et al., appears to have impacted on exon flanking regions and thus, in all
2003]. However, such a neutralist model would appear to be at likelihood, the regulation of alternative splicing in humans and
odds with: 1) the upregulation of gene expression that is specific chimpanzees [Xing et al., 2006].
to the human brain and not apparent either in the chimpanzee A rather specific type of alternative splicing, the differential
brain or in the liver of humans and chimpanzees [Gu and exclusion of exons due to pseudoexonization, may also have
Gu, 2003; Gilad et al., 2006]; and 2) the rapid evolution of contributed to the genetic divergence between humans and
primate transcription factors [Gilad et al., 2006]. Among possible chimpanzees. An example of this phenomenon is provided by
adaptive scenarios, it may be that the general level of physiological the glycophorin B (GYPB) gene, in which exon 3 is expressed
activity in the brain, and in particular the cerebral cortex, is higher in chimpanzees but silenced as a pseudoexon in humans [Huang
in humans than in chimpanzee with the higher levels of gene et al., 1995].
expression in humans being required to meet the correspondingly
higher requirements for brain proteins. Clearly, further studies are
CHROMOSOMAL DIFFERENCES AND VARIATION
needed to explore to what extent the higher gene expression levels
ACROSS CHROMOSOMES
characteristic of the human brain are reflected in the levels of their
protein products [Preuss et al., 2004]. Molecular cytogenetic analyses of hominoids have indicated that
humans and orangutans have retained karyotypes closer
to the putative ancestral hominoid karyotype than have the African
ALTERNATIVE SPLICING
great apes (chimpanzee and gorilla), which display more lineage-
Alternative splicing contributes very significantly to increased specific derived structural rearrangements [Dutrillaux, 1979; Muller
transcriptome and proteome diversity as well as to interspecies and Wienberg, 2001]. The chimpanzee and human genomes may
divergence [Boue et al., 2003]. be distinguished by 10 karyotypic differences that include nine
An estimated 60% of all human genes employ alternative pericentric inversions and one fusion that gave rise to human
splicing [Mironov et al., 1999; Lander et al., 2001]. Dual coding chromosome 2 [Yunis and Prakash, 1982; Gross et al., 2006]. Most
regions in particular are characterized by alternative splicing; the of these rearrangements have now been fully characterized by
same exon sequences are shared by different transcripts encoding analysis of the breakpoint regions; in none of these cases, however,
distinct amino acid sequences in different reading frames. At least has a gene been interrupted by the breakpoints [IJdo et al., 1991;
7% of all alternatively spliced genes in the human genome contain Nickerson and Nelson, 1998; Marzella et al., 2000; Fan et al., 2002;
multiple coding regions [Liang and Landweber, 2006]. Compara- Kehrer-Sawatzki et al., 2002, 2005a,b,c; Locke et al., 2003a;
tive analysis of orthologous sequences in the chimpanzee, mouse, Dennehey et al., 2004; Goidts et al., 2004, 2005; Shimada et al.,
rat, dog, and chicken genomes has indicated that most secondary 2005; Szamalek et al., 2005, 2006a]. Segmental duplications were
reading frames in dual coding regions have originated during identified in the breakpoint regions of six of the nine pericentric
mammalian evolution. Interestingly, 4% of human dual coding inversions, suggesting that these duplicates could have mediated or
regions are not conserved in chimpanzees, as indicated by the at least facilitated the inversions. Interestingly, human-specific
presence of stop codons that disrupt one of the two reading frames segmental duplications were identified at the breakpoints of both
[Liang and Landweber, 2006]. the chromosome 1 and 18 inversions, hinting that these sequence
Dual coding genes can be grouped into two categories: if the acquisitions could have rendered the respective breakpoint regions
dual reading frame is intact, it may be regarded as the ancestral susceptible to homologous recombination [Goidts et al., 2005;
reading frame (ARF), whereas the presence of a stop codon in Szamalek et al., 2006a].
one of the two reading frames defines it as the derived Seven of the large pericentric inversions appear to have been
reading frame (DRF). Comparative analysis between human fixed in the chimpanzee lineage (Supplementary Table S3) and
and chimpanzee has revealed that the reading frames associated comparative breakpoint analyses have shown that the two
with the original splicing patterns (ARFs) have acquired sister species, the common chimpanzee (Pan troglodytes) and the
considerably fewer substitutions than the derived ones, suggesting pygmy chimpanzee (Pan paniscus) share the same pericentric
that ARFs have been maintained by stronger selection pressure inversions [Locke et al., 2003a; Szamalek et al., 2006b]. Thus,
[Liang and Landweber, 2006]. This observation is in accord with these inversions must have predated the separation of the two

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 111

chimpanzee species 0.862 Mya [Yoder and Yang, 2000; Won Alu Sequence Insertions/Deletions
and Hey, 2005]. It is as yet unclear whether any of these
With up to 106 copies in the human genome, Alu sequences
rearrangements played a role during the process of speciation of
constitute the most frequently encountered type of short
humans and chimpanzees. According to the recombination
interspersed repeat element (SINEs). Alu mobilization has, how-
suppression model, these chromosomal rearrangements, in their
ever, proceeded at different rates in the human and chimpanzee
heterozygous state, could have acted as a partial barrier to gene
lineages. Thus, comparison of chimpanzee chromosome 22 and its
flow between early populations of emerging humans and
human homolog, chromosome 21, revealed a two-fold greater
chimpanzees at a time when interbreeding was still possible
frequency of Alu insertions in humans as compared to the
[Navarro and Barton, 2003; Rieseberg and Livingstone, 2003].
chimpanzee. Moreover, the comparison of human and chimpanzee
To test this hypothesis, Navarro and Barton [2003] analyzed 115
Y chromosomes has revealed higher Alu element retrotransposi-
genes and observed that the DNA sequence divergence on
tional activity in the human lineage [Hughes et al., 2005].
rearranged chromosomes was slightly higher than on colinear
However, by analogy with the higher rate of nucleotide sequence
chromosomes. Furthermore, the rate of protein evolution on
diversity in chimpanzees, the Alu sequence diversity (i.e., presence
rearranged chromosomes as measured by KA/KS ratios was twice
or absence of Alu elements at specific genomic locations within a
that observed on colinear chromosomes [Navarro and Barton,
given species) is 1.7 times higher in the chimpanzee as compared
2003]. These findings were therefore consistent with the view that
to human [Hedges et al., 2004].
large chromosomal rearrangements could have led to reduced gene
An expansion in the overall size of the human genome due to a
flow and accelerated adaptive evolution. However, reexamination
higher rate of Alu retrotranspositional activity in the human
of this hypothesis using larger datasets did not confirm these
lineage has been detected by Liu et al. [2003]. They compared
findings and failed to provide support for the hypothesis that gross
10.6 Mb of genomic sequence from lemur, baboon, chimpanzee,
chromosomal rearrangements affected the rate of genetic diver-
and human and noted a marked paucity of chimpanzee Alu inser-
gence between humans and chimpanzees [Lu et al., 2003; Zhang
tions as compared to human. Since the separation of chimpanzees
et al., 2004a; Vallender and Lahn, 2004b].
and humans from their common ancestor 5 to 7 Mya, 7000 Alu
During the course of their comprehensive comparative analysis,
elements have been fixed in the human genome including the
the CSAC also investigated the variation in protein divergence
human-specific subfamilies AluYa5 and AluYb8 [Carroll et al.,
across different chromosomes [Mikkelsen et al., 2005]. This re-
2001; Carter et al., 2004; Gibbons et al., 2004; Otieno et al., 2004;
examination did not, however, reveal any evidence for the
Mikkelsen et al., 2005; Mills et al., 2006]. Genomewide
accelerated evolution of gene products encoded by genes on
comparisons have indicated that the number of Alu insertions in
chromosomes with major rearrangements. Nevertheless, the large
humans has been 3.4-fold higher than in the chimpanzee and
inversions appear to be associated with an increase in interspecies
that the distributions of these elements among various Alu
differences in brain gene expression when genes on rearranged vs.
subfamilies also differed between the two species [Mills et al.,
colinear chromosomes are compared. Thus, the rearrangements
2006] (Supplementary Fig. S2). AluYa5, AluYb8, AluY, and
may have had indirect effects on the expression of linked genes
AluYc1 are highly abundant in humans, whereas only AluYc1
[Marques-Bonet et al., 2004].
and AluY are abundant in chimpanzees [Mills et al., 2006]. Clearly,
significantly more Alu retrotransposition has occurred in the
human lineage and a different set of Alu elements has been
DYNAMICS OF STRUCTURAL amplified in humans as compared to chimpanzees (Fig. 2).
DIFFERENCES BETWEEN THE HUMAN
AND CHIMPANZEE GENOMES
Insertions/Deletions 10000000

1000000
Recent progress in comparing the human and chimpanzee
Number of rearrangements

genomes has revealed that the structural divergence between 100000

them is not confined to gross chromosomal rearrangements but 10000


also includes a considerable number of submicroscopic structural 1000
differences [Britten, 2002; Britten et al., 2003; Frazer et al.,
100
2003; Liu et al., 2003; Fortna et al., 2004; Newman et al.,
10
2005; Mikkelsen et al., 2005; Kuroki et al., 2006]. These
include insertions and deletions that have considerably altered 1

the landscape of the human and chimpanzee genomes


after the separation of both lineages. Indeed, insertions of modest
f

nf
c

bd

se
ab

ns
kb

0k

o
0k

n
ns

io

si
10

size (1 bp to 15 kb) have given rise to a total of 32 Mb of


io

10
10

rs

Fu
tio

at
<

ve
>
<

lic
u

ns
tit

in
el

ns

human-specific DNA sequence by comparison with the chimpan-


up
d
bs

rio

ric
io
In

/d
su

er

ve

t
ns

en

zee genome [Mikkelsen et al., 2005] and a comparable figure for


nv

in
e

tio

ric
id

oi

ro
le
t

Pe
r

ic
eo

chimpanzee-specific sequences may reasonably be assumed.


ic

De

M
M
cl
Nu

The genomewide comparison performed by the CSAC has


Length of the rearrangement
provided comprehensive information about the nature and
frequency of lineage-specific insertions and deletions. Most inser- FIGURE 1. Graphical representation of the genetic and cytoge-
tions/deletions are small, with 96% being o20 bp and 98.6% being netic dierences between the human and chimpanzee genomes.
o80 bp (Fig. 1). Approximately 1.4% of all modestly-sized Rearrangements are ordered by size (x-axis). The number of
rearrangements is represented on a logarithmic scale (y-axis).
insertions/deletions are larger than 80 bp; these involve a variety a
Mikkelsen et al. [2005]; bChen and Li [2001]; cBritten [2002];
of different sequences but the majority comprise satellites, micro- d
Feuk et al. [2005]; eCheng et al. [2005]; f Yunis and Prakash
satellites, high copy number repeats, and transposons. [1982].

Human Mutation DOI 10.1002/humu


112 HUMAN MUTATION 28(2), 99^130, 2007

2,000 lineage-specific L1 elements [Mikkelsen et al., 2005]. This


contrasts with earlier assertions of a higher rate of L1 insertion in
the chimpanzee lineage [Mathews et al., 2003; Mills et al., 2006].
However, chromosome-specific differences may be assumed, since
on the chimpanzee Y chromosome, insertions of L1 elements and
endogenous retroviruses have been significantly more frequent
than on the human Y chromosome [Hughes et al., 2005].
Finally, L1 insertions into the human genome are often
accompanied by target site deletions. Han et al. [2005] have
characterized 50 such L1 insertion-mediated deletions in the
human and chimpanzee genomes resulting in the loss of a total of
18 kb from the human genome and 15 kb from the chimpanzee
FIGURE 2. Number of transposon insertions within genes, in
human and chimpanzee genomes, according to Mills et al. [2006].
genome.
The majority of these insertions areAlu, L1, and SVA elements.
SVA Elements
The youngest family of primate retrotransposons that arose after
In contrast to the expansion of repetitive sequences such as the separation of Old World monkeys from the hominoid ancestral
retroelements, the deletion of these sequences has also been lineage are the SVA elements, termed according to their main
observed in a lineage-specific manner. An estimated 0.5 to 1% components: a SINE-R sequence derived from an LTR element of
of the apparent retroelement insertions that distinguish humans the human endogenous retrovirus K (HERV-K) HERV-K10 [Ono
from chimpanzees actually represent deletions in one or other et al., 1987], VNTR, a central variable number tandem repeat
genome; this particularly affects members of the older Alu sub- that is rich in CpG sequences, and a partial Alu-sequence in
families, such as AluS and AluJ, which are common to both reverse orientation [Shen et al., 1994b]. SVA elements end with a
humans and chimpanzees. These deletions have probably been poly (A) tail and are flanked by target site duplications similar to
mediated by recombination between 10 to 20 bp direct repeats those that flank Alu and L1 elements [Ostertag et al., 2003;
flanking the retroelements [van de Lagemaat et al., 2005]. Bennett et al., 2004]. SVA elements are frequently polymorphic
Recombination between Alu elements is considered to be the among humans [Bennett et al., 2004] and some instances have
mechanism underlying the 492 human-specific deletions between been reported of SVA insertions causing disease [Kobayashi et al.,
101 and 7,255 bp. These deletions also affected exons and resulted 1998; Ostertag et al., 2003].
in the loss of 400 kb from the human genome as compared to the Six SVA subfamilies have been identified and exhibit consider-
chimpanzee [Sen et al., 2006]. Thus, Alu-mediated deletions have able lineage-specific activity in humans and chimpanzees, sprout-
counteracted, at least in part, the increase in the size of the human ing two human-specific subfamilies [Wang et al., 2005]. Direct
genome due to Alu insertions. Both processes have clearly genome comparisons have revealed 1,000 SVA copies specific
contributed to the evolution of the human genome. to the human lineage and a comparable number of chimpanzee
lineage-specific integrations [Mikkelsen et al., 2005]. Full-length
LINE Element Insertions SVA elements have a high G1C content (about 60%) and
contain transcription factor SP1 binding sites. This confers upon
Long interspersed elements (LINE-1 or L1) comprise 17%
SVAs the potential to mobilize CpG islands and hence their
of the human genome sequence and are the most abundant
insertion could influence the expression of flanking genes.
and the only active autonomous non-long terminal repeat (LTR)
Mikkelsen et al. [2005] identified human-specific SVA inser-
retrotransposons in the human genome. However, of 4500,000
tions near the promoters of three genes: MRGX3 (G protein-
L1 copies, only 80 to 100 are capable of active retrotransposition
coupled receptor MRGX3), SLC2A11 (glucose transporter protein
[Brouha et al., 2003].
10), and SULT1B1 (sulfotransferase family cytosolic 1B). Detailed
Some L1 retrotransposons have been specifically amplified since
analyses are, however, necessary to confirm or exclude effects of
the origin of the primates [Salem et al., 2003; Vincent et al., 2003;
the SVA integration with respect to the expression of the
Ho et al., 2005]. Two phases of L1 expansion may be discerned,
respective genes.
during which different L1 families experienced differing degrees of
Remarkably, a large number of lineage-specific transposon
replicative success. The most intense period of L1 activity involved
insertions have occurred within genes (2,642 in humans and
families L1PA8L1PA3 and lasted from 40 to 12 Mya [Khan
990 in chimpanzees; Fig. 2). In view of the extent of these
et al., 2006]. Their expansion was associated with the amplifica-
insertional events, it is very likely that at least some of them have
tion of AluY elements and many processed pseudogenes [Batzer
influenced gene expression [Mills et al., 2006]. Since the number
and Deininger, 2002; Ohshima et al., 2003]. Approximately 2,000
of transposon insertions has been higher in the human lineage, we
L1 elements have become inserted specifically into the human
surmise that the impact of transposon-mediated expression
genome since human-chimpanzee divergence, a process which
differences is likely to have been more pronounced in humans.
may have added as much as 1.3 Mb of DNA to the human genome
over the last 5 Myrs [Han et al., 2005; Mikkelsen et al., 2005].
Endogenous Retroviral Elements
This expansion may in part account for the larger genome sizes
of anthropoid primates (e.g., monkeys, apes, and human) as compared Endogenous retroviruses and LTRs comprise 8% of the human
with the prosimians (e.g., lemurs and galagos) [Liu et al., 2003]. genome. They arose early during primate evolution by infection
These L1 expansions, however, occurred prior to the separation of and integration of exogenous source viruses and were subsequently
hominoids. Unlike the human lineage-specific Alu expansion, the amplified by retrotransposition of preexisting endogenous
rate of L1 element retrotranspositional activity has been relatively retroviruses. The HERV-K subfamily is the most conserved
similar in the genomes of human and chimpanzee; they both possess evolutionarily and is capable of forming virus-like particles [Costas,

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 113

2001; reviewed in Bannert and Kurth, 2004]. About 30 to 50 deletions and other rearrangements (summarized in Bacolla and
HERV-K proviruses are found in the human genome; the youngest Wells [2004]; Bacolla et al. [2004, 2006]). Detailed comparison of
known HERV-K provirus, HERV-K113, has an estimated age of long RY tracts in human and mouse as well as in the human and
o200,000 years [Turner et al., 2001]. Divergent patterns of recent chimpanzee genomes have indicated that they have mutated
retroviral integrations into the human and chimpanzee genomes much faster than their flanking sequences due to mutational
have been observed [Buzdin et al., 2003; Jern et al., 2006; mechanisms such as replication slippage and recombination as well
Polavarapu et al., 2006]. Comparing the human and chimpanzee as nucleotide substitution. Furthermore, 80% of the pure RY tracts
genomes, 45 chimpanzee-specific insertions (one full-length 4250 bp are longer in humans than in chimpanzees, while the
and 44 solo LTRs) and 73 human-specific insertions (seven full- length divergence of RY tracts in the human as compared to the
length and 66 LTRs) have been identified corresponding to chimpanzee genome is significantly higher than the average rate of
lineage-specific retrotranspositional events [Mikkelsen et al., nucleotide substitution in both genomes [Bacolla et al., 2006].
2005; Romano et al., 2006]. Finally, a genomewide survey of
endogenous retroviral positional variation between humans and
Gross Insertions and Duplications
chimpanzees has shown that 7% of all human-chimpanzee
insertion/deletion variation is associated with endogenous retro- Human/chimpanzee genome comparisons have also revealed a
viral sequences [Polavarapu et al., 2006]. considerable number of human-specific insertions of larger size
These insertions could have influenced the expression of nearby (415 kb). These frequently contain genes (or partial gene
genes by virtue of the presence of their internal polyadenylation sequences) and have given rise to 8 Mb of human-specific
signals and promoter and enhancer sequences, as well as cryptic sequences and probably a similar amount of chimpanzee-specific
splice sites. Indeed, Yohn et al. [2005] have characterized the sequence [Mikkelsen et al., 2005]. Taken together, some 40 to
insertion sites of a PTERV1 retroviral element, sequences which 45 Mb of lineage-specific sequence results from insertions/
are specific to the African great apes including the chimpanzee but deletions. Thus, a total of 90 Mb, the sum of all lineage-specific
that are not found in human. Of 10 chimpanzee genes found to sequences, contribute to the divergence evident between the
harbor intronic PTERV1 elements, six exhibit significant differ- human and chimpanzee genomes [Mikkelsen et al., 2005].
ences in gene expression by comparison with human (in five of The total sequence divergence with respect to the number of
these, gene expression is lower in the chimpanzee). nucleotides affected by deletions and insertions is thus some three-
fold higher than the divergence due to nucleotide substitutions.
However, the number of underlying mutational events is clearly
Microsatellite and Triplet Repeat Expansion
much lower for insertions/deletions than for nucleotide substitu-
The tendency of the human genome to expand in size is also tions (Fig. 1).
reflected in microsatellite dynamics. Microsatellites are iterations It may be that the number of lineage-specific insertions/deletions
of short (16 bp) sequence motifs, with the length of the repeats detected by the CSAC using direct genome alignments was
being generally less than 30 bp. Comparative analyses of micro- underestimated owing to the draft version status of the chimpanzee
satellite length in humans and chimpanzees has revealed that genome. An alternative approach to investigating the extent of
human microsatellites are longer than their orthologous chimpan- structural divergence between the human and chimpanzee genomes
zee equivalents, even when the effects of ascertainment bias due to has involved the mapping of fosmid end-pairs of a chimpanzee
microsatellite length are allowed for [Cooper et al., 1998; Vowles library against the human reference sequence. By these means, a
and Amos, 2006]. This suggests that expansion mutations have total of 477 insertions/deletions 412 kb were identified [Newman
occurred disproportionately in the human lineage. Microsatellite et al., 2005]. These structural variants encompass a total of
evolution has, however, turned out to be quite heterogeneous. 424 Mb DNA and include more than 200 genes whose structure
Human dinucleotide repeats tend to be longer than their orthologs and/or expression could consequently be perturbed.
in chimpanzees, whereas the opposite trend is apparent in In addition to direct sequence comparisons, new methodologies
mononucleotide repeat arrays. Furthermore, the rate of divergence such as array comparative genomic hybridization (aCGH) using
between orthologous microsatellites is significantly higher at longer genomic clones have proved to be capable of detecting copy-
loci, which also display evidence of higher mutability per repeat number differences (CNDs) of 40 kb up to several Mb between the
number. Thus, locus-specific mutation rate variability between human and chimpanzee genomes [Locke et al., 2003b; Goidts
species suggests that microsatellite evolution is a highly dynamic et al., 2006a; Wilson et al., 2006a]. These CNDs represent
process [Webster et al., 2002]. deletions and duplications, also termed intermediate sized
Remarkably, triplet repeats within coding regions also seem to rearrangements since they are not visible by G-banding analysis
be longer in humans than in chimpanzees as observed for GAA of chromosomes in contrast to the cytogenetically visible gross
triplet repeat-containing loci [Clark et al., 2006], cancer-related rearrangements such as the nine pericentric inversions and the
genes [Puente et al., 2006], and genes associated with neurode- fusion that gave rise to human chromosome 2. The majority of
generative disorders [Choong et al., 1998; Andres et al., 2004b]. these CNDs map to regions of segmental duplication, indicating
With respect to the latter class of genes, not only do humans that this fraction of the genome is a major source of the variability
possess larger alleles as compared with the great apes, but they also evident between humans and chimpanzees.
display greater length variability, a finding that is suggestive of a A genomewide comparison of segmental duplications between
relationship between the level of variability and expansion the two primate species has been performed by Cheng et al.
potential [Andres et al., 2004b]. [2005]; although 66% of the autosomal sequence duplicated
Poly-purine/pyrimidine sequences (RY tracts) are able to form in humans was also found to be duplicated in the chimpanzee,
triplex DNA structures that can block DNA replication and a considerable fraction (33%; encompassing 26.5 Mb) was
interfere with transcription. Such non-B DNA conformations are duplicated only in the human lineage [Cheng et al., 2005]. These
highly mutagenic due to their propensity to induce double strand human-specific duplication intervals map to at least 500 distinct
breaks, and are frequently found at the breakpoints of gross regions (of average length 55 kb) distributed throughout the

