Você está na página 1de 8

REPORT

A Truncating Mutation of CEP135 Causes Primary


Microcephaly and Disturbed Centrosomal Function
Muhammad Sajid Hussain,1,2,3,4 Shahid Mahmood Baig,4 Sascha Neumann,2 Gudrun Nurnberg,1,3
Muhammad Farooq,4 Ilyas Ahmad,1,4 Thomas Alef,1 Hans Christian Hennies,1,5 Martin Technau,2
Janine Altmuller,1 Peter Frommolt,1,5 Holger Thiele,1 Angelika Anna Noegel,1,2,3,5,*
and Peter Nurnberg1,3,5,*

Autosomal-recessive primary microcephaly (MCPH) is a rare congenital disorder characterized by intellectual disability, reduced brain
and head size, but usually without defects in cerebral cortical architecture, and other syndromic abnormalities. MCPH is heterogeneous.
The underlying genes of the seven known loci code for centrosomal proteins. We studied a family from northern Pakistan with two
microcephalic children using homozygosity mapping and found suggestive linkage for regions on chromosomes 2, 4, and 9. We
sequenced two positional candidate genes and identified a homozygous frameshift mutation in the gene encoding the 135 kDa centro-
somal protein (CEP135), located in the linkage interval on chromosome 4, in both affected children. Post hoc whole-exome sequencing
corroborated this mutations identification as the causal variant. Fibroblasts obtained from one of the patients showed multiple and frag-
mented centrosomes, disorganized microtubules, and reduced growth rate. Similar effects were reported after knockdown of CEP135
through RNA interference; we could provoke them also by ectopic overexpression of the mutant protein. Our findings suggest an addi-
tional locus for MCPH at HSA 4q12 (MCPH8), further strengthen the role of centrosomes in the development of MCPH, and place
CEP135 among the essential components of this important organelle in particular for a normal neurogenesis.

Autosomal-recessive primary microcephaly (MCPH [MIM organization at the centrosome, resulting in altered cell
251200]) is a neurodevelopmental disorder characterized division and cortical development. Despite their similar
by reduced size of the cerebral cortex and mild to moderate subcellular localization, the biological functions of these
intellectual disability whereas the architecture of the brain genes may vary.1,3,4,10
is largely normal. Head circumference is already reduced at We studied a consanguineous family from northern
birth and usually more than 3 SD below the age- and sex- Pakistan; the two affected children had primary micro-
matched population mean throughout the patients life- cephaly at birth (Figures 1A and 1B). After informed
time. Many patients have a receding forehead. MCPH is consent of the parents was obtained, pedigree information
heterogeneous; it has seven known loci, MCPH1 was documented and clinical data and blood samples were
MCPH7,1 each of which is associated with an underlying collected from the affected siblings, their father, and an
genetic defect: MCPH1 (MIM 251200) is associated with unaffected sibling. Each affected child had a sloping fore-
mutations of MCPH1 (MIM 607117);2 MCPH2 with or head. By 5 years of age they showed severe cognitive defi-
without cortical malformations (MIM 604317) is associ- cits; their speech was not understandable. They were
ated with mutations of WDR62 (MIM 613583);3,4 unable to form sentences and even to say any clear words,
MCPH3 (MIM 604804) is associated with mutations of although their hearing was not impaired. Other abnormal-
CDK5RAP2 (MIM 608201);5 MCPH4 (MIM 604321) is asso- ities were not apparent. Individual V-2 died at 11 years. The
ciated with mutations of CEP152 (MIM 613529);6 MCPH5 head circumference of the affected individuals ranged
(MIM 608716) is associated with mutations of ASPM (MIM between 12 and 14.5 SD compared to the average pop-
605481);7 MCPH6 (MIM 608393) is associated with muta- ulation of the same age and sex. The parents were healthy
tions of CENPJ (MIM 609279);5 and MCPH7 (MIM 612703) with normal head circumference. Ethical approval for this
is associated with mutations of STIL (MIM 181590).8 study was obtained from the ethics review board at the
Recently, a new locus at HSA 10q11.23-21.3 was described National Institute for Biotechnology and Genetic Engi-
in a consanguineous Turkish family, but the authors were neering in Faisalabad according to the Declaration of Hel-
not able to find the disease-causing gene variant.9 The sinki.
MCPH-associated genes described to date have been impli- Standard procedures were used to extract DNA from the
cated in cell division and cell cycle regulation, and many of blood samples for homozygosity mapping. Initially, we
the corresponding gene products are localized to the excluded all known MCPH loci in the family, using STR
centrosome. It has been speculated that they affect neural markers to demonstrate heterozygosity in the relevant
progenitor cell number through disturbed microtubule genomic regions. We then performed a genome-wide

