Sunteți pe pagina 1din 8

Evolution of primary microcephaly genes and the enlargement

of primate brains
Chris Ponting1 and Andrew P Jackson2

Brain size, in relation to body size, has varied markedly during been accommodated topologically by folding of the cere-
the evolution of mammals. In particular, a large cerebral cortex bral cortex to form convolutions, so that there is a transition
is a feature that distinguishes humans from our fellow primates. from the smooth cortical surface seen in rodents to one of
Such anatomical changes must have a basis in genetic multiple folds (gyri and sulci; see Glossary) in humans.
alterations, but the molecular processes involved have yet to be
defined. However, recent advances from the cloning of two This review contrasts the pathological reduction in
human disease genes promise to make inroads in this human brain size (primary microcephaly) with the
important area. Microcephalin (MCPH1) and Abnormal spindle- increase in relative brain size during primate evolution.
like microcephaly associated (ASPM) are genes mutated in We describe the primary microcephaly genes and their
primary microcephaly, a human neurodevelopmental disorder. functions, and address the recently proposed roles of
In this atavistic condition, brain size is reduced in volume to a ASPM and microcephalin genes in determining brain
size comparable with that of early hominids. Hence, it has been size among different mammals.
proposed that these genes evolved adaptively with increasing
primate brain size. Subsequent studies have lent weight to this Cellular determinants of brain size
hypothesis by showing that both genes have undergone The relative rates of proliferation and cell death occurring
positive selection during great ape evolution. Further functional during neurogenesis (see Glossary) are likely to be major
characterisation of their proteins will contribute to an determinants of mammalian brain size. Evolutionary
understanding of the molecular and evolutionary processes expansion of the mammalian brain must, at least in part,
that have determined human brain size. be the result of increased numbers of cell divisions during
Addresses neurogenesis [5]. An evolutionary increase in the number
1
MRC Functional Genetics, University of Oxford, Department of Human of columnar functional units generated by proliferative
Anatomy and Genetics, South Parks Road, Oxford OX1 3QX, UK
2
units of progenitor cells has been proposed to explain
MRC Human Genetics Unit, Western General Hospital, Crewe Road, how increased cell number translates into expansion in
Edinburgh, EH4 2XU, UK
cortical surface area [6].
Corresponding authors: Ponting, Chris (chris.ponting@anat.ox.ac.uk);
Jackson, Andrew P (andrew.jackson@hgu.mrc.ac.uk) Neurogenesis first generates a pool of progenitor cells by
symmetric cell division, and neurons are subsequently
formed by asymmetric cell divisions. It has been proposed
Current Opinion in Genetics & Development 2005, 15:241248
that small changes in the relative proportion of such
This review comes from a themed issue on divisions could be responsible for marked alterations in
Genetics of disease cortical surface area [7]. Indeed, it has been demonstrated
Edited by Veronica van Heyningen and David FitzPatrick recently that increasing the proportion of cycling progeni-
Available online 22nd April 2005
tor cells is sufficient to increase cortical surface area,
producing a mouse cerebral cortex with sulci and gyri
0959-437X/$ see front matter [8]. Widespread neuronal cell death also occurs during
# 2005 Elsevier Ltd. All rights reserved.
foetal brain development, and the amount of cell death is
DOI 10.1016/j.gde.2005.04.009 also an important determinant of brain size. Inactivating
components of the apoptotic pathway also leads to an
enlarged sulcated (i.e. furrowed) mouse brain [9,10].

Introduction Candidate genes


There has been a dramatic expansion in brain size during Despite insights into the cellular mechanisms that regulate
mammalian evolution, with a thousand fold increase in brain size, the molecules and genes responsible for this
cortical surface area between mouse and man [1]. Morpho- process remain enigmatic. In general, genes expressed
metric studies (see Glossary) have revealed a scaling of the predominantly in the brain have evolved, between human
brain relative to body size during mammalian evolution and mouse, more slowly than those expressed in other
[2], with disproportionate enlargement of the cerebral tissues [11]. Nevertheless, some nervous system genes
cortex [3]. In primates, this might have been driven by exhibit more rapid evolution among primates than among
increased social, rather than environmental, complexity rodents [12], and so changes in these genes might have
[4]. Expansion has been through increased surface area been responsible for brain size modifications. Clearly,
rather than increased cortical thickness [1,2]. This has molecules involved in regulating cellular proliferation or