Human Mutation DOI 10.1002/humu


114 HUMAN MUTATION 28(2), 99^130, 2007

genome. By contrast, only 17% (11.4 Mb) of the duplicated hyperexpanded sequences has indicated the presence of a single
sequences in the chimpanzee are chimpanzee-specific in that they copy of a 36-kb segmental duplication and a 14.5-kb cluster of the
are not duplicated in the human genome [Cheng et al., 2005]. aforementioned 32-bp repeats [Cheng et al., 2005]. Subtelomeric
Segmental duplications have thus shaped the architecture of the regions of chimpanzee chromosomes would thus appear to have
human genome [Eichler et al., 1996; Trask et al., 1998; Bailey been susceptible to duplicative transposition, expansion of repeats,
et al., 2002a,b; Armengol et al., 2003; Cheung et al., 2003; and segmental duplications, leading to an asymmetrical increase in
Bailey et al., 2004; She et al., 2004; Stankiewicz et al., 2004; duplicated DNA in the chimpanzee lineage as compared to
Zhang et al., 2005b; Bailey and Eichler, 2006] and comparison humans [Cheng et al., 2005].
with the chimpanzee genome indicates that this has occurred in a Comparative analyses have revealed further significant differ-
lineage-specific manner [Cheng et al., 2005; Goidts et al., 2006a; ences between the human and chimpanzee genomes caused by
Wilson et al., 2006a; Wooding and Jorde, 2006]. In view of the copy number differences in subtelomeric segments [Trask et al.,
enrichment of genes within segmental duplications, this is likely to 1998; Monfouilloux et al., 1998; Martin et al., 2002]. Human
have had quite important implications for the emergence of new subtelomeric regions were massively rearranged after divergence
genes and gene expression differences between the species. from the chimpanzee lineage with at least 49% (1.13 Mb) of
human subtelomeric sequences being generated after the split
of both lineages [Linardopoulou et al., 2005]. Repeated trans-
SEGMENTAL DUPLICATIONS AND STRUCTURAL
locations between chromosomal ends then gave rise to complex
DIFFERENCES IN SUBTELOMERIC REGIONS
segmental duplications and at least one-half of the known subtelo-
BETWEEN HUMANS AND CHIMPANZEES
meric sequence in humans has been generated by interchromo-
Segmental duplications (also termed low copy repeats [LCRs]) somal sequence transfer between segmental duplications after the
are implicated in the genesis of disease-associated rearrangements divergence of humans and chimpanzees [Linardopoulou et al.,
owing to their inherent propensity to nonallelic homologous 2005]. Subtelomeric, pericentromeric, and centric regions of
recombination, the molecular basis of a wide range of so called chromosomes constituted preferred acceptor sites of duplica-
genomic disorders. The latter are caused by rearrangements tive transposition during the evolution of the human and
in genomic regions with complex architecture [reviewed in chimpanzee genomes, although to different degrees and with
Lupski, 1998; Ji et al., 2000; Shaw and Lupski, 2004; Lupski differing preferences of genomic locations [Fortna et al., 2004;
and Stankiewicz, 2005]. Analysis of the human genome assembly Cheng et al., 2005; Mikkelsen et al., 2005].
version 15 revealed that 4% of the human genome is covered by
sequence duplications and the extent of segmental duplication
EVOLUTION OF HUMAN CENTROMERES
varies from 1 to 14% between the 24 chromosomes.
Intrachromosomal duplications are more frequent than inter- The centromeres of human chromosomes are extremely
chromosomal duplications (Supplementary Fig. S3; Zhang et al. specialized loci that are critically important for orderly chromo-
[2005b]) and a peak of intrachromosomal duplication activity some segregation. Their conserved function contrasts, however,
appears to have occurred in the ancestral hominoid genome with the rapid evolution of their component DNA sequences
10 Mya. By contrast, interchromosomal duplication dispersal [reviewed by Henikoff et al., 2001; Henikoff and Dalal, 2005].
peaked somewhat earlier, during or after the separation of the Normal human centromeres contain huge amounts of a-satellite
Old World monkeys from the hominoid lineage 25 Mya [She sequence associated with centromere function and kinetochore
et al., 2006]. Segmental duplications are enriched in pericen- assembly [Schueler et al., 2001; Ikeno et al., 1998; Grimes et al.,
tromeric and subtelomeric regions of human chromosomes [Bailey 2002; Spence et al., 2002; Porter and Farr, 2004] and specific
et al., 2002a; Zhang et al., 2005b]. The dynamics of duplicative to the primate lineage [Willard, 1990; Alves et al., 1994]. These
transposition are significantly more prominent in these regions by sequences exist in two different forms, the higher-order and the
comparison with the genome average [Mefford and Trask, 2002; monomeric a-satellites, which evolved by complex patterns of
She et al., 2004; Linardopoulou et al., 2005]. One consequence of inter- and intrachromosomal exchanges [Warburton and Willard,
this is the extensive polymorphism present in human subtelomeric 1996; Alexandrov et al., 2001; Rudd and Willard, 2004]. Higher-
regions [Wilkie et al., 1991; Trask et al., 1998; Daniels et al., order a-satellite DNA comprises homogeneous 3- to 5-Mb
2001], which possess a high concentration of genes [Flint et al., arrays of repeat units in tandem orientation [Wevrick and Willard,
1997; Saccone et al., 1992]. 1989; Lee et al., 1997; Mahtani and Willard, 1998]. Monomeric
A considerable number of structural differences are evident a-satellites, however, lack detectable higher-order periodicity, and
between the subtelomeric regions of human and chimpanzee the monomers are more heterogeneous than the higher-order
chromosomes using classical cytogenetic techniques. In contrast to repeat units [Rudd and Willard, 2004]. In normal human
humans, chimpanzees and gorillas possess remarkable amounts of centromeres, monomeric a-satellites flank the large array of
additional positive G-banded chromatin at the chromosomal ends higher-order a-satellites [Horvath et al., 2000; Schueler et al.,
[Yunis and Prakash, 1982]. These telomeric caps contain arrays 2001; Guy et al., 2003; Rudd and Willard, 2004]. This
of a 32-bp AT-rich repeat sequence that is abundant within the organization within centromeres is also found in the great apes,
telomeric regions of the African great apes but absent from the whereas lower primates only have monomeric a-satellites at their
human genome [Royle et al., 1994]. In these subtelomeric regions centromeres [Thayer et al., 1981; Alves et al., 1994]. Human and
of chimpanzee chromosomes, massive expansions of one specific chimpanzee alphoid sequences underwent remarkably rapid
segmental duplication occurred, leading to the generation of an evolution in both species, mediated by massive interchromosomal
additional 16 Mb of genomic DNA that is not present in humans exchange and divergent intrachromosomal expansion leading to
[Cheng et al., 2005]. In addition to the subterminal portions of the generation of species-specific a-satellite DNA [Archidiacono
chimpanzee chromosomes, the hyperexpansion of these sequences et al., 1995; Warburton et al., 1996; Haaf and Willard, 1997].
also occurred in interstitial regions of chromosomes VII and XIII. Comparative analyses between humans and chimpanzees have
Sequence analysis of one chimpanzee locus corresponding to these revealed that monomeric a-satellites have diverged less rapidly

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 115

than higher-order a-satellites [Rudd et al., 2006]. These findings least 180 genes and partial genes have become duplicated
are remarkable since higher-order a-satellites are associated with specifically in the human genome, whereas at least 94 genes have
centromere function [Harrington et al., 1997; Ikeno et al., 1998; been duplicated specifically in the chimpanzee lineage. These
Schueler et al., 2001; Spence et al., 2002] and might therefore be lineage-specific segmental duplications serve as a reservoir of
expected to be subject to purifying selection. However, the more duplicated genes and it may be that some of these genes retained
rapid evolution of the higher-order a-satellites could have been their function and diverged such that they now confer hominid-
accompanied by rapidly coevolving centromere-associated pro- specific traits [Eichler, 2001; Bailey et al., 2002a,b; Samonte and
teins. In support of this postulate is the rapid evolution of CENPC Eichler, 2002; Courseaux et al., 2003].
proteins in plants and animals [Talbert et al., 2004]. Despite the The potential of duplicated genes to evolve new functions by
obvious functional significance of human centromeres, their highly sequence divergence was first mooted by Ohno [1970]. Released
repetitive sequences have greatly hampered their inclusion in the from the functional constraints that operated upon their
genome assembly [Eichler et al., 2004]. Only some chromosome parental source genes, duplicated gene copies are in principle free
assemblies (including human chromosomes 2, 4, 6, 7, 8, 10, 11, 17, to undergo neo-, sub-, or microfunctionalization [Hurles,
18, X, and Y) contain considerable amounts of higher-order 2004; Hancock, 2005]. Whereas neofunctionalization represents
a-satellite DNA [Rudd and Willard, 2004], and as yet none of the the acquisition of a new function, subfunctionalization entails
human centromeres have been completely sequenced. Further, as the adoption of part of the function of the parental gene
yet, none of the chimpanzee chromosome assemblies in the draft by the duplicate copy [Hurles, 2004]. Microfunctionalization
sequence have reached higher-order a-satellites. More is known, describes the process by which duplicated genes come to encode
however, about the sequence structure and evolution of the similar proteins with subtly different functions, such as olfactory
centromeric transition zone between the pericentromeric region receptors [Hancock, 2005]. These processes are frequently
and the higher-order a-satellite arrays [Horvath et al., 2000, 2005; accompanied by divergence of spatial expression patterns between
She et al., 2004; Schueler et al., 2005]. For human chromosome 8, human (and more generally eukaryotic) duplicated genes [Makova
a complete sequence contig is available that spans the entire and Li, 2003; Li et al., 2005]. However, most of the duplicates
pericentromeric region [Nusbaum et al., 2006]. Especially well- become degraded [Nadeau and Sankoff, 1997; Li et al., 2001]; this
characterized in this respect is the pericentromeric region of the has consequently been termed a birth and death process [Nei
human X chromosome. An age gradient is observed, with the et al., 1997; Nei and Rooney, 2005]. Some of the duplicated genes
most distal a-satellites in the transition zones being ancient, nevertheless adopt divergent expression profiles [Kim et al., 1989;
whereas more recently amplified repeats are located in the central Gu et al., 1997; TomHon et al., 1997] and acquire novel functions
functional region. This pattern has also been observed in the while evolving under different selective constraints and pressures
chimpanzee and other primates, implying that the primate X than the parental genes [Lynch and Conery, 2000; Prince and
centromere evolved through repeated expansion extending from Pickett, 2002; Kouprina et al., 2004b; Birtle et al., 2005; Yu et al.,
the central active region of centromeric a-satellites [Schueler 2006b; Svensson et al., 2006].
et al., 2001, 2005]. Several primate-specific genes and gene families are thought to
Comparative analyses of the pericentromeric regions in humans have arisen in this way, for example the melanin-concentrating
and chimpanzees, as well as in other primates, have revealed hormone-like genes [Courseaux and Nahon, 2001], the morpheus
insights into their extraordinary dynamism, characterized by gene family [Johnson et al., 2001], the chorionic gonadotropin
extensive duplication, deletion, and rearrangement of large genes [Maston and Ruvolo, 2002], the SPANX genes of cancer/
segments of DNA [Eichler et al., 1996, 1997; Regnier et al., testis-specific antigens [Kouprina et al., 2004b], the NBPF gene
1997; Zimonjic et al., 1997; Orti et al., 1998; Jackson et al., 1999; family [Vandepoele et al., 2005], and many others [Samuelson
Bailey et al., 2001, 2002b; Horvath et al., 2000, 2003; Schueler et al., 1990; reviewed in Gagneux and Varki, 2001; Zhang et al.,
et al., 2001; Crosier et al., 2002; She et al., 2004]. The 2002a; Paulding et al., 2003]. The widespread existence of gene
pericentromeric regions of human chromosomes have been formed families itself provides confirmation that gene duplication and
via extensive duplications that have colonized pericentromeric divergence has been of very considerable importance with respect
DNA (pericentromeric seeding). Secondary duplications of to the evolution of new biological functions. Quite frequently,
larger mosaic blocks (pericentromeric swapping) occurred members of gene families with multiple paralogs are highly
subsequent to the seeding events, giving rise to a differential rearranged in comparison with the source locus. These rearrange-
distribution of these blocks among the great ape and human ments are mostly associated with domain or exon accretion, a
pericentromeric regions [Jackson et al., 1999; Guy et al., 2000, process that allows avoidance of potentially deleterious negative
2003; Horvath et al., 2000; 2005; Locke et al., 2005]. The global gene dosage effects that might occur if two highly homologous
comparison of differences in the segmental duplication content of duplicate genes are maintained in the genome [reviewed by
human and chimpanzee genomes indicates that the human Nahon, 2003; Babcock et al., 2003; Ciccarelli et al., 2005]. It can
genome has experienced more pericentromeric duplications than be seen that the marked instability of segmental duplications
the chimpanzee genome, suggesting that pericentromeric duplica- characterized by extensive genomic remodeling during
tive expansion has been more pronounced in the human lineage hominoid evolution has facilitated the generation of great
[Cheng et al., 2005]. apespecific and human-specific chimeric genes [Eichler, 2001;
Nahon, 2003]. Furthermore, rearrangements in gene order during
the creation and/or the expansion of LCRs represent an important
GENE DUPLICATION AND OTHER
mechanism of genome evolution, a phenomenon exemplified
MECHANISMS RESPONSIBLE FOR CREATING
by the DPY19L2 gene, which is located within LCRs [Carson
HUMAN-SPECIFIC GENES
et al., 2006].
Gene Duplications
A genomewide survey of gene duplication across the great apes
As a consequence of the above-mentioned lineage-specific and humans has been performed using cDNA arrays covering
segmental duplications, Cheng et al. [2005] demonstrated that at 29,619 different genes [Fortna et al., 2004]. Interestingly, humans

Human Mutation DOI 10.1002/humu


116 HUMAN MUTATION 28(2), 99^130, 2007

were found to possess a larger number of lineage-specific gene Pavlicek et al., 2006]. Approximately 70% of the 20,000 human
duplications than the great apes. A total of 134 gene families were pseudogenes result from retroposition rather than genomic
identified that exhibited human lineage-specific gains, including duplication [Goncalves et al., 2000; Torrents et al., 2003].
a number of genes with possible neuronal functions. Quite Processed pseudogenes are usually intronless, lack regulatory
remarkable is the positional bias of these gains since many were elements, and are frequently inactivated by microdeletions or
found in the pericentromeric regions of chromosomes 1 and 9 premature stop codons. There is, however, increasing evidence
[Fortna et al., 2004]. Importantly, the great majority of these copy that at least a proportion of them are functional and that
number gains between humans and chimpanzees as detected retroposition constitutes an important mechanism for the genera-
by cDNA aCGH [Fortna et al., 2004] were also found in studies tion of new genes [reviewed in Brosius, 1999; Korneev et al., 1999;
using aCGH with genomic clones (BAC aCGH) [Cheng et al., reviewed by Betran and Long, 2002; Betran et al., 2002; Long
2005; Wilson et al., 2006a; Goidts et al., 2006a]. These array- et al., 2003; Hirotsune et al., 2003; Strichman-Almashanu et al.,
based analyses have therefore served to widen the spectrum of 2003; Emerson et al., 2004]. A high rate of retroposition occurred
identified human-specific genes that have arisen since the in the primate and rodent lineages [Ohshima et al., 2003; Zhang
separation of human and chimpanzee lineages (e.g., the SMN2 et al., 2004b], probably driven by the activity of transposable
gene) [Rochette et al., 2001], members of the olfactory receptor elements such as LINEs and SVAs [Esnault et al., 2000; Torrents
gene [Trask et al., 1998], NANOG gene/pseudogene [Fairbanks et al., 2003; Zhang et al., 2003], which are able to transduce
and Maughan, 2006], keratinocyte growth factor (KGF) [Zimonjic non-transposon-derived DNA flanking their 30 ends to new
et al., 1997], and immunoglobulin G receptor gene families (e.g., genomic locations, a process termed 30 transduction [Moran et al.,
FCGR1) [Maresco et al., 1996, 1998], as well as 13 human 1999; Goodier et al., 2000; Ostertag et al., 2003].
lineage-specific PRAME genes [Birtle et al., 2005]. In a genomewide approach, Marques et al. [2005] estimated, on
A recent study of 9,600 gene families, identified by means of the basis of a systematic analysis of selective signatures in
a likelihood method that makes efficient use of comparative processed pseudogenes, that between 57 and 76 such retrogenes
genomic data [Hahn et al., 2005], examined gene birth and loss emerged during primate evolution. The majority of these primate-
among gene family members across five mammalian species specific retrogenes are specifically expressed in the testis in
(human, chimpanzee, mouse, rat, and dog). By aligning 13,454 contrast to the widely expressed original source genes that they
unambiguous orthologous genes of human and chimpanzee were derived from. Therefore, several of these retrocopies could
genomes, a gain of 812 genes was specifically noted in the human have experienced a gain-of-function during spermatogenesis in
lineage, with 1,180 genes having been gained in the chimpanzee primates [Marques et al., 2005]. One such well-characterized
lineage (J.P. Demuth and M.W. Hahn, personal communication). retrogene is glutamate dehydrogenase 2 (GLUD2), which
Similarly, 332 genes were found to have been lost specifically in originated in the hominoid ancestor 23 Mya by retroposition
the human lineage, with 1,043 genes having been lost in the from the ubiquitously expressed glutamate dehydrogenase 1
chimpanzee lineage. Thus, in total, the two primate genomes (GLUD1) gene. The unique and brain-specific properties of this
were found to be distinguishable by the presence or absence enzyme, which plays a role in the brain metabolism of the key
of some 3,731 genes (J.P. Demuth and M.W. Hahn, personal neurotransmitter glutamate, are not intrinsic to the parental
communication). GLUD1 gene but rather originated from amino acid substitutions
Taken together, these findings confirm the view espoused by that occurred during a postduplication period of positive selection
Nahon [2003], that the concept of human-specific genes can no during which brain size increased in the ancestors of both humans
longer be regarded as being heretical and that some of these genes and great apes [Burki and Kaessmann, 2004]. Thus, the GLUD2
may well have been responsible for influencing the evolutionary gene represents a paradigm for the evolution of new functions
emergence of human-specific traits. Indeed, human-lineage mediated by gene duplication during hominoid evolution.
specific copy number expansions of genes important for brain Comparative analysis of human chromosome 22 and the
functions and/or speech development may have had a particularly orthologous chromosome in the chimpanzee revealed a total
important impact on the human-specific evolution of these of six human-specific processed pseudogenes that probably
processes [Fortna et al., 2004; Sikela, 2006]. integrated into the human genome after the divergence of both
lineages [Watanabe et al., 2004]. The human/chimpanzee genome
Intragenic Duplications comparison revealed the existence of 163 human-specific
retrotransposed gene copies and 246 that arose specifically in
Intragenic duplications may also contribute to the genomic
the chimpanzee lineage [Mikkelsen et al., 2005]. From this finding
divergence between humans and chimpanzees. Thus, Jensen-
and the incomplete coverage of the chimpanzee genome, one
Seaman and Li [2003] have reported an intragenic duplication
might infer that there may be in excess of 200 human lineage-
involving the 60amino acid residue tandem repeats of the
specific and 300 chimpanzee-specific retrocopies, some of which
chimpanzee semenogelin 1 gene, resulting in a protein nearly twice
could have subsequently acquired new functions. Moreover, as
as long as that encoded by the human SEMG1 gene. Semenogelins
suggested by Balakirev and Ayala [2003], although pseudogene
1 and 2 represent the predominant structural proteins in human
sequences are generally less well conserved than functional genes
semen and the different structure of semenogelin 1 in humans and
and intergenic regions [Zheng et al., 2005], some pseudogenes
chimpanzee may be of significance in the context of the observed
common to humans and other primates may nevertheless have
differences in the consistency of semen between the two species.
evolved under lineage-specific selective constraints and might
thus be associated with the evolution of human-specific
Pseudogene Creation by Retroposition
phenotypic traits. Among the variety of different functions
Another mechanism of gene creation is the integration acquired by pseudogenes is the regulation of the mRNA stability
of reverse-transcribed processed mRNAs into genomic DNA of the parental source gene, a phenomenon exemplified by the
mediated by the enzymatic machinery of L1 non-LTR retro- makorin 1 (MKRN1) gene and its retrocopy [Hirotsune et al.,
transposons (retroposition) [Brosius, 1991; Long et al., 2003; 2003]. Further comparative analysis of pseudogenes in the human

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 117

and chimpanzee genomes, including other primates as outgroups, pericentric inversions of chromosomes 4, 5, 15, and 18. Employing
is required in order to ascertain the full spectrum of primate- gene order comparisons for the human and chimpanzee genomes,
specific (and human lineage-specific) pseudogenes and their Szamalek et al. [2006c] succeeded in identifying 71 submicro-
functional significance. scopic inversions. Nine of these that contained more than three
genes were then experimentally validated and the authenticity
Pseudogene-Mediated Gene Conversion of five of the nine inversions, ranging in size from 800 kb to
Pseudocopies not only represent a reservoir of genes that are 4.4 Mb, was confirmed. There is some overlap between the
capable of developing new functions, they also have the potential inversions identified in the above three studies. However, owing to
to mediate conversion of the parental source genes. It has been experimental limitations inherent to all three studies, the precise
proposed that pseudogene-mediated gene conversion may number of inversions that distinguish the human and chimpanzee
contribute to adaptive evolution and, in so doing, potentiate an genomes is difficult to ascertain with any degree of accuracy. It is
adaptive peak shift [Cooper, 1999; Hansen et al., 2000]. In nevertheless clear that small inversions make a very significant
principle, the rapid evolution of a novel gene function could be contribution to the structural divergence between the two
accomplished by the introduction of several substitutions simulta- genomes. Since the great majority of the inverted regions contain
neously. An example of just such a conversion has recently been genes and in some cases are even associated with the disruption
identified by Hayakawa et al. [2005], who noted that the of genes at the inversion breakpoints [Newman et al., 2005],
SIGLECP16 pseudogene has converted its functional counterpart, their potential to cause lineage-specific gene loss and differences
SIGLEC11. This conversion occurred specifically in the human in expression through position effects would appear to be quite
lineage rather than in that of the chimpanzee. The converted considerable.
region includes a 50 upstream region and exons encoding the sialic The breakpoints of many of the identified inversions map to
acid recognition domain. Strong Siglec-11 expression was observed sites of segmental duplications, an observation already made in the
in human cortex microglia (in contrast to chimpanzee and context of deletions and duplications. This serves to underline
orangutan), possibly due to changes in 50 regulatory sequences. the key role that segmental duplications play as mediators of
Gene conversions do not seem to be rare events. Indeed, by rearrangements in hominoid genomes due to the intrinsic
analyzing 2,641 duplicated genes in mouse and rat, 18% were propensity of these regions to recombine [Feuk et al., 2005;
found to exhibit signs of gene conversion [Ezawa et al., 2006]. Newman et al., 2005; Szamalek et al., 2006c]. To assess the
Pathological changes in functional genes mediated by gene significance of these inversions for the evolution of humans and
conversion through a pseudogene are not uncommon [Chiu chimpanzees it is important to study the lineage specificity of the
et al., 1997; Gupta et al., 2005; reviewed by Cooper, 1999; different inversions. This is necessary because some of the
Balakirev and Ayala, 2003]. Detailed analyses of gene conversion inversions identified as being divergent sites between human and
events involving duplicated genes in humans and chimpanzees chimpanzee actually represent intraspecies polymorphisms [Feuk
should provide information as to how frequently human lineage- et al., 2005; Szamalek et al., 2006c].
specific gene conversion has contributed to human gene evolution.
STRUCTURAL DIVERGENCE (INTERSPECIES
STRUCTURAL DIVERGENCE MEDIATED VARIABILITY) AND DIVERSITY
BY INVERSIONS (INTRASPECIES VARIANCE)
Inversions
As outlined above, genomic duplications as well as lineage-
specific deletions have contributed significantly to genome Of the 23 verified submicroscopic inversions described by Feuk
evolution in great apes and humans. In addition, submicroscopic et al. [2005], three (1, 17, and 700 kb in size, respectively) turned
inversions represent structural variants of the human and out to be polymorphic variants in humans. The 700-kb inversion
chimpanzee genomes [Fortna et al., 2004; Cheng et al., 2005; located on chromosome 7 was also identified by Szamalek et al.
Feuk et al., 2005; Hughes et al., 2005; Newman et al., 2005; [2006c], together with four inversions of 1 to 4.4 Mb that were
Szamalek et al., 2006c]. Thus, for example, the Y chromosomes of polymorphic in the chimpanzee. These findings indicate that both
humans and chimpanzees differ by two inversions, one in the the structural divergence and the structural diversity found in
chimpanzee lineage spanning 5 Mb, the other, of length 1.5 Mb, humans and chimpanzees are rather greater than was initially
being specific to humans [Hughes et al., 2005]. expected [Carter, 2004; Buckley et al., 2005; Feuk et al., 2006].
In a genomewide human/chimpanzee comparison, a total Inversions contribute to the unexpectedly high degree of
of 174 putative inversions were identified by Newman et al. structural variation in the human genome. In the study of Tuzun
[2005]. Their comparative approach included the mapping of et al. [2005], 56 inversions were identified that presumably
1.8 million fosmid end sequences from a genomic library of a represent regions of human polymorphism. Structural diversity
single chimpanzee (10-fold physical coverage of the chimpanzee between humans is therefore more the rule rather than the
genome) against the human genome reference sequence. A total exception. Prior to these studies, some variant inversions or gene
of 15 of these rearrangements spanned more than 20 Mb and duplications were already known and may turn out to be important
correspond to the cytogenetically visible large pericentric inver- in a clinical genetics context (summarized in Supplementary
sions. The remaining 159 putative inversions of 41.5 kb clearly Table S4). An inversion variant in the Williams-Beuren syndrome
contribute to the virtually uncharacterized bulk of submicroscopic (WBS) region in 7q11.23 has been found in 25 to 33% of
structural variation between the human and chimpanzee genomes. transmitting parents of individuals with WBS, implying that the
In contrast to this fosmid mapping strategy, Feuk et al. [2005] inversion could predispose this region to the WBS deletion
performed direct alignments of the chimpanzee draft sequence [Osborne et al., 2001; Scherer et al., 2005]. Further, submicro-
against the human reference sequence. These comparisons scopic inversions of 15q11q13 have been detected at dispropor-
disclosed 1,576 inversions ranging in size from 23 bp to 62 Mb, tionately high frequency (67%) in the mothers of Angelman
with the largest representing the known karyotypically visible syndrome patients with class II (BP2/3) deletions [Gimelli et al.,

Human Mutation DOI 10.1002/humu


118 HUMAN MUTATION 28(2), 99^130, 2007

2003], as compared to 9% in the general population. Only these studies is rather small. We may surmise that the methods
recently, a heterozygous inversion encompassing the NSD1 gene applied do not systematically detect all common polymorphic
at 5q35.3 has been identified in fathers of children with deletions and that the actual number may eventually prove to be
Sotos syndrome [Visser et al., 2005]. Since the latter inversion considerably higher [Eichler, 2006].
polymorphism is very common in the general population, further
work will be required to confirm or exclude its involvement in Structural Variation and Disease Susceptibility
predisposition to Sotos syndrome [Visser et al., 2005]. Structural variants such as inversions may impart disease
De novo recurrent chromosome rearrangements, involving susceptibility to the offspring of heterozygote inversion carriers
human chromosomes 4 and 8, are the recombinant products of by virtue of their potential to give rise to additional genomic
heterozygous inversions present in the transmitting parent. rearrangements. Copy number variations can also be associated
Furthermore, these inversions on 4p16 and 8p23 have been with disease susceptibility as has been exemplified by the CC
detected in 12.5 and 26% of control subjects, respectively, and chemokine ligand 3-like 1 (CCL3L1) gene, located within a site
thus represent quite frequent polymorphisms [Giglio et al., 2001, of segmental duplication. Copy number variations of CCL3L1-
2002]. As with the inversions involving 5q35.3, 7q11.23, and containing duplicons have been identified in different human
15q11q13, these inversions are flanked by paralogous segmental populations and a CCL3L1 copy number that is lower than
duplications (SD), a finding which emphasizes the importance of the population average is associated with enhanced susceptibility
duplicated regions as actual and potential mediators of inversions. to human immunodeficiency virus (HIV)/AIDS [Gonzalez
et al., 2005]. Copy number variation of human FCRG3 genes
Duplications and Deletions also appears to determine susceptibility to immunologically
mediated glomerulonephritis [Aitman et al., 2006].
Copy number variations (CNVs; the accepted term for Leaving aside the potential pathological significance of some
polymorphic duplications and deletions) would appear to be structural variants, it may be that other structural variants have
responsible for millions of nucleotides of heterogeneity between been advantageous during human evolution. If a structural
extant human genomes. Indeed, it is likely to contribute to a much polymorphism such as an inversion has been maintained in a
larger extent to human diversity, and probably also to disease population for a long period of time, a selective advantage accruing
susceptibility, than has previously been assumed [Carter, 2004; to carriers may be inferred. In contrast to SNPs, heterozygous
Buckley et al., 2005; Feuk et al., 2006; Wilson et al., 2006b]. inversions are more likely to be naturally eliminated from the
DNA microarray analysis has proved to be a powerful tool for population since crossing over within the inverted segments results
the detection of CNVs 44050 kb in size [Iafrate et al., 2004; in genetically unbalanced offspring. If, however, such inversion
Sebat et al., 2004; Sharp et al., 2005; Goidts et al., 2006]. Thus, heterozygosity is maintained over an extended period of time, a
extrapolating to the entire genome, any two human individuals selective advantage resulting from this heterozygosity might be
are likely to be distinguishable by approximately 100 CNVs 440 assumed. A human polymorphic inversion that appears to have
50 kb [Feuk et al., 2006]. CNVs were also detected through the evolved under positive selection has recently been reported by
alignment of paired-end sequence data from a human fosmid Stefansson et al. [2005]: a 900-kb inversion polymorphism at
genomic library against the human reference sequence [Tuzun 17q21.31 within a region that includes the microtubule-associated
et al., 2005]. In this study, 102 regions of the human genome were protein tau (MAPT) gene. Chromosomes with the inverted
identified that constitute sites of polymorphic deletion of 48 kb in segment in different orientations represent two distinct lineages,
length. Human CNVs are significantly overrepresented in the termed H1 and H2, that appear to have diverged for as many as
proximity of telomeres and centromeres, and in simple tandem 3 million years with no evidence of recombination between them.
repeat sequences [Tuzun et al., 2005]. Furthermore, human CNVs The high frequency of H2 chromosomes in European populations
are nonrandomly distributed, with a four- to 12-fold greater is consistent with the rapid expansion of this allele under the
frequency of occurrence close to SDs [Sharp et al., 2005; Tuzun influence of positive selection in Europe and the Near East
et al., 2005; Goidts et al., 2006; Kriek et al., 2006]. In chimpanzee [Stefansson et al., 2005].
populations, a 20-fold enrichment of CNVs at sites of SDs has The detailed analyses of these polymorphic variants with regard
been noted [Perry et al., 2006]. This indicates that SDs have an to structure, segregation and frequency in the human population
increased tendency to vary in copy number, thereby mediating promises to lead to significant new insights into the nature and
common human (and chimpanzee) genetic variation [Mehan impact of both negative and positive selection pressure during
et al., 2004; Perry et al., 2006]. Further, CNVs are enriched in human evolution. Whether with regard to studies of human
protein-coding genes that have experienced significantly elevated evolution or disease susceptibility, the analysis of human genome
synonymous and nonsynonymous nucleotide substitution rates as diversity, and in particular those changes involving structural
compared to orthologous genes in the mouse. The elevated coding variation, will benefit enormously from comparisons with the
sequence divergence suggests that a subset of genes within CNVs genomes of other primates. These comparisons should help to
may be associated with an adaptive increase in gene dosage reveal ancestral states, lineage-specific evolutionary trends, and
[Nguyen et al., 2006b]. signatures of adaptive diversity, as well as their phenotypic
SNP genotyping and oligonucleotide microarray analyses of functional consequences.
long-range PCR products have revealed that polymorphic dele-
tions ranging from 0.510.5 kb are widespread in the human
OVERVIEW
genome [Conrad et al., 2006; Hinds et al., 2006; McCarroll et al.,
2006]; indeed, by these means, 1,000 regions harboring such The plethora of genetic changes that differentiate humans from
deletion polymorphisms were identified in the human genome. their closest relatives the chimpanzees affect multiple levels and
This number is, however, likely to represent a conservative are much more complex than was assumed prior to the advent
estimate of the total number of polymorphic deletions in the of genomewide comparative analyses. A considerable number of
human genome since the overlap of deletion variants identified in human genes have so far been identified that evolved under recent

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 119

positive selection. Some of these may turn out to have had a reveals rapid evolution by multiple mechanisms. Proc Natl Acad Sci
major impact on the evolution of human-specific traits. USA 101:1325113256.
Further, differential expression patterns of large sets of genes can Antonarakis SE, Rossiter JP, Young M, Horst J, de Moerloose P, Sommer SS,
discriminate the transcriptomes of humans and chimpanzees. Ketterling RP, Kazazian HH Jr, Negrier C, Vinciguerra C, Gitschier J,
Even more striking is that transcriptomes and proteomes appear Goossens M, Girodon E, Ghanem N, Plassa F, Lavergne JM, Vidaud M,
Costa JM, Laurian Y, Lin SW, Lin SR, Shen MC, Lillicrap D, Taylor SA,
to have coevolved in humans, implying a complex interplay of
Windsor S, Valleix SV, Nafa K, Sultan Y, Delpech M, Vnencak-Jones
evolutionary forces acting at different levels of the expression CL, Phillips JA 3rd, Ljung RC, Koumbarelis E, Gialeraki A, Mandalaki T,
pathway. Jenkins PV, Collins PW, Pasi KJ, Goodeve A, Peake I, Preston FE,
The continuing search for differences between the human and Schwartz M, Scheibel E, Ingerslev J, Cooper DN, Millar DS, Kakkar VV,
chimpanzee genomes has identified extensive structural variation Giannelli F, Naylor JA, Tizzano EF, Baiget M, Domenech M, Altisent C,
that has been frequently mediated by recombination-prone Tusell J, Beneyto M, Lorenzo JI, Gaucher C, Mazurier C, Peerlinck K,
segmentally duplicated sequences and is likely to be associated Matthijs G, Cassiman JJ, Vermylen J, Mori PG, Acquila M, Caprino D,
with a multitude of functional changes that still largely remain to Inaba H. 1995. Factor VIII gene inversions in severe hemophilia A:
be identified. The identification of the individual components results of an international consortium study. Blood 86:22062212.
of the structural divergence between the human and chimpanzee Arbiza L, Dopazo J, Dopazo H. 2006. Positive selection, relaxation, and
acceleration in the evolution of the human and chimp genome. PLoS
genomes has been accompanied by the discovery of very
Comput Biol 2:e38.
considerable lineage-specific structural diversity. Indeed, copy
Archidiacono N, Antonacci R, Marzella R, Finelli P, Lonoce A, Rocchi M.
number polymorphisms as well as inversions have turned out 1995. Comparative mapping of human alphoid sequences in great apes
to be a major contributor to the polymorphism manifest in using fluorescence in situ hybridization. Genomics 25:477484.
extant human genomes. At least some of these structural variants Armengol L, Pujana MA, Cheung J, Scherer SW, Estivill X. 2003.
are likely to turn out to be associated with disease susceptibility Enrichment of segmental duplications in regions of breaks of synteny
or complex trait development. The parallel investigation between the human and mouse genomes suggest their involvement in
of the corresponding regions in the chimpanzee and other primate evolutionary rearrangements. Hum Mol Genet 12:22012208.
genomes will not only help us to distinguish between Arnason U, Xu X, Gullberg A. 1996. Comparison between the complete
genetic divergence and diversity, but is also likely to provide mitochondrial DNA sequences of Homo and the common chimpanzee
based on nonchimeric sequences. J Mol Evol 42:145152.
important new insights into the evolutionary history of our
Asthana S, Schmidt S, Sunyaev S. 2005. A limited role for balancing
extended primate family.
selection. Trends Genet 21:3032.
Babcock M, Pavlicek A, Spiteri E, Kashork CD, Ioshikhes I, Shaffer LG,
ACKNOWLEDGMENTS Jurka J, Morrow BE. 2003. Shuffling of genes within low-copy repeats on
22q11 (LCR22) by Alu-mediated recombination events during evolu-
We thank Violaine Goidts, Justyna M. Szamalek, Catharina tion. Genome Res 13:25192532.
Sandig, Werner Schempp, Stefan Mu ller, and Matthias Platzer Bacolla A, Wells RD. 2004. Non-B DNA conformations, genomic
for their scientific contributions to the ongoing work in Ulm, rearrangements, and human disease. J Biol Chem 279:4741147414.
and Matthew Hahn for access to data prior to publication. We Bacolla A, Jaworski A, Larson JE, Jakupciak JP, Chuzhanova N, Abeysinghe
also thank Horst Hameister and James M. Sikela for helpful SS, OConnell CD, Cooper DN, Wells RD. 2004. Breakpoints of gross
discussions. deletions coincide with non-B DNA conformations. Proc Natl Acad Sci
USA 101:1416214167.
Bacolla A, Collins RA, Gold B, Chuzhanova N, Yi M, Stephens RM,
REFERENCES
Stefanov S, Olsh A, Jakupciak JP, Dean M, Lempicki RA, Cooper DN,
Aitken RJ, Marshall Graves AJ. 2002. The future of sex. Nature 41:963. Wells RD. 2006. Long homopurine  homopyrimidine sequences are
Aitman TJ, Dong R, Vyse TJ, Norsworthy PJ, Johnson MD, Smith J, characteristic of genes expressed in brain and the pseudoautosomal
Mangion J, Roberton-Lowe C, Marshall AJ, Petretto E, Hodges MD, region. Nucleic Acids Res 34:26632675.
Bhangal G, Patel SG, Sheehan-Rooney K, Duda M, Cook PR, Evans DJ, Bailey JA, Yavor AM, Massa HF, Trask BJ, Eichler EE. 2001. Segmental
Domin J, Flint J, Boyle JJ, Pusey CD, Cook HT. 2006. Copy number duplications: organization and impact within the current human genome
polymorphism in Fcgr3 predisposes to glomerulonephritis in rats and project assembly. Genome Res 11:10051017.
humans. Nature 439:851855. Bailey JA, Gu Z, Clark RA, Reinert K, Samonte RV, Schwartz S, Adams
Aldred PM, Hollox EJ, Armour JA. 2005. Copy number polymorphism and MD, Myers EW, Li PW, Eichler EE. 2002a. Recent segmental
expression level variation of the human alpha-defensin genes DEFA1 duplications in the human genome. Science 297:10031007.
and DEFA3. Hum Mol Genet 14:20452052. Bailey JA, Yavor AM, Viggiano L, Misceo D, Horvath JE, Archidiacono N,
Alexandrov I, Kazakov A, Tumeneva I, Shepelev V, Yurov Y. 2001. Alpha- Schwartz S, Rocchi M, Eichler EE. 2002b. Human-specific duplication
satellite DNA of primates: old and new families. Chromosoma 110: and mosaic transcripts: the recent paralogous structure of chromosome
253266. 22. Am J Hum Genet 70:83100.
Alves G, Seuanez HN, Fanning T. 1994. Alpha satellite DNA in Bailey JA, Baertsch R, Kent WJ, Haussler D, Eichler EE. 2004. Hotspots of
neotropical primates (Platyrrhini). Chromosoma 103:262267. mammalian chromosomal evolution. Genome Biol 5:R23.
Andres AM, Soldevila M, Navarro A, Kidd KK, Oliva B, Bertranpetit J. Bailey JA, Eichler EE. 2006. Primate segmental duplications: crucibles of
2004a. Positive selection in MAOA gene is human exclusive: evolution, diversity and disease. Nat Rev Genet 7:552564.
determination of the putative amino acid change selected in the human Bailey WJ, Fitch DH, Tagle DA, Czelusniak J, Slightom JL, Goodman M.
lineage. Hum Genet 115:377386. 1991. Molecular evolution of the psi eta-globin gene locus:
Andres AM, Soldevila M, Lao O, Volpini V, Saitou N, Jacobs HT, Hayasaka gibbon phylogeny and the hominoid slowdown. Mol Biol Evol 8:
I, Calafell F, Bertranpetit J. 2004b. Comparative genetics of functional 155184.
trinucleotide tandem repeats in humans and apes. J Mol Evol 59: Balakirev ES, Ayala FJ. 2003. Pseudogenes: are they junk or functional
329339. DNA? Annu Rev Genet 37:123151.
Angata T, Varki NM, Varki A. 2001. A second uniquely human mutation Balter M. 2005. Genetics. Expression of endorphin gene favored in human
affecting sialic acid biology. J Biol Chem 276:4028240287. evolution. Science 310:1257.
Angata T, Margulies EH, Green ED, Varki A. 2004. Large-scale sequencing Bamshad M, Wooding SP. 2003. Signatures of natural selection in the
of the CD33-related Siglec gene cluster in five mammalian species human genome. Nat Rev Genet 4:99111.