www.sciencedirect.com Current Opinion in Genetics & Development 2005, 15:241248


242 Genetics of disease

Glossary and a further pair at the C-terminus (Figure 2a). BRCT


Aneuploidy Condition of having an abnormal number of domains occur in many DNA repair and cell cycle pro-
chromosomes. teins, and they participate in proteinprotein and pro-
Atavistic Regarding a reversion to an ancestral form.
teinDNA interactions [33]. The functions of these
Early truncating mutations DNA mutations that give rise to a stop
codon in the 50 -most region of a protein coding sequence. domains have recently been further clarified by the
Gyri Elevations in the surface of the cerebral hemispheres. demonstration that they specifically bind proteins phos-
IQ domain A protein motif often containing conserved isoleucine phorylated on serine and/or threonine residues [34]. The
and glutamine amino acids. implication is, thus, that their binding is modulated by
Morphometric Regarding the measurement of shape and size.
Neurogenesis The developmental process of producing neural
phosphorylation as a result of either cell cycle stage-
cells. specific events or signals arising from DNA damage. This
Sulci Furrows in the surface of the cerebral hemisphere. year, a role in DNA damage response was confirmed for
microcephalin, after the demonstration that microcepha-
lin localises to DNA repair foci and that cells depleted for
apoptosis are likely candidates, given the processes known microcephalin by RNA interference exhibit failure in cell
to influence brain size outlined above. cycle checkpoints [35]. This gene has also been isolated
(as BRIT1) in a screen for negative regulators of telomer-
It has been suggested that genes responsible for human ase [36], and has a demonstrated role in the cell cycle in
neurogenetic disorders might be of particular interest in regulating chromosome condensation [17].
this regard because, through disease, they have demon-
strated the potential to alter developmental outcome of Both mutations identified in microcephalin have been early
brain morphology and function [13]. In respect to brain truncating mutations (see Glossary) and, therefore, are
size, genes for primary microcephaly are prime candidates. predicted to lead to a complete loss of gene function [28].
Given the diverse roles outlined above, it remains unclear
Primary microcephaly an evolutionary how abrogation of microcephalin leads to reduction in
condition? brain size. Loss of DNA repair molecules can lead to
Primary microcephaly is a Mendelian neurodevelopmen- increased cell death during neurogenesis [37]. Conver-
tal disorder in which there is marked reduction in human sely, embryonic proliferation might be reduced, as in the
brain size. Brain volume is reduced to a third of normal case of Brca1/ mice [38].
[14], to a size comparable with that of early hominids
(Figure 1) [15]. Surprisingly, in this disorder, the great loss ASPM
in volume of cerebral cortex does not lead to significant As with microcephalin, ASPM was positionally cloned
neurological dysfunction; apart from a moderate reduc- using consanguineous Pakistani families, and locus refine-
tion in cognitive ability [16]. The cerebral cortex has a ment by identifying shared ancestral haplotypes [18]. All
simplified gyral pattern [17,18], with no major abnorm- 21 mutations reported to date have been protein-truncat-
ality of cortical architecture [14,19]. These findings, along ing, and are distributed throughout the gene [30,31],
with the facial characteristics of a hypoplastic skull vault presumably implying that primary microcephaly is the
and sloping forehead, have led to the proposal that consequence of functionally null mutations.
primary microcephaly might be an evolutionary (i.e.
atavistic) condition (see Glossary) [19,20]. ASPM is a very large protein of 3477 amino acids, and
comprises a putative microtubule-binding domain at its N-
Microcephaly genes terminal, a calponin-homology domain, multiple IQ cal-
Linkage mapping studies in consanguineous families modulin-binding domains (see Glossary) and a terminal
have identified six gene loci for primary microcephaly region (Figure 2b). With the exception of some rodents, the
(MCPH16) [2127]. Subsequently, two of these genes number of ASPM IQ domains is constant among diverse
were positionally cloned: first microcephalin [28] and then mammals [39]. This contradicts an early proposal [18] that
abnormal spindle-like microcephaly-associated (ASPM) [18], IQ domain number positively correlates with increased
for the MCPH1 and MCPH5 loci, respectively. Mutations nervous system complexity. Like microcephalin, ASPM is
in ASPM appear to be a major cause of primary micro- expressed in the cerebral cortical ventricular zone and
cephaly [2931], whereas microcephalin mutations have proliferative zones of the medial and lateral ganglionic
only been reported in three families [17,28]. eminences during neurogenesis [18,28]. ASPM is also
expressed postnatally in regions of persistent neurogenesis.
Microcephalin
Microcephalin (MCPH1) is an 835 amino acid protein Although the cellular functions of ASPM have yet to be
containing BRCA1 C-terminal (BRCT) domains [28]. Its defined, its fly orthologue, abnormal-spindle protein
closest human homologues are topoisomerase II-binding (Asp), has been extensively studied and has roles in both
protein (TopBP1) and BRCA1. Three BRCT domains mitosis and meiosis [4043]. Asp is necessary for the
are predicted to be present [32], one at the N-terminus centrosome to function as a microtubule-organising

Current Opinion in Genetics & Development 2005, 15:241248 www.sciencedirect.com


Evolution of primary microcephaly genes and the enlargement of primate brains Ponting and Jackson 243