Human Mutation DOI 10.1002/humu


120 HUMAN MUTATION 28(2), 99^130, 2007

Bannert N, Kurth R. 2004. Retroelements and the human genome: new Buzdin A, Ustyugova S, Khodosevich K, Mamedov I, Lebedev Y, Hunsmann
perspectives on an old relation. Proc Natl Acad Sci USA 101(Suppl 2): G, Sverdlov E. 2003. Human-specific subfamilies of HERV-K (HML-2)
1457214579. long terminal repeats: three master genes were active simultaneously
Batzer MA, Deininger PL. 2002. Mammalian retroelements. Genome Res during branching of hominoid lineages. Genomics 81:149156.
12:14551465. Caceres M, Lachuer J, Zapala MA, Redmond JC, Kudo L, Geschwind DH,
Bennett EA, Coleman LE, Tsui C, Pittard WS, Devine SE. 2004. Natural Lockhart DJ, Preuss TM, Barlow C. 2003. Elevated gene expression
genetic variation caused by transposable elements in humans. Genetics levels distinguish human from non-human primate brains. Proc Natl
168:933951. Acad Sci USA 100:1303013035.
Bersaglieri T, Sabeti PC, Patterson N, Vanderploeg T, Schaffner SF, Drake Carroll ML, Roy-Engel AM, Nguyen SV, Salem AH, Vogel E, Vincent B,
JA, Rhodes M, Reich DE, Hirschhorn JN. 2004. Genetic signatures of Myers J, Ahmad Z, Nguyen L, Sammarco M, Watkins WS, Henke J,
strong recent positive selection at the lactase gene. Am J Hum Genet Makalowski W, Jorde LB, Deininger PL, Batzer MA. 2001. Large-scale
74:11111120. analysis of the Alu Ya5 and Yb8 subfamilies and their contribution to
Betran E, Long M. 2002. Expansion of genome coding regions by human genomic diversity. J Mol Biol 311:1740.
acquisition of new genes. Genetica 115:6580. Carroll SB. 2003. Genetics and the making of Homo sapiens. Nature
Betran E, Wang W, Jin L, Long M. 2002. Evolution of the phosphoglycerate 422:849857.
mutase processed gene in human and chimpanzee revealing the origin of Carroll SB. 2005. Evolution at two levels: on genes and form. PLoS Biol
a new primate gene. Mol Biol Evol 19:654663. 3:e245.
Birtle Z, Goodstadt L, Ponting C. 2005. Duplication and positive selection Carson AR, Cheung J, Scherer SW. 2006. Duplication and relocation of
among hominin-specific PRAME genes. BMC Genomics 13:120. the functional DPY19L2 gene within low copy repeats. BMC Genomics
Bishop DV. 2002. Putting language genes in perspective. Trends Genet 7:45.
18:5759. Carter NP. 2004. As normal as normal can be? Nat Genet 36:931932.
Bond J, Roberts E, Mochida GH, Hampshire DJ, Scott S, Askham JM, Carter AB, Salem AH, Hedges DJ, Keegan CN, Kimball B, Walker JA,
Springell K, Mahadevan M, Crow YJ, Markham AF, Walsh CA, Woods Watkins WS, Jorde LB, Batzer MA. 2004. Genome-wide analysis of the
CG. 2002. ASPM is a major determinant of cerebral cortical size. Nat human Alu Yb-lineage. Hum Genomics 1:167178.
Genet 32:316320. Charlesworth B, Charlesworth D. 2000. The degeneration of Y chromo-
Bondeson ML, Dahl N, Malmgren H, Kleijer WJ, Tonnesen T, Carlberg somes. Philos Trans R Soc Lond B Biol Sci 355:15631572.
BM, Pettersson U. 1995. Inversion of the IDS gene resulting from Chen FC, Li WH. 2001. Genomic divergences between humans and other
recombination with IDS-related sequences is a common cause of the hominoids and the effective population size of the common ancestor of
Hunter syndrome. Hum Mol Genet 4:615621. humans and chimpanzees. Am J Hum Genet 68:444456.
Boniotto M, Tossi A, DelPero M, Sgubin S, Antcheva N, Santon D, Chen FC, Vallender EJ, Wang H, Tzeng CS, Li WH. 2001. Genomic
Masters J, Crovella S. 2003. Evolution of the beta defensin 2 gene in divergence between human and chimpanzee estimated from large-scale
primates. Genes Immun 4:251257. alignments of genomic sequences. J Hered 92:481489.
Boue S, Letunic I, Bork P. 2003. Alternative splicing and evolution. Cheng HY, Pitcher GM, Laviolette SR, Whishaw IQ, Tong KI, Kockeritz
Bioessays 25:10311034. LK, Wada T, Joza NA, Crackower M, Goncalves J, Sarosi I, Woodgett JR,
Bray NJ, Buckland PR, Owen MJ, ODonovan MC. 2003. Cis-acting Oliveira-dos-Santos AJ, Ikura M, van der Kooy D, Salter MW, Penninger
variation in the expression of a high proportion of genes in human brain. JM. 2002. DREAM is a critical transcriptional repressor for pain
Hum Genet 113:149153. modulation. Cell 108:3143.
Britten RJ. 2002. Divergence between samples of chimpanzee and human Cheng Z, Ventura M, She X, Khaitovich P, Graves T, Osoegawa K, Church
DNA sequences is 5%, counting indels. Proc Natl Acad Sci USA 99: D, DeJong P, Wilson RK, Paabo S, Rocchi M, Eichler EE. 2005. A
1363313635. genome-wide comparison of recent chimpanzee and human segmental
Britten RJ, Rowen L, Williams J, Cameron RA. 2003. Majority of duplications. Nature 437:8893.
divergence between closely related DNA samples is due to indels. Proc Cheung J, Estivill X, Khaja R, MacDonald JR, Lau K, Tsui LC, Scherer SW.
Natl Acad Sci USA 100:46614665. 2003. Genome-wide detection of segmental duplications and potential
Brosius J. 1991. Retroposonsseeds of evolution. Science 251:753. assembly errors in the human genome sequence. Genome Biol 4:R25.
Brosius J. 1999. DNAs from all categories generate retrosequences that may Chiu CH, Schneider H, Slightom JL, Gumucio DL, Goodman M. 1997.
be exapted as novel genes or regulatory elements. Gene 238:115134. Dynamics of regulatory evolution in primate beta-globin gene clusters:
Brouha B, Schustak J, Badge RM, Lutz-Prigge S, Farley AH, Moran JV, cis-mediated acquisition of simian gamma fetal expression patterns. Gene
Kazazian HH Jr. 2003. Hot L1s account for the bulk of retrotransposition 205:4757.
in the human population. Proc Natl Acad Sci USA 100:52805285. Choi SS, Lahn BT. 2003. Adaptive evolution of MRG, a neuron-specific
Buckland PR. 2004. Allele-specific gene expression differences in humans. gene family implicated in nociception. Genome Res 13:22522259.
Hum Mol Genet 13:R255R260. Choong CS, Kemppainen JA, Wilson EM. 1998. Evolution of the primate
Buckley PG, Mantripragada KK, Piotrowski A, Diaz de Stahl T, Dumanski androgen receptor: a structural basis for disease. J Mol Evol 47:334342.
JP. 2005. Copy-number polymorphisms: mining the tip of an iceberg. Chou HH, Takematsu H, Diaz S, Iber J, Nickerson E, Wright KL,
Trends Genet 21:315317. Muchmore EA, Nelson DL, Warren ST, Varki A. 1998. A mutation in
Burki F, Kaessmann H. 2004. Birth and adaptive evolution of a hominoid human CMP-sialic acid hydroxylase occurred after the Homo-Pan
gene that supports high neurotransmitter flux. Nat Genet divergence. Proc Natl Acad Sci USA 95:1175111756.
36:10611063. Ciccarelli FD, von Mering C, Suyama M, Harrington ED, Izaurralde E,
Bush EC, Lahn BT. 2005. Selective constraint on noncoding regions of Bork P. 2005. Complex genomic rearrangements lead to novel primate
hominid genomes. PLoS Comput Biol 16:e73. gene function. Genome Res 15:343351.
Bussell JJ, Pearson NM, Kanda R, Filatov DA, Lahn BT. 2006. Human Clark AG, Glanowski S, Nielsen R, Thomas PD, Kejariwal A, Todd MA,
polymorphism and human-chimpanzee divergence in pseudoautosomal Tanenbaum DM, Civello D, Lu F, Murphy B, Ferriera S, Wang G, Zheng
region correlate with local recombination rate. Gene 368:94100. X, White TJ, Sninsky JJ, Adams MD, Cargill M. 2003. Inferring
Bustamante CD, Nielsen R, Hartl DL. 2002. A maximum likelihood nonneutral evolution from human-chimp-mouse orthologous gene trios.
method for analyzing pseudogene evolution: implications for silent site Science 302:19601963.
evolution in humans and rodents. Mol Biol Evol 19:110117. Clark NL, Swanson WJ. 2005. Pervasive adaptive evolution in primate
Bustamante CD, Fledel-Alon A, Williamson S, Nielsen R, Hubisz MT, seminal proteins. PLoS Genet 1:e35.
Glanowski S, Tanenbaum DM, White TJ, Sninsky JJ, Hernandez RD, Clark RM, Bhaskar SS, Miyahara M, Dalgliesh GL, Bidichandani SI. 2006.
Civello D, Adams MD, Cargill M, Clark AG. 2005. Natural selection on Expansion of GAA trinucleotide repeats in mammals. Genomics 87:
protein-coding genes in the human genome. Nature 437:11531157. 5767.

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 121

Conrad DF, Andrews TD, Carter NP, Hurles ME, Pritchard JK. 2006. A Eichler EE. 2001. Recent duplication, domain accretion and the dynamic
high-resolution survey of deletion polymorphism in the human genome. mutation of the human genome. Trends Genet 17:661669.
Nat Genet 38:7581. Eichler EE, Clark RA, She X. 2004. An assessment of the sequence gaps:
Conte C, Ebeling M, Marcuz A, Nef P, Andres-Barquin PJ. 2002. unfinished business in a finished human genome. Nat Rev Genet 5:
Identification and characterization of human taste receptor genes 345354.
belonging to the TAS2R family. Cytogenet Genome Res 98:4553. Eichler EE. 2006. Widening the spectrum of human genetic variation. Nat
Cooper G, Rubinsztein DC, Amos W. 1998. Ascertainment bias cannot Genet 38:911.
entirely account for human microsatellites being longer than their Elango N, Thomas JW, Yi SV. 2006. Variable molecular clocks in
chimpanzee homologues. Hum Mol Genet 7:14251429. hominoids. Proc Natl Acad Sci USA 103:13701375.
Cooper DN. 1999. Human gene evolution. Oxford: BIOS Scientific, Ellegren H. 2005. Evolution: natural selection in the evolution of humans
Oxford. pp.276278. and chimps. Curr Biol 15:R919R922.
Costas J. 2001. Evolutionary dynamics of the human endogenous retrovirus Emerson JJ, Kaessmann H, Betran E, Long M. 2004. Extensive gene traffic
family HERV-K inferred from full-length proviral genomes. J Mol Evol on the mammalian X chromosome. Science 303:537540.
53:237243. Enard W, Khaitovich P, Klose J, Zollner S, Heissig F, Giavalisco P, Nieselt-
Courseaux A, Nahon JL. 2001. Birth of two chimeric genes in the Struwe K, Muchmore E, Varki A, Ravid R, Doxiadis GM, Bontrop RE,
Hominidae lineage. Science 291:12931297. Paabo S. 2002. Intra-and interspecific variation in primate gene
Courseaux A, Richard F, Grosgeorge J, Ortola C, Viale A, Turc-Carel C, expression patterns. Science 296:340343.
Dutrillaux B, Gaudray P, Nahon JL. 2003. Segmental duplications in Enard W, Paabo S. 2004. Comparative primate genomics. Annu Rev
euchromatic regions of human chromosome 5: a source of evolutionary Genomics Hum Genet 5:351378.
instability and transcriptional innovation. Genome Res 13:369381. Enard W, Fassbender A, Model F, Adorjan P, Paabo S, Olek A. 2004.
Crosier M, Viggiano L, Guy J, Misceo D, Stones R, Wei W, Hearn T, Differences in DNA methylation patterns between humans and
Ventura M, Archidiacono N, Rocchi M, Jackson MS. 2002. Human chimpanzees. Curr Biol 14:R148149.
paralogs of KIAA0187 were created through independent pericentro- Escalante AA, Barrio E, Ayala FJ. 1995. Evolutionary origin of human and
meric-directed and chromosome-specific duplication mechanisms. primate malarias: evidence from the circumsporozoite protein gene. Mol
Genome Res 12:6780. Biol Evol 12:616626.
Daniels RJ, Peden JF, Lloyd C, Horsley SW, Clark K, Tufarelli C, Kearney L, Esnault C, Maestre J, Heidmann T. 2000. Human LINE retrotransposons
Buckle VJ, Doggett NA, Flint J, Higgs DR. 2001. Sequence, structure generate processed pseudogenes. Nat Genet 24:363367.
and pathology of the fully annotated terminal 2 Mb of the short arm of Evans PD, Anderson JR, Vallender EJ, Gilbert SL, Malcom CM, Dorus S,
human chromosome 16. Hum Mol Genet 10:339352. Lahn BT. 2004a. Adaptive evolution of ASPM, a major determinant of
Deeb SS, Jorgensen AL, Battisti L, Iwasaki L, Motulsky AG. 1994. cerebral cortical size in humans. Hum Mol Genet 13:489494.
Sequence divergence of the red and green visual pigments in great apes Evans PD, Anderson JR, Vallender EJ, Choi SS, Lahn BT. 2004b.
and humans. Proc Natl Acad Sci USA 91:72627266. Reconstructing the evolutionary history of microcephalin, a gene
Dennehey BK, Gutches DG, McConkey EH, Krauter KS. 2004. Inversion, controlling human brain size. Hum Mol Genet 13:11391145.
duplication, and changes in gene context are associated with human Evans PD, Gilbert SL, Mekel-Bobrov N, Vallender EJ, Anderson JR,
chromosome 18 evolution. Genomics 83:493501. Vaez-Azizi LM, Tishkoff SA, Hudson RR, Lahn BT. 2005. Micro-
Dolphin CT, Beckett DJ, Janmohamed A, Cullingford TE, Smith RL, cephalin, a gene regulating brain size, continues to evolve adaptively in
Shephard EA, Phillips IR. 1998. The flavin-containing monooxygenase 2 humans. Science 309:17171720.
gene (FMO2) of humans, but not of other primates, encodes a Eyre-Walker A, Keightley PD. 1999. High genomic deleterious mutation
truncated, nonfunctional protein. J Biol Chem 273:3059930607. rates in hominids. Nature 397:344347.
Donaldson IJ, Gottgens B. 2006. Evolution of candidate transcriptional Ezawa K, Oota S, Saitou N. 2006. Genomewide search of gene conversions
regulatory motifs since the human-chimpanzee divergence. Genome Biol in duplicated genes of mouse and rat. Mol Biol Evol 23:927940.
7:R52. Fairbanks DJ, Maughan PJ. 2006. Evolution of the NANOG pseudo-
Dorus S, Vallender EJ, Evans PD, Anderson JR, Gilbert SL, Mahowald M, gene family in the human and chimpanzee genomes. BMC Evol
Wyckoff GJ, Malcom CM, Lahn BT. 2004. Accelerated evolution of Biol 6:12.
nervous system genes in the origin of Homo sapiens. Cell 119: Fan Y, Linardopoulou E, Friedman C, Williams E, Trask BJ. 2002. Genomic
10271040. structure and evolution of the ancestral chromosome fusion site in
Dorus S, Anderson JR, Vallender EJ, Gilbert SL, Zhang L, Chemnick LG, 2q132q14.1 and paralogous regions on other human chromosomes.
Ryder OA, Li W, Lahn BT. 2006. Sonic Hedgehog, a key development Genome Res 12:16511662.
gene, experienced intensified molecular evolution in primates. Hum Mol Fay JC, Wu CI. 2000. Hitchhiking under positive Darwinian selection.
Genet 15:20312037. Genetics 155:14051413.
Dutrillaux B. 1979. Chromosomal evolution in primates: tentative Feuk L, Macdonald JR, Tang T, Carson AR, Li M, Rao G, Khaja R, Scherer
phylogeny from Microcebus murinus (Prosimian) to man. Hum Genet SW. 2005. Discovery of human inversion polymorphisms by comparative
48:251314. analysis of human and chimpanzee DNA sequence assemblies. PLoS
Ebersberger I, Metzler D, Schwarz C, Paabo S. 2002. Genomewide Genet 1:e56.
comparison of DNA sequences between humans and chimpanzees. Feuk L, Carson AR, Scherer SW. 2006. Structural variation in the human
Am J Hum Genet 70:14901497. genome. Nat Rev Genet 7:8597.
Ebersberger I, Meyer M. 2005. A genomic region evolving toward Filatov DA. 2004. A gradient of silent substitution rate in the human
different GC contents in humans and chimpanzees indicates a recent pseudoautosomal region. Mol Biol Evol 21:410417.
and regionally limited shift in the mutation pattern. Mol Biol Evol 22: Fischer H, Koenig U, Eckhart L, Tschachler E. 2002. Human caspase 12
12401245. has acquired deleterious mutations. Biochem Biophys Res Commun
Eichler EE, Lu F, Shen Y, Antonacci R, Jurecic V, Doggett NA, Moyzis RK, 293:722726.
Baldini A, Gibbs RA, Nelson DL. 1996. Duplication of a gene-rich Fisher SE, Marcus GF. 2006. The eloquent ape: genes, brains and the
cluster between 16p11.1 and Xq28: a novel pericentromeric-directed evolution of language. Nat Rev Genet 7:920.
mechanism for paralogous genome evolution. Hum Mol Genet 5: Flint J, Thomas K, Micklem G, Raynham H, Clark K, Doggett NA, King A,
899912. Higgs DR. 1997. The relationship between chromosome structure and
Eichler EE, Budarf ML, Rocchi M, Deaven LL, Doggett NA, Baldini A, function at a human telomeric region. Nat Genet 15:252257.
Nelson DL, Mohrenweiser HW. 1997. Interchromosomal duplications of Fortna A, Kim Y, MacLaren E, Marshall K, Hahn G, Meltesen L, Brenton
the adrenoleukodystrophy locus: a phenomenon of pericentromeric M, Hink R, Burgers S, Hernandez-Boussard T, Karimpour-Fard A,
plasticity. Hum Mol Genet 6:9911002. Glueck D, McGavran L, Berry R, Pollack J, Sikela JM. 2004. Lineage-