Figure 1

Evolutionary expansion of the primate brain contrasts with the pathological reduction in brain size seen in primary microcephaly. (a) Primate skulls
from the Museum of Comparative Zoology, Harvard University (Reproduced with permission from [54]).There has been a large increase in brain
size during primate evolution, reflected by enlargement of the skull vault and cranial capacity. (b) Magnetic resonance imaging of a normal adult
human head (left) and an individual with MCPH1 primary microcephaly (right). Midline sagittal (T1 weighted) sections; adapted with permission
from [28]). In primary microcephaly, there has been a marked decrease in brain volume. In particular, the cerebral cortex is much smaller and
exhibits reduced cortical folding with a simplified gyral pattern.

centre and to focus the poles of the mitotic spindle [40]. It orthologues are likely to be present in all chordates,
is also required for cytokinesis, where it is an organiser of because these genes are identifiable in all primates,
the central spindle [41]. Mutation of asp leads to mitotic carnivores, ruminants, marsupials, birds, fish, amphibians
arrest in larval neuroblasts [42]. and sea squirts for which we currently possess genomic
sequence. These MCPH1 sequences are each identical in
Positioning of the mitotic spindle perpendicular to the architecture to human MCPH1. Their N-terminal BRCT
epithelial surface is a regulated process in fly neuroblasts domain is separated from two C-terminal BRCT domains
and is required for correct orientation of cell cleavage by a long stretch of inter-BRCT domain sequence (IBS)
during asymmetric cell division [44]. Additionally, clea- that appears devoid of known domain structures and that
vage plane orientation might be important in localising is mostly encoded by a single exon.
asymmetric determinants in mammals [45]. Given the
known functions of Asp, it was, therefore, proposed that Similar MCPH1 genes are also present in other metazoan
mutation of ASPM might reduce brain size by influencing species, in particular the two main invertebrate model
mitotic spindle orientation, and reduce neural cell num- organisms, Drosophila melanogaster (gene CG8981) and
ber through an effect on the ratio of asymmetric to Caenorhabditis elegans (W04A8.1).
symmetric cell divisions [18].
The MCPH1 gene, however, might have arisen in eukar-
MCPH1 is ubiquitous in metazoans yotic evolution even earlier than the earliest metazoan.
A previous report [13] suggested that MCPH1 is found Other transcripts are known, from more distantly-related
only in mammals. By contrast, we believe that MCPH1 eukaryotic species, that all encode sequences similar to

www.sciencedirect.com Current Opinion in Genetics & Development 2005, 15:241248


244 Genetics of disease

Figure 2

(a) Microcephalin

Exons
1 2 3 4 5 6 7 8 9 10 11 12 13 14

Domains

BRCT IBS BRCT BRCT

(b) ASPM

Exons

1 2 3 13 17 18 28

Domains

Microtubule binding Calponin IQ (multiple) C terminus


homology

Current Opinion in Genetics & Development

Exon structure of microcephalin and ASPM genes (a) Microcephalin is an 835 amino acid (aa) protein, mutated in MCPH1 primary microcephaly.
The protein is predicted to contain three BRCA1 C-terminal (BRCT) domains. An N-terminal BRCT domain is separated by over 500 aa of
inter-BRCT domain sequence (IBS), which has no predicted domain structure, from a pair of BRCT domains at the C-terminus. Regions of the
gene have undergone high levels of adaptive evolution (red exons, KA/KS > 3, among ancestors of old world monkeys and great apes), whereas
purifying selection has occurred particularly in the gene regions encoding BRCT domains. Exon structure and KA/KS ratios are taken from [13],
and predicted domain structure from [28].(b) Abnormal spindle-like microcephaly-associated (ASPM) protein is a very large molecule of 3477
aa divided into four regions: a N-terminal putative microtubule binding domain (purple), calponin-homology domain (orange), multiple IQ,
calmodulin-binding, domains (red) and a C-terminal region (blue) [18]. Exons 3 and 18 have undergone adaptive evolution, with regions of
significantly elevated KA/KS in the ape lineages leading to humans (red exons) [39].

the BRCT domain-containing regions of human MCPH1. reduced functional constraint on sequence or by posi-
These include a fungus (Cryptococcus neoformans; gene tive selection, caused by the accumulation of nonsynon-
CNBK1030), an amoeba (Entamoeba histolytica), and a ymous changes that confer adaptive benefit.
plant (Sorghum bicolor; EST [expressed sequence tag]
CD430938). However, full-length MCPH1-like sequences Recently, two studies have attempted to distinguish posi-
containing N- and C-terminal BRCT domains, separated tive selection from neutrality during the evolution of
by an IBS region, have yet to be identified outside of the MCPH1 genes [13,46]. They demonstrated that
metazoans. whereas the flanking BRCT domains have accumulated
amino acid changes relatively slowly, as befits sequence
Adaptive evolution of microcephalin that is well-conserved back to at least the earliest metazoan
When human and mouse sequences are aligned, MCPH1 organism, the intervening IBS has acquired substantial
appears among the top 10% of rapidly evolving genes: numbers of amino acid alterations. This is also consistent
they have accumulated more amino acid-changing (non- with the considerably reduced sequence similarity
synonymous) nucleotide substitutions for each silent between, for example, sea squirt (Ciona intestinalis) and
(synonymous) substitution than most other genes. Such mammalian MCPH1 genes in their extended IBS regions,
rapid evolution can occur neutrally that is, as a result of or even between different Drosophila species (see below).

Current Opinion in Genetics & Development 2005, 15:241248 www.sciencedirect.com


Evolution of primary microcephaly genes and the enlargement of primate brains Ponting and Jackson 245

Using sequence differences in MCPH1 genes, both within Nevertheless, the extended IBS of cat and fly MCPH1
the human population and among different mammals, genes have each independently acquired different
these investigators demonstrated that although neutral repeats (Figure 3). In most Drosophila species, there
evolution could explain some of their changes Darwinian are three such repeats, but Drosophila pseudoobscura has
positive selection most likely also contributed changes. acquired an extra pair, making five in total. These repeats
These occurred specifically along the simian lineages that have been acquired relatively recently, certainly in the 55
led to modern apes and Old World monkeys. Similar million years since the last common ancestor of cat with
conclusions were drawn from human polymorphism data dog [47] and the 25 million years since the last common
[13,46]. ancestor of D. pseudoobscura and the other Drosophila
species [48]. Therefore, it is possible that these repeats
By contrast, the evolution of MCPH1 genes in species were acquired adaptively by flies and cats. The fruit fly
other than primates appears to have been more pedes- Mcph1 protein repeats are distinguished from those of the
trian, because there is no statistical evidence for Darwi- mosquito protein by their different conservation of serine
nian positive selection for any of these other animals. and threonine residues, respectively (Figure 3).
Figure 3

(a)
RRR*
D. melanogaster 1: PVRTTRRRSCVAEI 436-449
D. melanogaster 2: DTRMTRRRSSLLTS 456-469
D. melanogaster 3: EPRMTRRRSSLLGI 484-497
D. pseudoobscura 1: PVRTSRRRSSAHIS 437-450
D. pseudoobscura 2: EQRVTRRRSSAHIP 464-477
D. pseudoobscura 3: EPRMTRRRSSAHIL 515-528
D. pseudoobscura 4: EPRLKRRRSSAHIP 542-555
D. pseudoobscura 5: DLRMTRRRSSLLQA 569-582
D. yakuba 1: PVRTTRRRSCVAEI 439-452
D. yakuba 2: DTRMTRRRSSLLTS 459-472
D. yakuba 3: EPRMTRRRSSLLAT 487-500
D. ananassae 1: PVRTNRRRSCAAEI 437-450
D. ananassae 2: EPRMTRRRSSLHIN 454-467
D. ananassae 3: EPRMTRRRSSLHLA 481-494
D. mojavensis 1: PLRQLRRRSLAYVC 302-315
D. mojavensis 2: GSRLTRRRSQLNIT 328-341
D. mojavensis 3: EPRITRRRSLQLLT 364-378
D. virilis 1: PLRASRRRSSACGA 311-324
D. virilis 2: EPRMTRRRSSLLSM 337-350
D. virilis 3: EPRMPRRRSSLQLA 372-385
A. gambiae 1: GTPRGRRKTVMFTP 459-472
A. gambiae 2: PKQTFRRKTIVLPN 495-508
A. gambiae 3: SNTSNRRKTIVAAP 520-533
A. gambiae 4: VRVSSRRKTIVAAK 601-614
A. gambiae 5: KNKRSRRKTIAFGN 632-645
A. gambiae 6: NAPKDRRRTLFIPT 726-739
A. gambiae 7: LSLINRRRTLYTPS 762-775
A. gambiae 8: RAPDRRRRTVFDIT 858-871

(b)

Felis catus 1: LKTGSVHSSAPAGPSAS 274-290


Felis catus 2: LKTGSVHSSAPAGPSAS 291-307
Felis catus 3: LKTGSVHSLAPAGHLSQ 308-324
Canis familiaris LKTSCVPSLAPPGYLCQ 275-291