Human Mutation DOI 10.1002/humu


122 HUMAN MUTATION 28(2), 99^130, 2007

specific gene duplication and loss in human and great ape evolution. Giglio S, Calvari V, Gregato G, Gimelli G, Camanini S, Giorda R, Ragusa
PLoS Biol 2:e207. A, Guerneri S, Selicorni A, Stumm M, Tonnies H, Ventura M, Zollino
Fraser HB, Khaitovich P, Plotkin JB, Paabo S, Eisen MB. 2005. Aging and M, Neri G, Barber J, Wieczorek D, Rocchi M, Zuffardi O. 2002.
gene expression in the primate brain. PLoS Biol 3:e274. Heterozygous submicroscopic inversions involving olfactory receptor-
Frazer KA, Chen X, Hinds DA, Pant PV, Patil N, Cox DR. 2003. Genomic gene clusters mediate the recurrent t(4;8) (p16; p23) translocation. Am
DNA insertions and deletions occur frequently between humans and J Hum Genet 71:276285.
nonhuman primates. Genome Res 13:341346. Gilad Y, Bustamante CD, Lancet D, Paabo S. 2003a. Natural selection
Fujiyama A, Watanabe H, Toyoda A, Taylor TD, Itoh T, Tsai SF, Park HS, on the olfactory receptor gene family in humans and chimpanzees. Am J
Yaspo ML, Lehrach H, Chen Z, Fu G, Saitou N, Osoegawa K, de Jong PJ, Hum Genet 73:489501.
Suto Y, Hattori M, Sakaki Y. 2002. Construction and analysis of a Gilad Y, Man O, Paabo S, Lancet D. 2003b. Human specific loss of olfactory
human-chimpanzee comparative clone map. Science 295:131134. receptor genes. Proc Natl Acad Sci USA 100:33243327.
Gagneux P, Boesch C, Woodruff DS. 1999. Female reproductive strategies, Gilad Y, Oshlack A, Smyth GK, Speed TP, White KP. 2006. Expression
paternity and community structure in wild West African chimpanzees. profiling in primates reveals a rapid evolution of human transcription
Anim Behav 57:1932. factors. Nature 440:242245.
Gagneux P, Varki A. 2001. Genetic differences between humans and great Gimelli G, Pujana MA, Patricelli MG, Russo S, Giardino D, Larizza L,
apes. Mol Phylogenet Evol 18:213. Cheung J, Armengol L, Schinzel A, Estivill X, Zuffardi O. 2003.
Gao X, Nelson GW, Karacki P, Martin MP, Phair J, Kaslow R, Goedert JJ, Genomic inversions of human chromosome 15q11-q13 in mothers of
Buchbinder S, Hoots K, Vlahov D, OBrien SJ, Carrington M. 2001. Angelman syndrome patients with class II (BP2/3) deletions. Hum Mol
Effect of a single amino acid change in MHC class I molecules on the Genet 12:849858.
rate of progression to AIDS. N Engl J Med 344:16681675. Glazko GV, Nei M. 2003. Estimation of divergence times for major lineages
Gibbons R, Dugaiczyk LJ, Girke T, Duistermars B, Zielinski R, Dugaiczyk A. of primate species. Mol Biol Evol 20:424434.
2004. Distinguishing humans from great apes with AluYb8 repeats. J Mol Glazko G, Veeramachaneni V, Nei M, Makalowski W. 2005. Eighty percent
Biol 339:721729. of proteins are different between humans and chimpanzees. Gene
Gibbs RA, Weinstock GM, Metzker ML, Muzny DM, Sodergren EJ, 346:215219.
Scherer S, Scott G, Steffen D, Worley KC, Burch PE, Okwuonu G, Hines Glusman G, Yanai I, Rubin I, Lancet D. 2001. The complete human
S, Lewis L, DeRamo C, Delgado O, Dugan-Rocha S, Miner G, Morgan olfactory subgenome. Genome Res 11:685702.
M, Hawes A, Gill R, Celera, Holt RA, Adams MD, Amanatides PG, Go Y, Satta Y, Takenaka O, Takahata N. 2005. Lineage-specific loss of
Baden-Tillson H, Barnstead M, Chin S, Evans CA, Ferriera S, Fosler C, function of bitter taste receptor genes in humans and nonhuman
Glodek A, Gu Z, Jennings D, Kraft CL, Nguyen T, Pfannkoch CM, Sitter primates. Genetics 170:313326.
C, Sutton GG, Venter JC, Woodage T, Smith D, Lee HM, Gustafson E, Goidts V, Szamalek JM, Hameister H, Kehrer-Sawatzki H. 2004. Segmental
Cahill P, Kana A, Doucette-Stamm L, Weinstock K, Fechtel K, Weiss duplication associated with the human-specific inversion of chromosome
RB, Dunn DM, Green ED, Blakesley RW, Bouffard GG, De Jong PJ, 18: a further example of the impact of segmental duplications on
Osoegawa K, Zhu B, Marra M, Schein J, Bosdet I, Fjell C, Jones S, karyotype and genome evolution in primates. Hum Genet 115:116122.
Krzywinski M, Mathewson C, Siddiqui A, Wye N, McPherson J, Zhao S, Goidts V, Szamalek JM, de Jong PJ, Cooper DN, Chuzhanova N, Hameister
Fraser CM, Shetty J, Shatsman S, Geer K, Chen Y, Abramzon S, H, Kehrer-Sawatzki H. 2005. Independent intrachromosomal recombi-
Nierman WC, Havlak PH, Chen R, Durbin KJ, Egan A, Ren Y, Song XZ, nation events underlie the pericentric inversions of chimpanzee and
Li B, Liu Y, Qin X, Cawley S, Worley KC, Cooney AJ, DSouza LM, gorilla chromosomes homologous to human chromosome 16. Genome
Martin K, Wu JQ, Gonzalez-Garay ML, Jackson AR, Kalafus KJ, McLeod Res 15:12321242.
MP, Milosavljevic A, Virk D, Volkov A, Wheeler DA, Zhang Z, Bailey Goidts V, Armengol L, Schempp W, Conroy J, Nowak N, Muller S, Cooper
JA, Eichler EE, Tuzun E, Birney E, Mongin E, Ureta-Vidal A, Woodwark DN, Estivill X, Enard W, Szamalek JM, Hameister H, Kehrer-Sawatzki H.
C, Zdobnov E, Bork P, Suyama M, Torrents D, Alexandersson M, Trask 2006a. Identification of large-scale human-specific copy number
BJ, Young JM, Huang H, Wang H, Xing H, Daniels S, Gietzen D, differences by inter-species array comparative genomic hybridization.
Schmidt J, Stevens K, Vitt U, Wingrove J, Camara F, Mar Alba M, Abril Hum Genet 119:185198.
JF, Guigo R, Smit A, Dubchak I, Rubin EM, Couronne O, Poliakov A, Goidts V, Cooper DN, Armengol L, Schemp W, Conroy J, Estivill S,
Hubner N, Ganten D, Goesele C, Hummel O, Kreitler T, Lee YA, Monti Nowak N, Hameister H, Kehrer-Sawatzki H. 2006b. Complex pattern
J, Schulz H, Zimdahl H, Himmelbauer H, Lehrach H, Jacob HJ, of copy number variation at sites of segmental duplications: an
Bromberg S, Gullings-Handley J, Jensen-Seaman MI, Kwitek AE, Lazar important category of structural variation in the human genome. Hum
J, Pasko D, Tonellato PJ, Twigger S, Ponting CP, Duarte JM, Rice S, Genet 120:270284.
Goodstadt L, Beatson SA, Emes RD, Winter EE, Webber C, Brandt P, Goncalves I, Duret L, Mouchiroud D. 2000. Nature and structure
Nyakatura G, Adetobi M, Chiaromonte F, Elnitski L, Eswara P, Hardison of human genes that generate retropseudogenes. Genome Res 10:
RC, Hou M, Kolbe D, Makova K, Miller W, Nekrutenko A, Riemer C, 672678.
Schwartz S, Taylor J, Yang S, Zhang Y, Lindpaintner K, Andrews TD, Gonzalez E, Kulkarni H, Bolivar H, Mangano A, Sanchez R, Catano G,
Caccamo M, Clamp M, Clarke L, Curwen V, Durbin R, Eyras E, Searle Nibbs RJ, Freedman BI, Quinones MP, Bamshad MJ, Murthy KK, Rovin
SM, Cooper GM, Batzoglou S, Brudno M, Sidow A, Stone EA, Venter BH, Bradley W, Clark RA, Anderson SA, OConnell RJ, Agan BK,
JC, Payseur BA, Bourque G, Lopez-Otin C, Puente XS, Chakrabarti K, Ahuja SS, Bologna R, Sen L, Dolan MJ, Ahuja SK. 2005. The influence
Chatterji S, Dewey C, Pachter L, Bray N, Yap VB, Caspi A, Tesler G, of CCL3L1 gene-containing segmental duplications on HIV-1/AIDS
Pevzner PA, Haussler D, Roskin KM, Baertsch R, Clawson H, Furey TS, susceptibility. Science 307:14341440.
Hinrichs AS, Karolchik D, Kent WJ, Rosenbloom KR, Trumbower H, Goodier JL, Ostertag EM, Kazazian HH, Jr. 2000. Transduction of
Weirauch M, Cooper DN, Stenson PD, Ma B, Brent M, Arumugam M, 30 -flanking sequences is common in L1 retrotransposition. Hum Mol
Shteynberg D, Copley RR, Taylor MS, Riethman H, Mudunuri U, Genet 9:653657.
Peterson J, Guyer M, Felsenfeld A, Old S, Mockrin S, Collins F, Rat Goodman M. 1962. Evolution of the immunologic species specificity of
Genome Sequencing Project Consortium. 2004. Genome sequence of human serum proteins. Hum Biol 34:104150.
the Brown Norway rat yields insights into mammalian evolution. Nature Goodman M, Grossman LI, Wildman DE. 2005. Moving primate genomics
428:493521. beyond the chimpanzee genome. Trends Genet 21:511517.
Giglio S, Broman KW, Matsumoto N, Calvari V, Gimelli G, Neumann T, Greenberg AJ, Moran JR, Coyne JA, Wu CI. 2003. Ecological adaptation
Ohashi H, Voullaire L, Larizza D, Giorda R, Weber JL, Ledbetter DH, during incipient speciation revealed by precise gene replacement.
Zuffardi O. 2001. Olfactory receptor-gene clusters, genomic-inversion Science 302:17541757.
polymorphisms, and common chromosome rearrangements. Am J Hum Greenberg AJ, Moran JR, Fang S, Wu CI. 2006. Adaptive loss of an old
Genet 68:874883. duplicated gene during incipient speciation. Mol Biol Evol 23:401410.

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 123

Grimes BR, Rhoades AA, Willard HF. 2002. Alpha-satellite DNA and Hasegawa M, Kishino H, Yano T. 1987. Mans place in Hominoidea as
vector composition influence rates of human artificial chromosome inferred from molecular clocks of DNA. J Mol Evol 26:132147.
formation. Mol Ther 5:798805. Hayakawa T, Satta Y, Gagneux P, Varki A, Takahata N. 2001. Alu-mediated
Gross M, Starke H, Trifonov V, Claussen U, Liehr T, Weise A. 2006. A inactivation of the human CMP-N-acetylneuraminic acid hydroxylase
molecular cytogenetic study of chromosome evolution in chimpanzee. gene. Proc Natl Acad Sci USA 98:1139911404.
Cytogenet Genome Res 112:6775. Hayakawa T, Angata T, Lewis AL, Mikkelsen TS, Varki NM, Varki A.
Grus WE, Shi P, Zhang YP, Zhang J. 2005. Dramatic variation of the 2005. A human-specific gene in microglia. Science 309:1693.
vomeronasal pheromone receptor gene repertoire among five orders of Hedges DJ, Callinan PA, Cordaux R, Xing J, Barnes E, Batzer MA. 2004.
placental and marsupial mammals. Proc Natl Acad Sci USA 102: Differential Alu mobilization and polymorphism among the human and
57675772. chimpanzee lineages. Genome Res 14:10681075.
Gu JJ, Spychala J, Mitchell BS. 1997. Regulation of the human inosine Heissig F, Krause J, Bryk J, Khaitovich P, Enard W, Paabo S. 2005.
monophosphate dehydrogenase type I gene. Utilization of alternative Functional analysis of human and chimpanzee promoters. Genome Biol
promoters. J Biol Chem 272:44584466. 6:R57.
Gu J, Gu X. 2003. Induced gene expression in human brain after the split Hellmann I, Zollner S, Enard W, Ebersberger I, Nickel B, Paabo S. 2003a.
from chimpanzee. Trends Genet 19:6365. Selection on human genes as revealed by comparisons to chimpanzee
Gupta PK, Adamtziki E, Budde U, Jaiprakash M, Kumar H, Harbeck-Seu cDNA. Genome Res 13:831837.
A, Kannan M, Oyen F, Obser T, Wedekind I, Saxena R, Schneppenheim Hellmann I, Ebersberger I, Ptak SE, Paabo S, Przeworski M. 2003b. A
R. 2005. Gene conversions are a common cause of von Willebrand neutral explanation for the correlation of diversity with recombination
disease. Br J Haematol 130:752758. rates in humans. Am J Hum Genet 72:15271535.
Guy J, Spalluto C, McMurray A, Hearn T, Crosier M, Viggiano L, Miolla V, Henikoff S, Ahmad K, Malik HS. 2001. The centromere paradox: stable
Archidiacono N, Rocchi M, Scott C, Lee PA, Sulston J, Rogers J, Bentley inheritance with rapidly evolving DNA. Science 293:10981102.
D, Jackson MS. 2000. Genomic sequence and transcriptional profile of Henikoff S, Dalal Y. 2005. Centromeric chromatin: what makes it unique?
the boundary between pericentromeric satellites and genes on human Curr Opin Genet Dev 15:177184.
chromosome arm 10q. Hum Mol Genet 9:20292042. Hinds DA, Kloek AP, Jen M, Chen X, Frazer KA. 2006. Common deletions
Guy J, Hearn T, Crosier M, Mudge J, Viggiano L, Koczan D, Thiesen HJ, and SNPs are in linkage disequilibrium in the human genome. Nat
Bailey JA, Horvath JE, Eichler EE, Earthrowl ME, Deloukas P, French L, Genet 38:8285.
Rogers J, Bentley D, Jackson MS. 2003. Genomic sequence and Hirotsune S, Yoshida N, Chen A, Garrett L, Sugiyama F, Takahashi S,
transcriptional profile of the boundary between pericentromeric satellites Yagami K, Wynshaw-Boris A, Yoshiki A. 2003. An expressed pseudogene
and genes on human chromosome arm 10p. Genome Res 13:159172. regulates the messenger-RNA stability of its homologous coding gene.
Gygi SP, Rochon Y, Franza BR, Aebersold R. 1999. Correlation between Nature 423:9196.
protein and mRNA abundance in yeast. Mol Cell Biol 19:17201730. Ho HJ, Ray DA, Salem AH, Myers JS, Batzer MA. 2005. Straightening out
Haaf T, Willard HF. 1997. Chromosome-specific alpha-satellite DNA from the LINEs: LINE-1 orthologous loci. Genomics 85:201207.
the centromere of chimpanzee chromosome 4. Chromosoma 106: Hollox EJ, Armour JA, Barber JC. 2003. Extensive normal copy number
226232. variation of a beta-defensin antimicrobial-gene cluster. Am J Hum Genet
Hacia JG, Fan JB, Ryder O, Jin L, Edgemon K, Ghandour G, Mayer RA, 73:591600.
Sun B, Hsie L, Robbins CM, Brody LC, Wang D, Lander ES, Lipshutz R, Holmquist GP. 1992. Chromosome bands, their chromatin flavors, and
Fodor SP, Collins FS. 1999. Determination of ancestral alleles for human their functional features. Am J Hum Genet 51:1737.
single-nucleotide polymorphisms using high-density oligonucleotide Hoogendoorn B, Coleman SL, Guy CA, Smith K, Bowen T, Buckland PR,
arrays. Nat Genet 22:164167. ODonovan MC. 2003. Functional analysis of human promoter
Hahn MW, Rockman MV, Soranzo N, Goldstein DB, Wray GA. 2004. polymorphisms. Hum Mol Genet 12:22492254.
Population genetic and phylogenetic evidence for positive selection on Horai S, Satta Y, Hayasaka K, Kondo R, Inoue T, Ishida T, Hayashi S,
regulatory mutations at the factor VII locus in humans. Genetics 167: Takahata N. 1992. Mans place in Hominoidea revealed by mitochon-
867877. drial DNA genealogy. J Mol Evol 35:3243.
Hahn Y, Lee B. 2005. Identification of nine human-specific frameshift Horvath JE, Viggiano L, Loftus BJ, Adams MD, Archidiacono N, Rocchi M,
mutations by comparative analysis of the human and the chimpanzee Eichler EE. 2000. Molecular structure and evolution of an alpha satellite/
genome sequences. Bioinformatics 21(Suppl 1):i186i194. non-alpha satellite junction at 16p11. Hum Mol Genet 9:113123.
Hahn MW, De Bie T, Stajich JE, Nguyen C, Cristianini N. 2005. Estimating Horvath JE, Gulden CL, Bailey JA, Yohn C, McPherson JD, Prescott A, Roe
the tempo and mode of gene family evolution from comparative genomic BA, de Jong PJ, Ventura M, Misceo D, Archidiacono N, Zhao S, Schwartz
data. Genome Res 15:11531160. S, Rocchi M, Eichler EE. 2003. Using a pericentromeric interspersed
Hahn Y, Lee B. 2006. Human-specific nonsense mutations identified by repeat to recapitulate the phylogeny and expansion of human
genome sequence comparisons. Hum Genet 119:169178. centromeric segmental duplications. Mol Biol Evol 20:14631479.
Hamann J, Kwakkenbos MJ, de Jong EC, Heus H, Olsen AS, van Lier RA. Horvath JE, Gulden CL, Vallente RU, Eichler MY, Ventura M, McPherson
2003. Inactivation of the EGF-TM7 receptor EMR4 after the Pan-Homo JD, Graves TA, Wilson RK, Schwartz S, Rocchi M, Eichler EE. 2005.
divergence. Eur J Immunol 33:13651371. Punctuated duplication seeding events during the evolution of human
Hamilton AT, Huntley S, Tran-Gyamfi M, Baggott DM, Gordon L, chromosome 2p11. Genome Res 15:914927.
Stubbs L. 2006. Evolutionary expansion and divergence in the Hsieh WP, Chu TM, Wolfinger RD, Gibson G. 2003. Mixed-model
ZNF91 subfamily of primate-specific zinc finger genes. Genome Res 16: reanalysis of primate data suggests tissue and species biases in
584594. oligonucleotide-based gene expression profiles. Genetics 165:747757.
Han K, Sen SK, Wang J, Callinan PA, Lee J, Cordaux R, Liang P, Batzer Huang CH, Xie SS, Socha W, Blumenfeld OO. 1995. Sequence
MA. 2005. Genomic rearrangements by LINE-1 insertion-mediated diversification and exon inactivation in the glycophorin A gene family
deletion in the human and chimpanzee lineages. Nucleic Acids Res 33: from chimpanzee to human. J Mol Evol 41:478486.
40404052. Huby T, Dachet C, Lawn RM, Wickings J, Chapman MJ, Thillet J. 2001.
Hancock JM. 2005. Gene factories, microfunctionalization and the Functional analysis of the chimpanzee and human apo(a) promoter
evolution of gene families. Trends Genet 21:591595. sequences: identification of sequence variations responsible for elevated
Hansen TF, Carter AJ, Chiu CH. 2000. Gene conversion may aid adaptive transcriptional activity in chimpanzee. J Biol Chem 276:2220922214.
peak shifts. J Theor Biol 207:495511. Hughes JF, Skaletsky H, Pyntikova T, Minx PJ, Graves T, Rozen S, Wilson
Harrington JJ, Van Bokkelen G, Mays RW, Gustashaw K, Willard HF. 1997. RK, Page DC. 2005. Conservation of Y-linked genes during human
Formation of de novo centromeres and construction of first-generation evolution revealed by comparative sequencing in chimpanzee. Nature
human artificial microchromosomes. Nat Genet 15:345355. 437:100103.