Current Opinion in Genetics & Development

Amino acid conservation of MCPH1 proteins (a) Repetitive arginine- (R) and serine- (S) or threonine- (T) rich motifs in arthropod MCPH1 proteins.
Note that Drosophila pseudoobscura MCPH1 possesses five repeats, whereas the other drosophilids possess three, and the mosquito Anopheles
gambiae MCPH1 possesses eight. In addition, Drosophila repeats conserve a serine in the position that the mosquito repeats always contain a
threonine. Alignment columns shaded in light grey contain at least 80% hydrophobic amino acids. (b) Triplicated repeat in cat (Felis catus)
MCPH1 that is present as a single copy in all other mammals known including dog (Canis familiaris).

www.sciencedirect.com Current Opinion in Genetics & Development 2005, 15:241248


246 Genetics of disease

Given this pattern of conservation, and the fact that that these modern pathological changes, which are loss-
BRCT domains are phosphoserine- or phosphothreo- of-function mutations, do not recapitulate the ancient
nine-binding domains [34,49], we speculate that fly, hominid genotype. Instead microcephalin and ASPM genes
and possibly human, microcephalin function could be have been preserved as functional sequences, without
regulated by phosphorylated serines or threonines in degenerating into pseudogenes since at least an early
the IBS repeats. Such residues might well interact with metazoan ancestor to extant animals.
the BRCT domains of other proteins, or even interact
with the flanking N- or C-terminal BRCT domains of In general, microcephalin or ASPM gene function does not
microcephalin itself. promote enlargement of the neocortex, because this
structure is specific to mammals, and the origin of each
Adaptive evolution of ASPM genes of these genes predates the first mammal. Instead, incre-
The ASPM gene also appears to have accumulated sig- mental adaptive changes to these genes might have subtly
nificant numbers of adaptive changes in the terminal altered cell cycle proliferation or reduced the propensity
human evolutionary branch since the last common ances- of neurons to apoptosis. For ASPM, alterations in mitotic
tor of chimpanzees and humans [39,50]. By comparing spindle function, perhaps as a consequence of altered
ASPM gene sequences, gorillas, chimpanzees and humans spindle orientation, might have increased symmetric cell
were distinguished from other primates by having accu- division at the expense of asymmetric cell division in
mulated large numbers of nonsynonymous nucleotide neurogenesis, thereby increasing the eventual neuronal
substitutions in this gene [39,50]. Moreover, during number. Alternatively, it is suggested that sequence
the human lineage since the last common ancestor with changes might have resulted in increased neuronal survi-
chimpanzees, approximately 15 of these substitutions in val, through more robust chromosome segregation [51]
this gene have been adaptive, and these are proposed to resulting from reduction in the high levels of aneuploidy
have contributed to the progressive enlargement of the (see Glossary) seen in rodent neuroblasts [53].
hominid cerebral cortex [39]. Significantly more amino
acid changes have occurred between chimpanzee and Conclusions
human sequences than they have recently within the Despite answers to the question of whether primate
human population, which again argues for positive selec- microcephalin or ASPM genes evolved adaptively, it still
tion in the terminal human branch [39,50]. remains to be established why they did so. The expres-
sion of these genes is not restricted to the central nervous
Among apes, the largest concentrations of nonsynon- system and, because their known functions are not con-
ymous substitutions occur within two long exons (3 fined to processes that regulate brain morphology, it is still
and 18) that encode large portions of likely microtu- possible that their evolution is causally unconnected with
bule-binding and calmodulin-binding regions, respec- the expansion of the cerebral cortex in great apes. Clearly,
tively [39,51] (Figure 2). The ASPM calponin- an improved understanding of the function of these
homology domain has experienced substantially fewer proteins would also assist in resolving this issue, as would
such substitutions. Hence, for both ASPM and MCPH1 analyses of the evolutionary histories of other primary
gene sequences, it is the extended exons that have microcephaly genes, once they have been identified. We
accumulated disproportionately large numbers of nonsy- wait with interest for developmental and cellular studies
nonymous substitutions, whereas regions encoding large that will address the biological significance of the bio-
folded protein domains have attracted relatively few. It is chemical changes identified from these evolutionary stu-
also noteworthy that adaptive sites are also clustered in a dies.
large exon (exon 11) of BRCA1 [52] that provides much of
the sequence intervening between its terminal structural Acknowledgements
domains. We believe it improbable that large exons are This work was funded by the UK Medical Research Council. We thank
Ian Hope for bringing the C.elegans MCPH1 orthologue to our attention.
more likely, in general, to accumulate adaptive mutations.
Rather, such exons (exon 8 of microcephalin and exon 18 of
References and recommended reading
ASPM) might have been extended by successive internal Papers of particular interest, published within the annual period of
duplications over a time period that was sufficiently short review, have been highlighted as:
to have not suffered from intron insertion.  of special interest
 of outstanding interest
How might microcephaly genes have been
involved in evolution of human brain size? 1. Rakic P: A small step for the cell, a giant leap for mankind:
a hypothesis of neocortical expansion during evolution.
Pathological changes in microcephalin and ASPM reduce Trends Neurosci 1995, 18:383-388.
brain size, therefore demonstrating that these genes have 2. Northcutt RG, Kaas JH: The emergence and evolution of the
the capability of altering the developmental outcome of mammalian neocortex. Trends Neurosci 1995, 18:373-379.
neurogenesis. These mutations apparently recapitulate 3. Clark DA, Mitra PP, Wang SS: Scalable architecture in
an earlier hominid phenotypic state. It is clear, however, mammalian brains. Nature 2001, 411:189-193.