Human Mutation DOI 10.1002/humu


124 HUMAN MUTATION 28(2), 99^130, 2007

Hurles M. 2004. Gene duplication: the genomic trade in spare parts. PLoS Kehrer-Sawatzki H, Sandig C, Chuzhanova N, Goidts V, Szamalek JM,
Biol 2:E206. Tanzer S, Mu ller S, Platzer M, Cooper DN, Hameister H. 2005a.
Huttley GA, Easteal S, Southey MC, Tesoriero A, Giles GG, McCredie Breakpoint analysis of the pericentric inversion distinguishing human
MR, Hopper JL, Venter DJ. 2000. Adaptive evolution of the tumour chromosome 4 from the homologous chromosome in the chimpanzee
suppressor BRCA1 in humans and chimpanzees. Australian Breast (Pan troglodytes). Hum Mut 25:4555.
Cancer Family Study. Nat Genet 25:410413. Kehrer-Sawatzki H, Szamalek JM, Tanzer S, Platzer M, Hameister H.
Iafrate AJ, Feuk L, Rivera MN, Listewnik ML, Donahoe PK, Qi Y, Scherer 2005b. Molecular characterization of the pericentric inversion of
SW, Lee C. 2004. Detection of large-scale variation in the human chimpanzee chromosome 11 homologous to human chromosome 9.
genome. Nat Genet 36:949951. Genomics 85:542550.
IJdo JW, Baldini A, Ward DC, Reeders ST, Wells RA. 1991. Origin of Kehrer-Sawatzki H, Sandig CA, Goidts V, Hameister H. 2005c. Breakpoint
human chromosome 2: an ancestral telomere-telomere fusion. Proc Natl analysis of the pericentric inversion between chimpanzee chromosome
Acad Sci USA 88:90519055. 10 and the homologous chromosome 12 in humans. Cytogenet Genome
Ikeno M, Grimes B, Okazaki T, Nakano M, Saitoh K, Hoshino H, McGill Res 108:9197.
NI, Cooke H, Masumoto H. 1998. Construction of YAC-based Keightley PD, Lercher MJ, Eyre-Walker A. 2005. Evidence for widespread
mammalian artificial chromosomes. Nat Biotechnol 16:431439. degradation of gene control regions in hominid genomes. PLoS
Irie A, Koyama S, Kozutsumi Y, Kawasaki T, Suzuki A. 1998. The Biol 3:e42.
molecular basis for the absence of N-glycolylneuraminic acid in humans. Kelley J, Walter L, Trowsdale J. 2005. Comparative genomics of major
J Biol Chem 273:1586615871. histocompatibility complexes. Immunogenetics 56:683695.
Jackson MS, Rocchi M, Thompson G, Hearn T, Crosier M, Guy J, Kirk D, Kennedy GC, Matsuzaki H, Dong S, Liu WM, Huang J, Liu G, Su X, Cao
Mulligan L, Ricco A, Piccininni S, Marzella R, Viggiano L, Archidiacono M, Chen W, Zhang J, Liu W, Yang G, Di X, Ryder T, He Z, Surti U,
N. 1999. Sequences flanking the centromere of human chromosome 10 Phillips MS, Boyce-Jacino MT, Fodor SP, Jones KW. 2003. Large-scale
are a complex patchwork of arm-specific sequences, stable duplications genotyping of complex DNA. Nat Biotechnol 21:12331237.
and unstable sequences with homologies to telomeric and other Khaitovich P, Weiss G, Lachmann M, Hellmann I, Enard W, Muetzel B,
centromeric locations. Hum Mol Genet 8:205215. Wirkner U, Ansorge W, Paabo S. 2004. A neutral model of transcriptome
Jackson AP, Eastwood H, Bell SM, Adu J, Toomes C, Carr IM, Roberts E, evolution. PLoS Biol 2:e132.
Hampshire DJ, Crow YJ, Mighell AJ, Karbani G, Jafri H, Rashid Y, Khaitovich P, Hellmann I, Enard W, Nowick K, Leinweber M, Franz H,
Mueller RF, Markham AF, Woods CG. 2002. Identification of Weiss G, Lachmann M, Paabo S. 2005a. Parallel patterns of evolution in
microcephalin, a protein implicated in determining the size of the the genomes and transcriptomes of humans and chimpanzees. Science
human brain. Am J Hum Genet 71:136142. 309:18501854.
Jaeger EE, Bontrop RE, Parham P, Wickings EJ, Kadwell M, Lanchbury JS. Khaitovich P, Paabo S, Weiss G. 2005b.Toward a neutral evolutionary
1998. Characterization of chimpanzee TCRV gene polymorphism: how model of gene expression. Genetics 170:929939.
old are human TCRV alleles? Immunogenetics 47:115123. Khan H, Smit A, Boissinot S. 2006. Molecular evolution and tempo of
Jaruzelska J, Zietkiewicz E, Labuda D. 1999. Is selection responsible for the amplification of human LINE-1 retrotransposons since the origin of
low level of variation in the last intron of the ZFY locus? Mol Biol Evol primates. Genome Res 16:7887.
16:16331640. Kim JH, Yu CY, Bailey A, Hardison R, Shen CK. 1989. Unique sequence
Jensen-Seaman MI, Li WH. 2003. Evolution of the hominoid semenogelin organization and erythroid cell-specific nuclear factor-binding of
genes, the major proteins of ejaculated semen. J Mol Evol 57:261270. mammalian theta 1 globin promoters. Nucleic Acids Res 17:56875700.
Jensen-Seaman MI, Furey TS, Payseur BA, Lu Y, Roskin KM, Chen CF, Kimura M. 1968. Evolutionary rate at the molecular level. Nature
Thomas MA, Haussler D, Jacob HJ. 2004. Comparative recombination 217:624626.
rates in the rat, mouse, and human genomes. Genome Res 14:528538. King MC, Wilson AC. 1975. Evolution at two levels in humans and
Jern P, Sperber GO, Blomberg J. 2006. Divergent patterns of recent chimpanzees. Science 188:107116.
retroviral integrations in the human and chimpanzee genomes: probable Kitano T, Liu YH, Ueda S, Saitou N. 2004. Human-specific amino
transmissions between other primates and chimpanzees. J Virol 80: acid changes found in 103 protein-coding genes. Mol Biol Evol 21:
13671375. 936944.
Ji Y, Eichler EE, Schwartz S, Nicholls RD. 2000. Structure of chromosomal Kivisild T, Shen P, Wall DP, Do B, Sung R, Davis K, Passarino G, Underhill
duplicons and their role in mediating human genomic disorders. Genome PA, Scharfe C, Torroni A, Scozzari R, Modiano D, Coppa A, de Knijff P,
Res 10:597610. Feldman M, Cavalli-Sforza LL, Oefner PJ. 2006. The role of selection
Jobling MA, Tyler-Smith C. 2003. The human Y chromosome: an in the evolution of human mitochondrial genomes. Genetics 172:
evolutionary marker comes of age. Nat Rev Genet 4:598612. 373387.
Johnson ME, Viggiano L, Bailey JA, Abdul-Rauf M, Goodwin G, Rocchi M, Kobayashi K, Nakahori Y, Miyake M, Matsumura K, Kondo-Iida E,
Eichler EE. 2001. Positive selection of a gene family during the Nomura Y, Segawa M, Yoshioka M, Saito K, Osawa M, Hamano K,
emergence of humans and African apes. Nature 413:514519. Sakakihara Y, Nonaka I, Nakagome Y, Kanazawa I, Nakamura Y,
Jury JA, Frayne J, Hall L. 1998. Sequence analysis of a variety of primate Tokunaga K, Toda T. 1998. An ancient retrotransposal insertion causes
fertilin alpha genes: evidence for non-functional genes in the gorilla and Fukuyama-type congenital muscular dystrophy. Nature 394:388392.
man. Mol Reprod Dev 51:9297. Korneev SA, Park JH, OShea M. 1999. Neuronal expression of neural
Kaessmann H, Heissig F, von Haeseler A, Paabo S. 1999. DNA sequence nitric oxide synthase (nNOS) protein is suppressed by an antisense RNA
variation in a non-coding region of low recombination on the human X transcribed from an NOS pseudogene. J. Neurosci 19:77117720.
chromosome. Nat Genet 22:7881. Kouprina N, Pavlicek A, Mochida GH, Solomon G, Gersch W, Yoon YH,
Kaessmann H, Wiebe V, Weiss G, Paabo S. 2001. Great ape DNA Collura R, Ruvolo M, Barrett JC, Woods CG, Walsh CA, Jurka J,
sequences reveal a reduced diversity and an expansion in humans. Nat Larionov V. 2004a. Accelerated evolution of the ASPM gene controlling
Genet 27:155156. brain size begins prior to human brain expansion. PLoS Biol 2:e126.
Karaman MW, Houck ML, Chemnick LG, Nagpal S, Chawannakul D, Kouprina N, Mullokandov M, Rogozin IB, Collins NK, Solomon G, Otstot
Sudano D, Pike BL, Ho VV, Ryder OA, Hacia JG. 2003. Comparative J, Risinger JI, Koonin EV, Barrett JC, Larionov V. 2004b. The SPANX
analysis of gene-expression patterns in human and African great ape gene family of cancer/testis-specific antigens: rapid evolution and
cultured fibroblasts. Genome Res 13:16191630. amplification in African great apes and hominids. Proc Natl Acad Sci
Kehrer-Sawatzki H, Schreiner B, Tanzer S, Platzer M, Mu ller S, Hameister USA 101:30773082.
H. 2002. Molecular characterization of the pericentric inversion that Kriek M, White SJ, Szuhai K, Knijnenburg J, van Ommen GJ, den Dunnen
causes differences between chimpanzee chromosome 19 and human JT, Breuning MH. 2006. Copy number variation in regions flanked (or
chromosome 17. Am J Hum Genet 71:375388. unflanked) by duplicons among patients with developmental delay

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 125

and/or congenital malformations; detection of reciprocal and partial T, Lara M, Lee W, Leger JP, Lennon N, Leuper L, LeVine S, Liu J, Liu X,
Williams-Beuren duplications. Eur J Hum Genet 14:180189. Lokyitsang Y, Lokyitsang T, Lui A, Macdonald J, Major J, Marabella R,
Kumar S, Hedges SB. 1998. A molecular timescale for vertebrate evolution. Maru K, Matthews C, McDonough S, Mehta T, Meldrim J, Melnikov A,
Nature 392:917920. Meneus L, Mihalev A, Mihova T, Miller K, Mittelman R, Mlenga V,
Kumar S, Filipski A, Swarna V, Walker A, Hedges SB. 2005. Placing Mulrain L, Munson G, Navidi A, Naylor J, Nguyen T, Nguyen N,
confidence limits on the molecular age of the human-chimpanzee Nguyen C, Nguyen T, Nicol R, Norbu N, Norbu C, Novod N, Nyima T,
divergence. Proc Natl Acad Sci USA 102:1884218847. Olandt P, ONeill B, ONeill K, Osman S, Oyono L, Patti C, Perrin D,
Kuroki Y, Toyoda A, Noguchi H, Taylor TD, Itoh T, Kim DS, Kim DW, Phunkhang P, Pierre F, Priest M, Rachupka A, Raghuraman S, Rameau
Choi SH, Kim IC, Choi HH, Kim YS, Satta Y, Saitou N, Yamada T, R, Ray V, Raymond C, Rege F, Rise C, Rogers J, Rogov P, Sahalie J,
Morishita S, Hattori M, Sakaki Y, Park HS, Fujiyama A. 2006. Settipalli S, Sharpe T, Shea T, Sheehan M, Sherpa N, Shi J, Shih D,
Comparative analysis of chimpanzee and human Y chromosomes unveils Sloan J, Smith C, Sparrow T, Stalker J, Stange-Thomann N,
complex evolutionary pathway. Nat Genet 38:158167. Stavropoulos S, Stone C, Stone S, Sykes S, Tchuinga P, Tenzing P,
Lahn BT, Page DC. 1999. Four evolutionary strata on the human X Tesfaye S, Thoulutsang D, Thoulutsang Y, Topham K, Topping I, Tsamla
chromosome. Science 286:964967. T, Vassiliev H, Venkataraman V, Vo A, Wangchuk T, Wangdi T, Weiand
Lai CS, Fisher SE, Hurst JA, Vargha-Khadem F, Monaco AP. 2001. A M, Wilkinson J, Wilson A, Yadav S, Yang S, Yang X, Young G, Yu Q,
forkhead-domain gene is mutated in a severe speech and language Zainoun J, Zembek L, Zimmer A, Lander ES. 2005. Genome sequence,
disorder. Nature 413:519523. comparative analysis and haplotype structure of the domestic dog.
Lamkanfi M, Declercq W, Kalai M, Saelens X, Vandenabeele P. 2002. Alice Nature 438:803819.
in caspase land. A phylogenetic analysis of caspases from worm to man. Liu G, Zhao S, Bailey JA, Sahinalp SC, Alkan C, Tuzun E, Green ED,
Cell Death Differ 9:358361. Eichler EE. 2003. Analysis of primate genomic variation reveals a repeat-
Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon driven expansion of the human genome. Genome Res 13:358368.
K, Dewar K, Doyle M, FitzHugh W. et al. 2001. Initial sequencing and Locke DP, Archidiacono N, Misceo D, Cardone MF, Deschamps S,
analysis of the human genome. Nature 409:860921. Roe B, Rocchi M, Eichler EE. 2003a. Refinement of a chimpanzee
Leakey MG, Feibel CS, McDougall I, Ward C, Walker A. 1998. New pericentric inversion breakpoint to a segmental duplication cluster.
specimens and confirmation of an early age for Australopithecus Genome Biol 4:R50.
anamensis. Nature 393:6266. Locke DP, Segraves R, Carbone L, Archidiacono N, Albertson DG, Pinkel
Lee C, Wevrick R, Fisher RB, Ferguson-Smith MA, Lin CC. 1997. Human D, Eichler EE. 2003b. Large-scale variation among human and great ape
centromeric DNAs. Hum Genet 100:291304. genomes determined by array comparative genomic hybridization.
Lehrer RI. 2004. Primate defensins. Nat Rev Microbiol 2:727738. Genome Res 13:347357.
Lercher MJ, Hurst LD. 2002. Human SNP variability and mutation rate are Locke DP, Jiang Z, Pertz LM, Misceo D, Archidiacono N, Eichler EE. 2005.
higher in regions of high recombination. Trends Genet 18:337340. Molecular evolution of the human chromosome 15 pericentromeric
Li WH, Tanimura M, Sharp PM. 1987. An evaluation of the molecular region. Cytogenet Genome Res 108:7382.
clock hypothesis using mammalian DNA sequences. J Mol Evol Long M, Betran E, Thornton K, Wang W. 2003. The origin of new genes:
25:330342. glimpses from the young and old. Nat Rev Genet 4:865875.
Li WH, Ellsworth DL, Krushkal J, Chang BH, Hewett-Emmett D. 1996. Lu J, Li WH, Wu CI. 2003. Comment on Chromosomal speciation and
Rates of nucleotide substitution in primates and rodents and the molecular divergence-accelerated evolution in rearranged chromo-
generation-time effect hypothesis. Mol Phylogenet Evol 5:182187. somes. Science 302:988.
Li WH, Gu Z, Wang H, Nekrutenko A. 2001. Evolutionary analyses of the Lu J, Wu CI. 2005. Weak selection revealed by the whole-genome
human genome. Nature 409:847849. comparison of the X chromosome and autosomes of human and
Li WH, Yi S, Makova K. 2002. Male-driven evolution. Curr Opin Genet chimpanzee. Proc Natl Acad Sci USA 102:40634067.
Dev 12:650656. Lupski JR. 1998. Genomic disorders: structural features of the genome can
Li WH, Yang J, Gu X. 2005. Expression divergence between duplicate lead to DNA rearrangements and human disease traits. Trends Genet
genes. Trends Genet 21:602607. 14:417422.
Liang H, Landweber LF. 2006. A genome-wide study of dual coding regions Lupski JR, Stankiewicz P. 2005. Genomic disorders: molecular mechanisms
in human alternatively spliced genes. Genome Res 16:190196. for rearrangements and conveyed phenotypes. PLoS Genet 1:e49.
Linardopoulou EV, Williams EM, Fan Y, Friedman C, Young JM, Trask BJ. Lynch M, Conery JS. 2000. The evolutionary fate and consequences of
2005. Human subtelomeres are hot spots of interchromosomal duplicate genes. Science 290:11511155.
recombination and segmental duplication. Nature 437:94100. Mahtani MM, Willard HF. 1998. Physical and genetic mapping of the
Lindblad-Toh K, Wade CM, Mikkelsen TS, Karlsson EK, Jaffe DB, Kamal human X chromosome centromere: repression of recombination.
M, Clamp M, Chang JL, Kulbokas EJ 3rd, Zody MC, Mauceli E, Xie X, Genome Res 8:100110.
Breen M, Wayne RK, Ostrander EA, Ponting CP, Galibert F, Smith DR, Maier AG, Duraisingh MT, Reeder JC, Patel SS, Kazura JW, Zimmerman
DeJong PJ, Kirkness E, Alvarez P, Biagi T, Brockman W, Butler J, Chin PA, Cowman AF. 2003. Plasmodium falciparum erythrocyte invasion
CW, Cook A, Cuff J, Daly MJ, DeCaprio D, Gnerre S, Grabherr M, Kellis through glycophorin C and selection for Gerbich negativity in human
M, Kleber M, Bardeleben C, Goodstadt L, Heger A, Hitte C, Kim L, populations. Nat Med 9:8792.
Koepfli KP, Parker HG, Pollinger JP, Searle SM, Sutter NB, Thomas R, Makova KD, Li WH. 2002. Strong male-driven evolution of DNA
Webber C, Baldwin J, Abebe A, Abouelleil A, Aftuck L, Ait-Zahra M, sequences in humans and apes. Nature 416:624626.
Aldredge T, Allen N, An P, Anderson S, Antoine C, Arachchi H, Aslam Makova KD, Li WH. 2003. Divergence in the spatial pattern of gene
A, Ayotte L, Bachantsang P, Barry A, Bayul T, Benamara M, Berlin A, expression between human duplicate genes. Genome Res 13:16381645.
Bessette D, Blitshteyn B, Bloom T, Blye J, Boguslavskiy L, Bonnet C, Maresco DL, Chang E, Theil KS, Francke U, Anderson CL. 1996. The
Boukhgalter B, Brown A, Cahill P, Calixte N, Camarata J, Cheshatsang three genes of the human FCGR1 gene family encoding Fc gamma RI
Y, Chu J, Citroen M, Collymore A, Cooke P, Dawoe T, Daza R, Decktor flank the centromere of chromosome 1 at 1p12 and 1q21. Cytogenet
K, DeGray S, Dhargay N, Dooley K, Dooley K, Dorje P, Dorjee K, Dorris Cell Genet 73:157163.
L, Duffey N, Dupes A, Egbiremolen O, Elong R, Falk J, Farina A, Faro S, Maresco DL, Blue LE, Culley LL, Kimberly RP, Anderson CL, Theil KS.
Ferguson D, Ferreira P, Fisher S, FitzGerald M, Foley K, Foley C, Franke 1998. Localization of FCGR1 encoding Fcgamma receptor class I in
A, Friedrich D, Gage D, Garber M, Gearin G, Giannoukos G, Goode T, primates: molecular evidence for two pericentric inversions during the
Goyette A, Graham J, Grandbois E, Gyaltsen K, Hafez N, Hagopian D, evolution of human chromosome 1. Cytogenet Cell Genet 82:7174.
Hagos B, Hall J, Healy C, Hegarty R, Honan T, Horn A, Houde N, Marques AC, Dupanloup I, Vinckenbosch N, Reymond A, Kaessmann H.
Hughes L, Hunnicutt L, Husby M, Jester B, Jones C, Kamat A, Kanga B, 2005. Emergence of young human genes after a burst of retroposition in
Kells C, Khazanovich D, Kieu AC, Kisner P, Kumar M, Lance K, Landers primates. PLoS Biol 3:e357.