Current Opinion in Genetics & Development 2005, 15:241248 www.sciencedirect.com


Evolution of primary microcephaly genes and the enlargement of primate brains Ponting and Jackson 247

4. Dunbar RIM: Coevolution of neocortical size, group size and 23. Jamieson CR, Fryns JP, Jacobs J, Matthijs G, Abramowicz MJ:
language in humans. Behav Brain Sci 1993, 16:681-735. Primary autosomal recessive microcephaly: MCPH5 maps to
1q25-q32. Am J Hum Genet 2000, 67:1575-1577.
5. Kornack DR, Rakic P: Changes in cell-cycle kinetics during the
development and evolution of primate neocortex. Proc Natl 24. Leal GF, Roberts E, Silva EO, Costa SM, Hampshire DJ,
Acad Sci USA 1998, 95:1242-1246. Woods CG: A novel locus for autosomal recessive primary
microcephaly (MCPH6) maps to 13q12.2. J Med Genet 2003,
6. Rakic P: Specification of cerebral cortical areas. Science 1988, 40:540-542.
241:170-176.
25. Moynihan L, Jackson AP, Roberts E, Karbani G, Lewis I, Corry P,
7. Caviness VS Jr, Takahashi T, Nowakowski RS: Numbers, time Turner G, Mueller RF, Lench NJ, Woods CG: A third novel locus
and neocortical neuronogenesis: a general developmental for primary autosomal recessive microcephaly maps to
and evolutionary model. Trends Neurosci 1995, 18:379-383. chromosome 9q34. Am J Hum Genet 2000, 66:724-727.
8. Chenn A, Walsh CA: Regulation of cerebral cortical size by 26. Pattison L, Crow YJ, Deeble VJ, Jackson AP, Jafri H, Rashid Y,
control of cell cycle exit in neural precursors. Science 2002, Roberts E, Woods CG: A fifth locus for primary autosomal
297:365-369. recessive microcephaly maps to chromosome 1q31. Am J Hum
Genet 2000, 67:1578-1580.
9. Kuida K, Zheng TS, Na S, Kuan C, Yang D, Karasuyama H,
Rakic P, Flavell RA: Decreased apoptosis in the brain and 27. Roberts E, Jackson AP, Carradice AC, Deeble VJ, Mannan J,
premature lethality in CPP32- deficient mice. Nature 1996, Rashid Y, Jafri H, McHale DP, Markham AF, Lench NJ et al.: The
384:368-372. second locus for autosomal recessive primary microcephaly
(MCPH2) maps to chromosome 19q13.1-13.2. Eur J Hum Genet
10. Kuida K, Haydar TF, Kuan CY, Gu Y, Taya C, Karasuyama H,
1999, 7:815-820.
Su MS, Rakic P, Flavell RA: Reduced apoptosis and cytochrome
c-mediated caspase activation in mice lacking caspase 9. 28. Jackson AP, Eastwood H, Bell SM, Adu J, Toomes C, Carr IM,
Cell 1998, 94:325-337. Roberts E, Hampshire DJ, Crow YJ, Mighell AJ et al.:
Identification of microcephalin, a protein implicated in
11. Winter EE, Goodstadt L, Ponting CP: Elevated rates of protein
determining the size of the human brain. Am J Hum Genet 2002,
secretion, evolution, and disease among tissue-specific
71:136-142.
genes. Genome Res 2004, 14:54-61.
29. Roberts E, Hampshire DJ, Pattison L, Springell K, Jafri H, Corry P,
12. Dorus S, Vallender EJ, Evans PD, Anderson JR, Gilbert SL,
Mannon J, Rashid Y, Crow Y, Bond J et al.: Autosomal recessive
Mahowald M, Wyckoff GJ, Malcom CM, Lahn BT: Accelerated
primary microcephaly: an analysis of locus heterogeneity and
evolution of nervous system genes in the origin of Homo
phenotypic variation. J Med Genet 2002, 39:718-721.
sapiens. Cell 2004, 119:1027-1040.
30. Bond J, Scott S, Hampshire DJ, Springell K, Corry P,
13. Evans PD, Anderson JR, Vallender EJ, Choi SS, Lahn BT:
Abramowicz MJ, Mochida GH, Hennekam RC, Maher ER, Fryns JP
 Reconstructing the evolutionary history of microcephalin,
et al.: Protein-truncating mutations in ASPM cause variable
a gene controlling human brain size. Hum Mol Genet 2004,
reduction in brain size. Am J Hum Genet 2003, 73:1170-1177.
13:1139-1145.
These investigators showed that the microcephalin amino acid sequence 31. Kumar A, Blanton S, Babu M, Markandaya M, Girimaji S: Genetic
has been modified adaptively during its evolution from simian ancestors analysis of primary microcephaly in Indian families: novel
to humans and chimpanzees. ASPM mutations. Clin Genet 2004, 66:341-348.
14. Mcreary BD, Rossiter JP, Robertson DM: Recessive (true) 32. Schultz J, Milpetz F, Bork P, Ponting CP: SMART, a simple
microcephaly: a case report with neuropathological modular architecture research tool: identification of signaling
observations. J Intellect Disabil Res 1996, 40:66-70. domains. Proc Natl Acad Sci USA 1998, 95:5857-5864.
15. Wood B, Collard M: The human genus. Science 1999, 33. Huyton T, Bates PA, Zhang X, Sternberg MJ, Freemont PS:
284:65-71. The BRCA1 C-terminal domain: structure and function.
Mutat Res 2000, 460:319-332.
16. Bundey S: Microcephaly. In Genetics and Neurology, 2nd edn.
Edited by: Churchill Livingstone; 1992:20-24. Emery AEH (Series 34. Yu X, Chini CC, He M, Mer G, Chen J: The BRCT domain is a
Editor): Genetics in Medicine and Surgery. phospho-protein binding domain. Science 2003, 302:639-642.
17. Trimborn M, Bell SM, Felix C, Rashid Y, Jafri H, Griffiths PD, 35. Xu X, Lee J, Stern DF: Microcephalin is a DNA damage response
 Neumann LM, Krebs A, Reis A, Sperling K et al.: Mutations in  protein involved in regulation of CHK1 and BRCA1. J Biol Chem
microcephalin cause aberrant regulation of chromosome 2004, 279:34091-34094.
condensation. Am J Hum Genet 2004, 75:261-266. Epitope-tagged microcephalin proteins were observed by the authors to
Patients with microcephaly caused by mutations in microcephalin were localise to ionising radiation-induced foci, suggesting a role in DNA
shown in this study to have an associated cellular phenotype of aberrant damage repair. Further work using RNA interference-mediated inhibition
regulation in chromosome condensation. Cells deficient in microcephalin of microcephalin expression, found that depletion of microcephalin led to
spend extended periods of time with chromosome condensation in the G2 reduced levels of Brca1 and Chk1, and also defective intra-S and G2/M
phase of the cell cycle. Furthermore, chromosome condensation con- checkpoints after ionising radiation.
tinues for some time post-mitosis.
36. Lin SY, Elledge SJ: Multiple tumor suppressor pathways
18. Bond J, Roberts E, Mochida GH, Hampshire DJ, Scott S,  negatively regulate telomerase. Cell 2003, 113:881-889.
Askham JM, Springell K, Mahadevan M, Crow YJ, Markham AF Microcephalin was isolated in a cellular screen for negative regulators of
et al.: ASPM is a major determinant of cerebral cortical size. hTERT transcription. The authors called the gene BRIT1 (BRCT-repeat
Nat Genet 2002, 32:316-320. inhibitor of hTERT expression) and demonstrated that small interfering
19. Mochida GH, Walsh CA: Molecular genetics of human RNA of BRIT1 (microcephalin) could derepress hTERT expression in
microcephaly. Curr Opin Neurol 2001, 14:151-156. U2OS cells.

20. Komai T, Kishimoto K, Ozaki Y: Genetic study of microcephaly 37. Gao Y, Sun Y, Frank KM, Dikkes P, Fujiwara Y, Seidl KJ,
based on Japanese material. Am J Hum Genet 1955, 7:51-65. Sekiguchi JM, Rathbun GA, Swat W, Wang J et al.: A critical role
for DNA end-joining proteins in both lymphogenesis and
21. Jackson AP, McHale DP, Campbell DA, Jafri H, Rashid Y, neurogenesis. Cell 1998, 95:891-902.
Mannan J, Karbani G, Corry P, Levene MI, Mueller RF et al.:
Primary autosomal recessive microcephaly (MCPH1) maps 38. Hakem R, de la Pompa JL, Sirard C, Mo R, Woo M, Hakem A,
to chromosome 8p22-pter. Am J Hum Genet 1998, Wakeham A, Potter J, Reitmair A, Billia F et al.: The tumor
63:541-546. suppressor gene Brca1 is required for embryonic cellular
proliferation in the mouse. Cell 1996, 85:1009-1023.
22. Jamieson CR, Govaerts C, Abramowicz MJ: Primary autosomal
recessive microcephaly: homozygosity mapping of MCPH4 to 39. Evans PD, Anderson JR, Vallender EJ, Gilbert SL, Malcom CM,
chromosome 15. Am J Hum Genet 1999, 65:1465-1469.  Dorus S, Lahn BT: Adaptive evolution of ASPM, a major