Human Mutation DOI 10.1002/humu


126 HUMAN MUTATION 28(2), 99^130, 2007

Marques-Bonet T, Caceres M, Bertranpetit J, Preuss TM, Thomas JW, Mills RE, Bennett EA, Iskow RC, Luttig CT, Tsui C, Pittard WS, Devine
Navarro A. 2004. Chromosomal rearrangements and the genomic SE. 2006. Recently mobilized transposons in the human and chimpanzee
distribution of gene-expression divergence in humans and chimpanzees. genomes. Am J Hum Genet 78:671679.
Trends Genet 20:524529. Mironov AA, Fickett JW, Gelfand MS. 1999. Frequent alternative splicing
Mars WM, Patmasiriwat P, Maity T, Huff V, Weil MM, Saunders GF. of human genes. Genome Res 9:12881293.
1995. Inheritance of unequal numbers of the genes encoding the Modrek B, Lee CJ. 2003. Alternative splicing in the human, mouse and rat
human neutrophil defensins HP-1 and HP-3. J Biol Chem 270: genomes is associated with an increased frequency of exon creation and/
3037130376. or loss. Nat Genet 34:1780.
Martin CL, Wong A, Gross A, Chung J, Fantes JA, Ledbetter DH. 2002. Moles A, Kieffer BL, DAmato FR. 2004. Deficit in attachment behavior in
The evolutionary origin of human subtelomeric homologiesor where mice lacking the mu-opioid receptor gene. Science 304:19831986.
the ends begin. Am J Hum Genet 70:972984. Monfouilloux S, Avet-Loiseau H, Amarger V, Balazs I, Pourcel C, Vergnaud
Martin MJ, Rayner JC, Gagneux P, Barnwell JW, Varki A. 2005. Evolution G. 1998. Recent human-specific spreading of a subtelomeric domain.
of human-chimpanzee differences in malaria susceptibility: relationship Genomics 51:165176.
to human genetic loss of N-glycolylneuraminic acid. Proc Natl Acad Sci Moran JV, DeBerardinis RJ, Kazazian HH Jr. 1999. Exon shuffling by L1
USA 102:1281912824. retrotransposition. Science 283:15301534.
Martinko JM, Vincek V, Klein D, Klein J. 1993. Primate ABO Morley M, Molony CM, Weber TM, Devlin JL, Ewens KG, Spielman RS,
glycotransferases: evidence for trans-species evolution. Immunogenetics Cheung VG. 2004. Genetic analysis of genome-wide variation in human
37:274278. gene expression. Nature 430:743747.
Marvanova M, Menager J, Bezard E, Bontrop RE, Pradier L, Wong G. 2003. Mountain JL, Cavalli-Sforza LL. 1994. Inference of human evolution
Microarray analysis of nonhuman primates: validation of experimental through cladistic analysis of nuclear DNA restriction polymorphisms.
models in neurological disorders. FASEB J 17:929931. Proc Natl Acad Sci USA 91:65156519.
Marzella R, Viggiano L, Miolla V, Storlazzi CT, Ricco A, Gentile E, Roberto Muchmore EA, Diaz S, Varki A. 1998. A structural difference between the
R, Surace C, Fratello A, Mancini M, Archidiacono N, Rocchi M. 2000. cell surfaces of humans and the great apes. Am J Phys Anthropol
Molecular cytogenetic resources for chromosome 4 and comparative 107:187198.
analysis of phylogenetic chromosome IV in great apes. Genomics 63: Muller S, Wienberg J. 2001. Bar-coding primate chromosomes: molecular
307313. cytogenetic screening for the ancestral hominoid karyotype. Hum Genet
Maston GA, Ruvolo M. 2002. Chorionic gonadotropin has a recent origin 109:8594.
within primates and an evolutionary history of selection. Mol Biol Evol Nadeau JH, Sankoff D. 1997. Comparable rates of gene loss and functional
19:320335. divergence after genome duplications early in vertebrate evolution.
Mathews LM, Chi SY, Greenberg N, Ovchinnikov I, Swergold GD. 2003. Genetics 147:12591266.
Large differences between LINE-1 amplification rates in the human and Nahon JL. 2003. Birth of human-specific genes during primate evolution.
chimpanzee lineages. Am J Hum Genet 72:739748. Genetica 118:193208.
Maxwell AI, Morrison GM, Dorin JR. 2003. Rapid sequence divergence in Nakagawa T, Zhu H, Morishima N, Li E, Xu J, Yankner BA, Yuan J. 2000.
mammalian beta-defensins by adaptive evolution. Mol Immunol Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and
40:413421. cytotoxicity by amyloid-beta. Nature 403:98103.
Maynard Smith J, Haigh J. 1974. The hitch-hiking effect of a favourable Navarro A, Barton NH. 2003. Chromosomal speciation and molecular
gene. Genet Res 23:2335. divergenceaccelerated evolution in rearranged chromosomes. Science
McCarroll SA, Hadnott TN, Perry GH, Sabeti PC, Zody MC, Barrett JC, 300:321324.
Dallaire S, Gabriel SB, Lee C, Daly MJ, Altshuler DM, The International Nei M, Gu X, Sitnikova T. 1997. Evolution by the birth-and-death process
HapMap Consortium. 2006. Common deletion polymorphisms in the in multigene families of the vertebrate immune system. Proc Natl Acad
human genome. Nat Genet 38:8692. Sci USA 94:77997806.
McConkey EH, Goodman M. 1997. A human genome evolution project is Nei M, Rooney AP. 2005. Concerted and birth-and-death evolution of
needed. Trends Genet 13:350351. multigene families. Annu Rev Genet 39:121152.
McEvoy SM, Maeda N. 1988. Complex events in the evolution of the Newman TL, Tuzun E, Morrison VA, Hayden KE, Ventura M,
haptoglobin gene cluster in primates. J Biol Chem 263:1574015747. McGrath SD, Rocchi M, Eichler EE. 2005. A genome-wide survey of
Mefford HC, Trask BJ. 2002. The complex structure and dynamic structural variation between human and chimpanzee. Genome Res 15:
evolution of human subtelomeres. Nat Rev Genet 3:91102. 13441356.
Mehan MR, Freimer NB, Ophoff RA. 2004. A genome-wide survey of Nguyen DH, Hurtado-Ziola N, Gagneux P, Varki A. 2006a. Loss of Siglec
segmental duplications that mediate common human genetic variation expression on T lymphocytes during human evolution. Proc Natl Acad
of chromosomal architecture. Hum Genomics 1:335344. Sci USA 103:77657770.
Mekel-Bobrov N, Gilbert SL, Evans PD, Vallender EJ, Anderson JR, Nguyen DQ, Webber C, Ponting CP. 2006b. Bias of selection on human
Hudson RR, Tishkoff SA, Lahn BT. 2005. Ongoing adaptive evolution copy-number variants. PLoS Genet 2:e20.
of ASPM, a brain size determinant in Homo sapiens. Science 309: Nickerson E, Nelson DL. 1998. Molecular definition of pericentric
17201722. inversion breakpoints occurring during the evolution of humans and
Meunier J, Duret L. 2004. Recombination drives the evolution of chimpanzees. Genomics 50:368372.
GC-content in the human genome. Mol Biol Evol 21:984990. Nielsen R, Bustamante C, Clark AG, Glanowski S, Sackton TB, Hubisz MJ,
Mikkelsen TS, Hillier LW, Eichler EE, Zody MC, Jaffe DB, Yang S-P, Enard Fledel-Alon A, Tanenbaum DM, Civello D, White TJ, Sninsky J, Adams
W, Hellmann I, Lindblad-Toh K, Altheide TK, Archidiacono N, Bork P, MD, Cargill M. 2005. A scan for positively selected genes in the genomes
Butler J, Chang JL, Cheng Z, Chinwalla AT, deJong P, Delehaunty KD, of humans and chimpanzees. PLoS Biol 3:e170.
Fronick CC, Fulton LL, Gilad Y, Glusman G, Gnerre S, Graves TA, Nielsen R. 2006. Comparative genomics: difference of expression. Nature
Hayakawa T, Hayden KE, Huang X, Ji H, Kent WJ, King M-C, Kulbokas 440:161.
EJ, Lee MK, Liu G, Lopez-Otin C, Makova KD, Man O, Mardis ER, Nishant KT, Rao MR. 2006. Molecular features of meiotic recombination
Mauceli E, Miner TL, Nash WE, Nelson JO, Paabo S, Patterson NJ, Poh hot spots. Bioessays 28:4556.
CS, Pollard KS, Pru fer K, Puente XS, Reich D, Rocchi M, Rosenbloom Novembre FJ, Saucier M, Anderson DC, Klumpp SA, ONeil SP, Brown CR
K, Ruvolo M, Richter DJ, Schaffner SF, Smit AFA, Smith SM, Suyama 2nd, Hart CE, Guenthner PC, Swenson RB, McClure HM. 1997.
M, Taylor J, Torrents D, Tuzun E, Varki A, Velasco G, Ventura M, Wallis Development of AIDS in a chimpanzee infected with human
JW, Wend MC, Wilson RK, Lander ES, Waterston RJ. 2005. Initial immunodeficiency virus type 1. J Virol 71:40864091.
sequence of the chimpanzee genome and comparison with the human Nusbaum C, Mikkelsen TS, Zody MC, Asakawa S, Taudien S, Garber M,
genome. Nature 437:6987. Kodira CD, Schueler MG, Shimizu A, Whittaker CA, Chang JL, Cuomo

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 127

CA, Dewar K, FitzGerald MG, Yang X, Allen NR, Anderson S, Asakawa Ptak SE, Roeder AD, Stephens M, Gilad Y, Paabo S, Przeworski M. 2004.
T, Blechschmidt K, Bloom T, Borowsky ML, Butler J, Cook A, Corum B, Absence of the TAP2 human recombination hotspot in chimpanzees.
DeArellano K, DeCaprio D, Dooley KT, Dorris L 3rd, Engels R, PLoS Biol 2:e155.
Glockner G, Hafez N, Hagopian DS, Hall JL, Ishikawa SK, Jaffe DB, Ptak SE, Hinds DA, Koehler K, Nickel B, Patil N, Ballinger DG, Przeworski
Kamat A, Kudoh J, Lehmann R, Lokitsang T, Macdonald P, Major JE, M, Frazer KA, Paabo S. 2005. Fine-scale recombination patterns differ
Matthews CD, Mauceli E, Menzel U, Mihalev AH, Minoshima S, between chimpanzees and humans. Nat Genet 37:429434.
Murayama Y, Naylor JW, Nicol R, Nguyen C, OLeary SB, ONeill K, Puente XS, Gutierrez-Fernandez A, Ordonez GR, Hillier LW, Lopez-Otin
Parker SC, Polley A, Raymond CK, Reichwald K, Rodriguez J, Sasaki T, C. 2005. Comparative genomic analysis of human and chimpanzee
Schilhabel M, Siddiqui R, Smith CL, Sneddon TP, Talamas JA, Tenzin P, proteases. Genomics 86:638647.
Topham K, Venkataraman V, Wen G, Yamazaki S, Young SK, Zeng Q, Puente XS, Velasco G, Gutierrez-Fernandez A, Bertranpetit J, King MC,
Zimmer AR, Rosenthal A, Birren BW, Platzer M, Shimizu N, Lander ES. Lopez-Otin C. 2006. Comparative analysis of cancer genes in the human
2006. DNA sequence and analysis of human chromosome 8. Nature and chimpanzee genomes. BMC Genomics 7:15.
439:331335. Regnier V, Meddeb M, Lecointre G, Richard F, Duverger A, Nguyen VC,
Ohno S. 1970. Evolution by gene duplication. London: George Allen and Dutrillaux B, Bernheim A, Danglot G. 1997. Emergence and scattering
Unwin. p.160. of multiple neurofibromatosis (NF1)-related sequences during hominoid
Ohshima M, Hattori T, Yada T, Gojobori Y, Sakaki Y, Okada N. 2003. evolution suggest a process of pericentromeric interchromosomal
Whole-genome screening indicates a possible burst of formation of transposition. Hum Mol Genet 6:916.
processed pseudogenes and Alu repeats by particular L1 subfamilies in Resch A, Xing Y, Alekseyenko A, Modrek B, Lee C. 2004. Evidence for a
ancestral primates. Genome Biol 4:R74. subpopulation of conserved alternative splicing events under selection
Olson MV. 1999. When less is more: gene loss as an engine of evolutionary pressure for protein reading frame preservation. Nucleic Acids Res
change. Am J Hum Genet 64:1823. 32:12611269.
Olson MV, Varki A. 2003. Sequencing the chimpanzee genome: insights Ricchetti M, Tekaia F, Dujon B. 2004. Continued colonization of the
into human evolution and disease. Nat Rev Genet 4:2028. human genome by mitochondrial DNA. PLoS Biol 2:E273.
Ono M, Kawakami M, Takezawa T. 1987. A novel human nonviral Rice WR. 1987. Genetic hitchhiking and the evolution of reduced genetic
retroposon derived from an endogenous retrovirus. Nucleic Acids Res activity of the Y sex chromosome. Genetics 116:161167.
15:87258737. Rieseberg LH, Livingstone K. 2003. Evolution. Chromosomal speciation in
Orti R, Potier MC, Maunoury C, Prieur M, Creau N, Delabar JM. 1998. primates. Science 300:267268.
Conservation of pericentromeric duplications of a 200-kb part Roberts E, Hampshire DJ, Pattison L, Springell K, Jafri H, Corry P, Mannon
of the human 21q22.1 region in primates. Cytogenet Cell Genet 83: J, Rashid Y, Crow Y, Bond J, Woods CG. 2002. Autosomal recessive
262265. primary microcephaly: an analysis of locus heterogeneity and phenotypic
Osada N, Wu CI. 2005. Inferring the mode of speciation from genomic variation. J Med Genet 39:718721.
data: a study of the great apes. Genetics 169:259264. Rochette CF, Gilbert N, Simard LR. 2001. SMN gene duplication and the
Osborne LR, Li M, Pober B, Chitayat D, Bodurtha J, Mandel A, Costa T, emergence of the SMN2 gene occurred in distinct hominids: SMN2 is
Grebe T, Cox S, Tsui LC, Scherer SW. 2001. A 1.5 million-base pair unique to Homo sapiens. Hum Genet 108:255266.
inversion polymorphism in families with Williams-Beuren syndrome. Nat Rockman MV, Hahn MW, Soranzo N, Zimprich F, Goldstein DB, Wray GA.
Genet 29:321325. 2005. Ancient and recent positive selection transformed opioid cis-
Ostertag EM, Goodier JL, Zhang Y, Kazazian HH, Jr. 2003. SVA elements regulation in humans. PLoS Biol 3:e387.
are nonautonomous retrotransposons that cause disease in humans. Am Rodgers RJ, Cooper SJ, editors. 1988. Endorphins, opiates and behavioural
J Hum Genet 73:14441451. processes. New York: Wiley. p 361.
Otieno AC, Carter AB, Hedges DJ, Walker JA, Ray DA, Garber RK, Romano CM, Ramalho RF, Zanotto PM. 2006. Tempo and mode of ERV-K
Anders BA, Stoilova N, Laborde ME, Fowlkes JD, Huang CH, Perodeau evolution in human and chimpanzee genomes. Arch Virol July 10; [Epub
B, Batzer MA. 2004. Analysis of the human Alu Ya-lineage. J Mol Biol ahead of print].
342:109118. Rose SD, MacDonald RJ. 1997. Evolutionary silencing of the human
Patterson N, Richter DJ, Gnerre S, Lander ES, Reich D. 2006. Genetic elastase I gene (ELA1). Hum Mol Genet 6:897903.
evidence for complex speciation of humans and chimpanzees. Nature Rouquier S, Friedman C, Delettre C, van den Engh G, Blancher A,
441:11031108. Crouau-Roy B, Trask BJ, Giorgi D. 1998. A gene recently inactivated in
Paulding CA, Ruvolo M, Haber DA. 2003. The Tre2 (USP6) oncogene is a human defines a new olfactory receptor family in mammals. Hum Mol
hominoid-specific gene. Proc Natl Acad Sci USA 100:25072511. Genet 7:13371345.
Pavlicek A, Noskov VN, Kouprina N, Barrett JC, Jurka J, Larionov V. 2004. Royle NJ, Baird DM, Jeffreys AJ. 1994. A subterminal satellite located
Evolution of the tumor suppressor BRCA1 locus in primates: implica- adjacent to telomeres in chimpanzees is absent from the human genome.
tions for cancer predisposition. Hum Mol Genet 13:27372751. Nat Genet 6:5256.
Pavlicek A, Gentles AJ, Paces J, Paces V, Jurka J. 2006. Retroposition of Rozen S, Skaletsky H, Marszalek JD, Minx PJ, Cordum HS, Waterston RH,
processed pseudogenes: the impact of RNA stability and translational Wilson RK, Page DC. 2003. Abundant gene conversion between arms of
control. Trends Genet 22:6973. palindromes in human and ape Y chromosomes. Nature 423:873876.
Perry GH, Tchinda J, McGrath SD, Zhang J, Picker SR, Caceres AM, Rudd MK, Willard HF. 2004. Analysis of the centromeric regions of the
Iafrate AJ, Tyler-Smith C, Scherer SW, Eichler EE, Stone AC, Lee C. human genome assembly. Trends Genet 20:529533.
2006. Hotspots for copy number variation in chimpanzees and humans. Rudd MK, Wray GA, Willard HF. 2006. The evolutionary dynamics
Proc Natl Acad Sci USA 103:80068011. of a-satellite. Genome Res 16:8896.
Pilpel Y, Sudarsanam P, Church GM. 2001. Identifying regulatory networks Sabeti PC, Schaffner SF, Fry B, Lohmueller J, Varilly P, Shamovsky O,
by combinatorial analysis of promoter elements. Nat Genet 29:153159. Palma A, Mikkelsen TS, Altshuler D, Lander ES. 2006. Positive natural
Polavarapu N, Bowen NJ, McDonald JF. 2006. Identification, characteriza- selection in the human lineage. Science 312:16141620.
tion and comparative genomics of chimpanzee endogenous retroviruses. Saccone S, De Sario A, Della Valle G, Bernardi G. 1992. The highest gene
Genome Biol 7:R51. concentrations in the human genome are in telomeric bands of
Porter AC, Farr CJ. 2004. Topoisomerase II: untangling its contribution at metaphase chromosomes. Proc Natl Acad Sci USA 89:49134917.
the centromere. Chromosome Res 12:569583. Saleh M, Vaillancourt JP, Graham RK, Huyck M, Srinivasula SM, Alnemri
Preuss TM, Caceres M, Oldham MC, Geschwind DH. 2004. Human brain ES, Steinberg MH, Nolan V, Baldwin CT, Hotchkiss RS, Buchman TG,
evolution: insights from microarrays. Nat Rev Genet 5:850860. Zehnbauer BA, Hayden MR, Farrer LA, Roy S, Nicholson DW. 2004.
Prince VE, Pickett FB. 2002. Splitting pairs: the diverging fates of Differential modulation of endotoxin responsiveness by human caspase-
duplicated genes. Nat Rev Genet 3:827837. 12 polymorphisms. Nature 429:7579.

Human Mutation DOI 10.1002/humu


128 HUMAN MUTATION 28(2), 99^130, 2007

Salem AH, Myers JS, Otieno AC, Watkins WS, Jorde LB, Batzer MA. exonintron structure, composite retroposon, and breakpoint of gene
2003. LINE-1 preTa elements in the human genome. J Mol Biol 326: duplication. J Biol Chem 269:84668476.
11271146. Shepherd GM. 2004. The human sense of smell: are we better than we
Samonte RV, Eichler EE. 2002. Segmental duplications and the evolution think? PLoS Biol 2:e146.
of the primate genome. Nat Rev Genet 3:6572. Shi J, Xi H, Wang Y, Zhang C, Jiang Z, Zhang K, Shen Y, Jin L,
Samuelson LC, Wiebauer K, Snow CM, Meisler MH. 1990. Retroviral and Zhang K, Yuan W, Hua Q, Wang F, Xu S, Ren S, Xu S, Zhao G, Chen Z,
pseudogene insertion sites reveal the lineage of human salivary and Jin L, Huang W. 2003a. Divergence of the genes on human chromosome
pancreatic amylase genes from a single gene during primate evolution. 21 between human and other hominoids and variation of substitution
Mol Cell Biol 10:25132520. rates among transcription units. Proc Natl Acad Sci USA 100:
Saunier S, Calado J, Benessy F, Silbermann F, Heilig R, Weissenbach J, 83318336.
Antignac C. 2000. Characterization of the NPHP1 locus: mutational Shi P, Zhang J, Yang H, Zhang YP. 2003b. Adaptive diversification of
mechanism involved in deletions in familial juvenile nephronophthisis. bitter taste receptor genes in mammalian evolution. Mol Biol Evol 20:
Am J Hum Genet 66:778789. 805814.
Scherer SW, Gripp KW, Lucena J, Nicholson L, Bonnefont J-P, Perez-Jurado Shimada MK, Kim CG, Kitano T, Ferrell RE, Kohara Y, Saitou N. 2005.
LA, Osborne LR. 2005. Observation of a parental inversion variant in a Nucleotide sequence comparison of a chromosome rearrangement on
rare Williams-Beuren syndrome family with two affected children. Hum human chromosome 12 and the corresponding ape chromosomes.
Genet 117:383388. Cytogenet Genome Res 108:8390.
Schueler MG, Higgins AW, Rudd MK, Gustashaw K, Willard HF. 2001. Shin HD, Winkler C, Stephens JC, Bream J, Young H, Goedert JJ, OBrien
Genomic and genetic definition of a functional human centromere. TR, Vlahov D, Buchbinder S, Giorgi J, Rinaldo C, Donfield S,
Science 294:109115. Willoughby A, OBrien SJ, Smith MW. 2000. Genetic restriction of
Schueler MG, Dunn JM, Bird CP, Ross MT, Viggiano L, Rocchi M, Willard HIV-1 pathogenesis to AIDS by promoter alleles of IL10. Proc Natl Acad
HF, Green ED, NISC Comparative Sequencing Program. 2005. Sci USA 97:1446714472.
Progressive proximal expansion of the primate X chromosome Sikela JM. 2006. The jewels of our genome: the search for the genomic
centromere. Proc Natl Acad Sci USA 102:1056310568. changes underlying the evolutionarily unique capacities of the human
Sebat J, Lakshmi B, Troge J, Alexander J, Young J, Lundin P, Maner S, brain. PLoS Genet 2:e80.
Massa H, Walker M, Chi M, Navin N, Lucito R, Healy J, Hicks J, Ye K, Skaletsky H, Kuroda-Kawaguchi T, Minx PJ, Cordum HS, Hillier L, Brown
Reiner A, Gilliam TC, Trask B, Patterson N, Zetterberg A, Wigler M. LG, Repping S, Pyntikova T, Ali J, Bieri T, Chinwalla A, Delehaunty A,
2004. Large-scale copy number polymorphism in the human genome. Delehaunty K, Du H, Fewell G, Fulton L, Fulton R, Graves T, Hou SF,
Science 305:525528. Latrielle P, Leonard S, Mardis E, Maupin R, McPherson J, Miner T,
Semple CA, Rolfe M, Dorin JR. 2003. Duplication and selection in the Nash W, Nguyen C, Ozersky P, Pepin K, Rock S, Rohlfing T, Scott K,
evolution of primate beta-defensin genes. Genome Biol 4:R31. Schultz B, Strong C, Tin-Wollam A, Yang SP, Waterston RH, Wilson RK,
Semple CA, Maxwell A, Gautier P, Kilanowski FM, Eastwood H, Barran Rozen S, Page DC. 2003. The male-specific region of the human Y
PE, Dorin JR. 2005. The complexity of selection at the major primate chromosome is a mosaic of discrete sequence classes. Nature 423:
beta-defensin locus. BMC Evol Biol 5:32. 825837.
Sen SK, Han K, Wang J, Lee J, Wang H, Callinan PA, Dyer M, Small K, Iber J, Warren ST. 1997. Emerin deletion reveals a common X-
Cordaux R, Liang P, Batzer MA. 2006. Human genomic deletions chromosome inversion mediated by inverted repeats. Nat Genet 16:
mediated by recombination between Alu elements. Am J Hum Genet 9699.
79:4153. Smith NG, Webster MT, Ellegren H. 2002. Deterministic mutation rate
Sharon D, Glusman G, Pilpel Y, Khen M, Gruetzner F, Haaf T, Lancet D. variation in the human genome. Genome Res 12:13501356.
1999. Primate evolution of an olfactory receptor cluster: diversification Sonnenburg JL, Altheide TK, Varki A. 2004. A uniquely human
by gene conversion and recent emergence of pseudogenes. Genomics consequence of domain-specific functional adaptation in a sialic acid-
61:2436. binding receptor. Glycobiology 14:339346.
Sharp PM, Li WH. 1986. Codon usage in regulatory genes in Escherichia Spence JM, Critcher R, Ebersole TA, Valdivia MM, Earnshaw WC,
coli does not reflect selection for rare codons. Nucleic Acids Res 14: Fukagawa T, Farr CJ. 2002. Co-localization of centromere activity,
77377749. proteins and topoisomerase II within a subdomain of the major human X
Sharp AJ, Locke DP, McGrath SD, Cheng Z, Bailey JA, Vallente RU, Pertz alpha-satellite array. EMBO J 21:52695280.
LM, Clark RA, Schwartz S, Segraves R, Oseroff VV, Albertson DG, Stankiewicz P, Shaw CJ, Withers M, Inoue K, Lupski JR. 2004. Serial
Pinkel D, Eichler EE. 2005. Segmental duplications and copy number segmental duplications during primate evolution result in complex
variation in the human genome. Am J Hum Genet 77:7888. human genome architecture. Genome Res 14:22092220.
Shaw CJ, Lupski JR. 2004. Implications of human genome architecture for Stedman HH, Kozyak BW, Nelson A, Thesier DM, Su LT, Low DW, Bridges
rearrangement-based disorders: the genomic basis of disease. Hum Mol CR, Shrager JB, Minugh-Purvis N, Mitchell MA. 2004. Myosin gene
Genet 13:R57R64. mutation correlates with anatomical changes in the human lineage.
She X, Horvath JE, Jiang Z, Liu G, Furey TS, Christ L, Clark R, Graves T, Nature 428:415418.
Gulden CL, Alkan C, Bailey JA, Sahinalp C, Rocchi M, Haussler D, Stefansson H, Helgason A, Thorleifsson G, Steinthorsdottir V, Masson G,
Wilson RK, Miller W, Schwartz S, Eichler EE. 2004. The structure and Barnard J, Baker A, Jonasdottir A, Ingason A, Gudnadottir VG, Desnica
evolution of centromeric transition regions within the human genome. N, Hicks A, Gylfason A, Gudbjartsson DF, Jonsdottir GM, Sainz J,
Nature 430:857864. Agnarsson K, Birgisdottir B, Ghosh S, Olafsdottir A, Cazier JB,
She X, Liu G, Ventura M, Zhao S, Misceo D, Roberto R, Cardone MF, Kristjansson K, Frigge ML, Thorgeirsson TE, Gulcher JR, Kong A,
Rocchi M, Green ED, Archidiacano N, Eichler EE. 2006. A preliminary Stefansson K. 2005. A common inversion under selection in Europeans.
comparative analysis of primate segmental duplications shows elevated Nat Genet 37:129137.
substitution rates and a great-ape expansion of intrachromosomal Stern R, Woods CG. 2006. Evolutionary genetics: Is brain evolution still
duplications. Genome Res 16:576583. continuing in modern humans? Eur J Hum Genet 14:799800.
Shen JC, Rideout WM 3rd, Jones PA. 1994a. The rate of hydrolytic Stranger BE, Forrest MS, Clark AG, Minichiello MJ, Deutsch S, Lyle R,
deamination of 5-methylcytosine in double-stranded DNA. Nucleic Hunt S, Kahl B, Antonarakis SE, Tavare S, Deloukas P, Dermitzakis ET.
Acids Res 22:972976. 2005. Genome-wide associations of gene expression variation in humans.
Shen L, Wu LC, Sanlioglu S, Chen R, Mendoza AR, Dangel AW, Carroll PLoS Genet 1(6):e78.
MC, Zipf WB, Yu CY. 1994b. Structure and genetics of the partially Strichman-Almashanu LZ, Bustin M, Landsman D. 2003. Retroposed
duplicated gene RP located immediately upstream of the complement copies of the HMG genes: a window to genome dynamics. Genome Res
C4A and the C4B genes in the HLA class III region. Molecular cloning, 13:800812.