www.sciencedirect.com Current Opinion in Genetics & Development 2005, 15:241248


248 Genetics of disease

determinant of cerebral cortical size in humans. Hum Mol 47. Springer MS, Murphy WJ, Eizirik E, OBrien SJ: Placental
Genet 2004, 13:489-494. mammal diversification and the CretaceousTertiary
See annotation [51]. boundary. Proc Natl Acad Sci USA 2003, 100:1056-1061.
40. do Carmo Avides M, Glover DM: Abnormal spindle protein, Asp, 48. Russo CA, Takezaki N, Nei M: Molecular phylogeny and
and the integrity of mitotic centrosomal microtubule divergence times of drosophilid species. Mol Biol Evol 1995,
organizing centers. Science 1999, 283:1733-1735. 12:391-404.
41. Riparbelli MG, Callaini G, Glover DM, Avides Mdo C: A 49. Manke IA, Lowery DM, Nguyen A, Yaffe MB: BRCT repeats as
requirement for the abnormal spindle protein to organise phosphopeptide-binding modules involved in protein
microtubules of the central spindle for cytokinesis in targeting. Science 2003, 302:636-639.
Drosophila. J Cell Sci 2002, 115:913-922.
50. Zhang J: Evolution of the human ASPM gene, a major
42. Gonzalez C, Saunders RD, Casal J, Molina I, Carmena M, Ripoll P,  determinant of brain size. Genetics 2003, 165:2063-2070.
Glover DM: Mutations at the asp locus of Drosophila lead to See annotation [51].
multiple free centrosomes in syncytial embryos, but restrict
centrosome duplication in larval neuroblasts. J Cell Sci 1990, 51. Kouprina N, Pavlicek A, Mochida GH, Solomon G, Gersch W,
96:605-616.  Yoon YH, Collura R, Ruvolo M, Barrett JC, Woods CG et al.:
Accelerated evolution of the ASPM gene controlling brain
43. Riparbelli MG, Massarelli C, Robbins LG, Callaini G: The abnormal size begins prior to human brain expansion. PLoS Biol 2004,
spindle protein is required for germ cell mitosis and oocyte 2:126.
differentiation during Drosophila oogenesis. Exp Cell Res 2004, This study, together with that of Evans et al. [39] and Zhang [50], identifies
298:96-106. the signatures of strong positive selection in the ASPM gene of great apes.
Impressively, the authors sequence and analyse complete primate ASPM
44. Kraut R, Chia W, Jan LY, Jan YN, Knoblich JA: Role of genes including their promoters, non-coding and coding sequences.
inscuteable in orienting asymmetric cell divisions in
Drosophila. Nature 1996, 383:50-55. 52. Pavlicek A, Noskov VN, Kouprina N, Barrett JC, Jurka J,
Larionov V: Evolution of the tumor suppressor BRCA1 locus
45. Chenn A, McConnell SK: Cleavage orientation and the in primates: implications for cancer predisposition. Hum Mol
asymmetric inheritance of Notch1 immunoreactivity in Genet 2004, 13:2737-2751.
mammalian neurogenesis. Cell 1995, 82:631-641.
53. Rehen SK, McConnell MJ, Kaushal D, Kingsbury MA, Yang AH,
46. Wang Y-q, Su B: Molecular evolution of microcephalin, a gene Chun J: Chromosomal variation in neurons of the developing
 determining human brain size. Hum Mol Genet 2004, and adult mammalian nervous system. Proc Natl Acad Sci USA
13:1131-1137. 2001, 98:13361-13366.
In addition to demonstrating rapid evolution of microcephalin in the
common ancestor of great apes, these authors predict four substitutions 54. Evolutionary history of a gene controlling brain size. PLoS Biol
that have arisen by positive selection in the human population. 2004, 2:e134.

Reproduction of material from Elsevier articles


Interested in reproducing part or all of an article published by Elsevier, or one of our article figures? If so, please
contact our Global Rights Department with details of how and where the requested material will be used. To submit
a permission request on-line, please visit:

www.elsevier.com/wps/find/obtainpermissionform.cws_home/obtainpermissionform

Alternatively, please contact:

Elsevier
Global Rights Department
PO Box 800
Oxford OX5 1DX, UK
Phone: (44) 1865-843830
Fax: (44) 1865-853333
permissions@elsevier.com

Current Opinion in Genetics & Development 2005, 15:241248 www.sciencedirect.com

S-ar putea să vă placă și