Human Mutation DOI 10.1002/humu


HUMAN MUTATION 28(2), 99^130, 2007 129

Svensson O, Arvestad L, Lagergren J. 2006. Genome-wide survey for Tuzun E, Sharp AJ, Bailey JA, Kaul R, Morrison VA, Pertz LM, Haugen E,
biologically functional pseudogenes. PLoS Comput Biol 2:e46. Hayden H, Albertson D, Pinkel D, Olson MV, Eichler EE. 2005. Fine-
Swanson WJ, Vacquier VD. 2002. The rapid evolution of reproductive scale structural variation of the human genome. Nat Genet 37:727732.
proteins. Nat Rev Genet 3:137144. Tyler-Smith C, Howe K, Santos FR. 2006. The rise and fall of the ape Y
Szamalek JM, Goidts V, Chuzhanova N, Hameister H, Cooper DN, Kehrer- chromosome? Nat Genet 38:141143.
Sawatzki H. 2005. Molecular characterisation of the pericentric Uddin M, Wildman DE, Liu G, Xu W, Johnson RM, Hof PR, Kapatos G,
inversion that distinguishes human chromosome 5 from the homologous Grossman LI, Goodman M. 2004. Sister grouping of chimpanzees and
chimpanzee chromosome. Hum Genet 117:168176. humans as revealed by genome-wide phylogenetic analysis of brain gene
Szamalek JM, Cooper DN, Goidts V, Hameister H, Kehrer-Sawatzki H. expression profiles. Proc Natl Acad Sci USA 101:29572962.
2006a. Characterization of the human-specific pericentric inversion that Urrutia AO, Hurst LD. 2001. Codon usage bias covaries with expression
discriminates human chromosome 1 from the homologous chromosomes breadth and the rate of synonymous evolution in humans, but this is not
in great apes. Hum Genet 120:126138. evidence for selection. Genetics 159:11911199.
Szamalek JM, Goidts V, Searle JB, Cooper DN, Hameister H, Kehrer- Vallender EJ, Lahn BT. 2004a. Positive selection on the human genome.
Sawatzki H. 2006b. The chimpanzee-specific pericentric inversions that Hum Mol Genet 13(Suppl 2):R245R254.
distinguish humans and chimpanzees have identical breakpoints in Pan Vallender EJ, Lahn BT. 2004b. Effects of chromosomal rearrangements on
troglodytes and Pan paniscus. Genomics 87:3945. human-chimpanzee molecular evolution. Genomics 84:757761.
Szamalek JM, Cooper DN, Schempp W, Minich P, Kohn M, Hoegel J, van de Lagemaat LN, Gagnier L, Medstrand P, Mager DL. 2005. Genomic
Goidts V, Hameister H, Kehrer-Sawatzki H. 2006c. Polymorphic micro- deletions and precise removal of transposable elements mediated by
inversions contribute to the genomic variability of humans and short identical DNA segments in primates. Genome Res 15:12431249.
chimpanzees. Hum Genet 119:103112. Vandepoele K, Van Roy N, Staes K, Speleman F, van Roy F. 2005. A novel
Takahashi A, Tsaur SC, Coyne JA, Wu CI. 2001. The nucleotide changes gene family NBPF: intricate structure generated by gene duplications
governing cuticular hydrocarbon variation and their evolution in during primate evolution. Mol Biol Evol 22:22652274.
Drosophila melanogaster. Proc Natl Acad Sci USA 98:39203925. Varki A, Altheide TK. 2005. Comparing the human and chimpanzee
Talbert PB, Bryson TD, Henikoff S. 2004. Adaptive evolution of genomes: searching for needles in a haystack. Genome Res 15:
centromere proteins in plants and animals. J Biol 3:18. 17461758.
Tang J, Shelton B, Makhatadze NJ, Zhang Y, Schaen M, Louie LG, Goedert Verrelli BC, Tishkoff SA, Stone AC, Touchman JW. 2006. Contrasting
JJ, Seaberg EC, Margolick JB, Mellors J, Kaslow RA. 2002. Distribution histories of G6PD molecular evolution and malarial resistace in humans
of chemokine receptor CCR2 and CCR5 genotypes and their relative and chimpanzees. Mol Biol Evol 23:15921601.
contribution to human immunodeficiency virus type 1 (HIV-1) Vincent BJ, Myers JS, Ho HJ, Kilroy GE, Walker JA, Watkins WS, Jorde
seroconversion, early HIV-1 RNA concentration in plasma, and later LB, Batzer MA. 2003. Following the LINEs: an analysis of primate
disease progression. J Virol 76:662672. genomic variation at human-specific LINE-1 insertion sites. Mol Biol
Taudien S, Galgoczy P, Huse K, Reichwald K, Schilhabel M, Szafranski K, Evol 20:13381348.
Shimizu A, Asakawa S, Frankish A, Loncarevic IF, Shimizu N, Siddiqui Visser R, Shimokawa O, Harada N, Kinoshita A, Ohta T, Niikawa N,
R, Platzer M. 2004. Polymorphic segmental duplications at 8p23.1 Matsumoto N. 2005. Identification of a 3.0-kb major recombination
challenge the determination of individual defensin gene repertoires hotspot in patients with Sotos syndrome who carry a common 1.9-Mb
and the assembly of a contiguous human reference sequence. BMC microdeletion. Am J Hum Genet 76:5267.
Genomics 5:92. Voight BF, Kudaravalli S, Wen X, Pritchard JK. 2006. A map of recent
Taudien S, Ebersberger I, Glockner G, Platzer M. 2006. Should the draft positive selection in the human genome. PLoS Biol 4:e72.
chimpanzee sequence be finished? Trends Genet 22:122125. Vowles EJ, Amos W. 2006. Quantifying ascertainment bias and species-
Taylor J, Tyekucheva S, Zody M, Chiaromonte F, Makova KD. 2006. Strong specific length differences in human and chimpanzee microsatellites
and weak male mutation bias at different sites in the primate genomes: using genome sequences. Mol Biol Evol 23:598607.
insights from the human-chimpanzee comparison. Mol Biol Evol 23: Wagner JJ, Terman GW, Chavkin C. 1993. Endogenous dynorphins inhibit
565573. excitatory neurotransmission and block LTP induction in the hippo-
Thanaraj TA, Clark F, Muilu J. 2003. Conservation of human alternative campus. Nature 363:451454.
splice events in mouse. Nucleic Acids Res 31:25442552. Wall JD, Frisse LA, Hudson RR, Di Rienzo A. 2003. Comparative linkage-
Thayer RE, Singer MF, McCutchan TF. 1981. Sequence relationships disequilibrium analysis of the beta-globin hotspot in primates. Am J Hum
between single repeat units of highly reiterated African Green monkey Genet 73:13301340.
DNA. Nucleic Acids Res 9:169181. Wang HY, Tang H, Shen CK, Wu CI. 2003. Rapidly evolving genes in
Thomas MA, Weston B, Joseph M, Wu W, Nekrutenko A, Tonellato PJ. human. I. The glycophorins and their possible role in evading malaria
2003. Evolutionary dynamics of oncogenes and tumor suppressor genes: parasites. Mol Biol Evol 20:17951804.
higher intensities of purifying selection than other genes. Mol Biol Evol Wang X, Thomas SD, Zhang J. 2004. Relaxation of selective constraint and
20:964968. loss of function in the evolution of human bitter taste receptor genes.
TomHon C, Zhu W, Millinoff D, Hayasaka K, Slightom JL, Goodman M, Hum Mol Genet 13:26712678.
Gumucio DL. 1997. Evolution of a fetal expression pattern via cis Wang H, Xing J, Grover D, Hedges Kyudong Han DJ, Walker JA, Batzer
changes near the gamma globin gene. J Biol Chem 272:1406214066. MA. 2005. SVA elements: a hominid-specific retroposon family. J Mol
Torrents D, Suyama M, Zdobnov E, Bork P. 2003. A genome-wide survey of Biol 354:9941007.
human pseudogenes. Genome Res 13:25592567. Wang X, Grus WE, Zhang J. 2006. Gene losses during human origins. PLoS
Trask BJ, Friedman C, Martin-Gallardo A, Rowen L, Akinbami C, Blanken- Biol 4:e52.
ship J, Collins C, Giorgi D, Iadonato S, Johnson F, Kuo WL, Massa H, Warburton PE, Willard HF. 1996. Evolution of centromeric a-satellite
Morrish T, Naylor S, Nguyen OT, Rouquier S, Smith T, Wong DJ, DNA: molecular organization within and between human and
Youngblom J, van den Engh G. 1998. Members of the olfactory receptor primate chromosomes. In: Jackson ST, Dover G, editors. Human
gene family are contained in large blocks of DNA duplicated polymorphi- genome evolution. Oxford: BIOS Scientific Publishers. pp 121145.
cally near the ends of human chromosomes. Hum Mol Genet 7:1326. Warburton PE, Haaf T, Gosden J, Lawson D, Willard HF. 1996.
Trinklein ND, Aldred SJ, Saldanha AJ, Myers RM. 2003. Identification and Characterization of a chromosome-specific chimpanzee alpha satellite
functional analysis of human transcriptional promoters. Genome Res subset: evolutionary relationship to subsets on human chromosomes.
13:308312. Genomics 33:220228.
Turner G, Barbulescu M, Su M, Jensen-Seaman MI, Kidd KK, Lenz J. 2001. Watanabe H, Fujiyama A, Hattori M, Taylor TD, Toyoda A, Kuroki Y,
Insertional polymorphisms of full-length endogenous retroviruses in Noguchi H, BenKahla A, Lehrach H, Sudbrak R, Kube M, Taenzer S,
humans. Curr Biol 11:15311535. Galgoczy P, Platzer M, Scharfe M, Nordsiek G, Blocker H, Hellmann I,

Human Mutation DOI 10.1002/humu


130 HUMAN MUTATION 28(2), 99^130, 2007

Khaitovich P, Paabo S, Reinhardt R, Zheng HJ, Zhang XL, Zhu GF, Wang Xue Y, Daly A, Yngvadottir B, Liu M, Coop G, Kim Y, Sabeti P, Chen Y,
BF, Fu G, Ren SX, Zhao GP, Chen Z, Lee YS, Cheong JE, Choi SH, Wu Stalker J, Huckle E, Burton J, Leonard S, Rogers J, Tyler-Smith C. 2006.
KM, Liu TT, Hsiao KJ, Tsai SF, Kim CG, OOta S, Kitano T, Kohara Y, Spread of an inactive form of caspase-12 in humans is due to recent
Saitou N, Park HS, Wang SY, Yaspo ML, Sakaki Y. 2004. DNA sequence positive selection. Am J Hum Genet 78:659670.
and comparative analysis of chimpanzee chromosome 22. Nature 429: Yang Z. 1998. Likelihood ratio tests for detecting positive selection and
382388. application to primate lysozyme evolution. Mol Biol Evol 15:568573.
Watkins DI. 1995. The evolution of major histocompatibility class I genes Yang Z, Bielawski JP. 2000. Statistical methods for detecting molecular
in primates. Crit Rev Immunol 15:129. adaptation. Trends Ecol Evol 15:496503.
Webster MT, Smith NG, Ellegren H. 2002. Microsatellite evolution inferred Yoder AD, Yang Z. 2000. Estimation of primate speciation dates using local
from human-chimpanzee genomic sequence alignments. Proc Natl Acad molecular clocks. Mol Biol Evol 17:10811090.
Sci USA 99:87488753. Yohn CT, Jiang Z, McGrath SD, Hayden KE, Khaitovich P, Johnson ME,
Wevrick R, Willard HF. 1989. Long-range organization of tandem arrays of Eichler MY, McPherson JD, Zhao S, Paabo S, Eichler EE. 2005. Lineage-
alpha satellite DNA at the centromeres of human chromosomes: high- specific expansions of retroviral insertions within the genomes of African
frequency array-length polymorphism and meiotic stability. Proc Natl great apes but not humans and orangutans. PLoS Biol 3:e110.
Acad Sci USA 86:93949398. Young JM, Trask BJ. 2002. The sense of smell: genomics of vertebrate
Wildman DE, Uddin M, Liu G, Grossman LI, Goodman M. 2003. odorant receptors. Hum Mol Genet 11:11531160.
Implications of natural selection in shaping 99.4% nonsynonymous DNA Young JM, Kambere M, Trask BJ, Lane RP. 2005. Divergent V1R repertoires
identity between humans and chimpanzees: enlarging genus Homo. Proc in five species: amplification in rodents, decimation in primates, and a
Natl Acad Sci USA 100:71817188. surprisingly small repertoire in dogs. Genome Res 15:231240.
Wilkie AO, Higgs DR, Rack KA, Buckle VJ, Spurr NK, Fischel-Ghodsian Yu N, Jensen-Seaman MI, Chemnick L, Kidd JR, Deinard AS, Ryder O,
N, Ceccherini I, Brown WR, Harris PC. 1991. Stable length Kidd KK, Li WH. 2003. Low nucleotide diversity in chimpanzees and
polymorphism of up to 260 kb at the tip of the short arm of human bonobos. Genetics 164:15111518.
chromosome 16. Cell 64:595606. Yu X, Zheng H, Wang J, Wang W, Su B. 2006a. Detecting lineage-specific
Willard HF. 1990. Centromeres of mammalian chromosomes. Trends Genet adaptive evolution of brain-expressed genes in human using rhesus
6:410416. macaque as outgroup. Genomics July 17; [Epub ahead of print].
Wilson GM, Flibotte S, Missirlis PI, Marra MA, Jones S, Thornton K, Clark Yu H, Jiang H, Zhou Q, Yang J, Cun Y, Su B, Xiao C, Wang W. 2006b.
AG, Holt RA. 2006a. Identification by full-coverage array CGH of Origination and evolution of a human-specific transmembrane protein
human DNA copy number increases relative to chimpanzee and gorilla. gene, c1orf37-dup. Hum Mol Genet 15:18701875.
Genome Res 16:173181. Yunis JJ, Prakash O. 1982. The origin of man: a chromosomal pictorial
Wilson GM, Flibotte S, Chopra V, Melnyk BL, Honer WG, Holt RA. legacy. Science 215:15251530.
2006b. DNA copy-number analysis in bipolar disorder and schizophrenia Zhang XM, Cathala G, Soua Z, Lefranc MP, Huck S. 1996. The human T-
reveals aberrations in genes involved in glutamate signaling. Hum Mol cell receptor gamma variable pseudogene V10 is a distinctive marker of
Genet 15:743749. human speciation. Immunogenetics 43:196203.
Winckler W, Myers SR, Richter DJ, Onofrio RC, McDonald GJ, Bontrop Zhang J, Zhang YP, Rosenberg HF. 2002a. Adaptive evolution of a
RE, McVean GA, Gabriel SB, Reich D, Donnelly P, Altshuler D. 2005. duplicated pancreatic ribonuclease gene in a leaf-eating monkey. Nat
Comparison of fine-scale recombination rates in humans and chimpan- Genet 30:411415.
zees. Science 308:107111. Zhang J, Webb DM, Podlaha O. 2002b. Accelerated protein evolution and
Winter H, Langbein L, Krawczak M, Cooper DN, Jave-Suarez LF, Rogers origins of human-specific features: FOXP2 as an example. Genetics
MA, Praetzel S, Heidt PJ, Schweizer J. 2001. Human type I hair keratin 162:18251835.
pseudogene phihHaA has functional orthologs in the chimpanzee and Zhang J, Rosenberg HF. 2002. Diversifying selection of the tumor-
gorilla: evidence for recent inactivation of the human gene after the growth promoter angiogenin in primate evolution. Mol Biol Evol 19:
Pan-Homo divergence. Hum Genet 108:3742. 438445.
Wise CA, Sraml M, Rubinsztein DC, Easteal S. 1997. Comparative nuclear Zhang Z, Harrison PM, Liu Y, Gerstein M. 2003. Millions of years of
and mitochondrial genome diversity in humans and chimpanzees. Mol evolution preserved: a comprehensive catalog of the processed
Biol Evol14:707716. pseudogenes in the human genome. Genome Res 13:25412558.
Won YJ, Hey J. 2005. Divergence population genetics of chimpanzees. Mol Zhang J, Wang X, Podlaha O. 2004a. Testing the chromosomal speciation
Biol Evol 22:297307. hypothesis for humans and chimpanzees. Genome Res 14:845851.
Wooding S, Jorde LB. 2006. Duplication and divergence in humans and Zhang Z, Carriero N, Gerstein M. 2004b. Comparative analysis of processed
chimpanzees. Bioessays 28:335338. pseudogenes in the mouse and human genomes. Trends Genet 20:
Wooding S, Bufe B, Grassi C, Howard MT, Stone AC, Vazquez M, Dunn 6267.
DM, Meyerhof W, Weiss RB, Bamshad MJ. 2006. Independent evolution Zhang J, Nielsen R, Yang Z. 2005a. Evaluation of an improved branch-site
of bitter-taste sensitivity in humans and chimpanzees. Nature 440: likelihood method for detecting positive selection at the molecular level.
930934. Mol Biol Evol 22:24722479.
Wray GA, Hahn MW, Abouheif E, Balhoff JP, Pizer M, Rockman MV, Zhang L, Lu HH, Chung WY, Yang J, Li WH. 2005b. Patterns of segmental
Romano LA. 2003. The evolution of transcriptional regulation in duplication in the human genome. Mol Biol Evol 22:135141.
eukaryotes. Mol Biol Evol 20:13771419. Zheng D, Zhang Z, Harrison PM, Karro J, Carriero N, Gerstein M. 2005.
Wyckoff GJ, Wang W, Wu CI. 2000. Rapid evolution of male reproductive Integrated pseudogene annotation for human chromosome 22: evidence
genes in the descent of man. Nature 403:304309. for transcription. J Mol Biol 349:2745.
Xiao Y, Hughes AL, Ando J, Matsuda Y, Cheng JF, Skinner-Noble D, Zhang Zimonjic DB, Kelley MJ, Rubin JS, Aaronson SA, Popescu NC. 1997.
G. 2004. A genome-wide screen identifies a single beta-defensin gene Fluorescence in situ hybridization analysis of keratinocyte growth factor
cluster in the chicken: implications for the origin and evolution of gene amplification and dispersion in evolution of great apes and humans.
mammalian defensins. BMC Genomics 5:56. Proc Natl Acad Sci USA 94:1146111465.
Xing Y, Wang Q, Lee C. 2006. Evolutionary divergence of exon Zimprich A, Kraus J, Woltje M, Mayer P, Rauch E, Hollt V. 2000. An allelic
flanks: a dissection of mutability and selection. Genetics 173: variation in the human prodynorphin gene promoter alters stimulus-
17871791. induced expression. J Neurochem 74:472477.

Human Mutation DOI 10.1002/humu

You might also like