1
Cologne Center for Genomics (CCG), University of Cologne, 50931 Cologne, Germany; 2Institute of Biochemistry I, Medical Faculty, University of
Cologne, 50931 Cologne, Germany; 3Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; 4Health Biotech-
nology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad 38000, Pakistan; 5Cologne Excellence Cluster on
Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50674 Cologne, Germany
*Correspondence: noegel@uni-koeln.de (A.A.N.), nuernberg@uni-koeln.de (P.N.)
DOI 10.1016/j.ajhg.2012.03.016. 2012 by The American Society of Human Genetics. All rights reserved.

The American Journal of Human Genetics 90, 871878, May 4, 2012 871
Figure 1. Identification of an MCPH-Causing Mutation in CEP135
(A) Clinical features of two siblings of a family from northern Pakistan presenting with microcephaly, sloping forehead, and retrogna-
thia. Informed consent to publish the photographs was obtained from the subjects parents.
(B) A simplified pedigree of the Pakistani family. Filled circles indicate individuals with MCPH. Their parents are second cousins. Only
the core family is shown. DNA was available for individuals IV-1, V-1, V-2, and V-3.
(C) G-banded chromosome 4 showing the position of the linkage interval at 4p14-4q12 along with the position of CEP135. The homo-
zygous region was delimited by the markers SNP_A-2154951 (rs12498424, physical position 40,631,476 bp) and SNP_A-1894332
(rs13134527, physical position 58,702,130 bp).
(D) Sequence chromatograms of a part of CEP135 exon 8 as obtained by Sanger sequencing. Traces of the male patient (V-1) and his
father (IV-1) show the mutation c.970delC in homozygous and heterozygous status, respectively. The mutant sequences are shown
along with a wild-type trace from a control individual.
(E) Schematic representation of the genomic structure of human CEP135. The 26 exons of CEP135 are drawn to scale, while introns show
just artificial lines. Black boxes represent untranslated regions. The position of mutation c.970delC in exon 8 is indicated.
(F) CEP135 structure as predicted by SMART (Simple Modular Architecture Research Tool) database. This software predicted six coiled-
coil domains (orange rods) which cover almost the entire region of the protein. The position of mutation p.Q324Sfs*2 in the fourth
coiled-coil domain is indicated.

linkage analysis using the Affymetrix GeneChip Human hg19). The genes were prioritized with Endeavour and
Mapping 250K Sty Array. Data handling, evaluation, and GeneWanderer.12,13 Highly ranked genes were further
statistical analysis were performed as described previ- scrutinized manually with the NCBI, Ensembl, and UCSC
ously.11 We observed three peaks on chromosomes 2, 4, genome databases. Finally, we gave top priority to genes
and 9 that were suggestive for linkage with maximum mul- that were very likely to have important functions related
tipoint LOD scores of 2.07, 2.53, and 2.53, respectively to cell division or chromosome segregation and decided
(Figure S1). The underlying common homozygous regions to sequence the following two strong candidate genes
defined a candidate region of 8.6 Mb on chromosome first: the cell division cycle-14 homolog B gene (CDC14B
2 (90,183,48498,795,792; hg19), a candidate region of [MIM 603505]) at cytoband 9q22.32-q31.1 and the gene
18.1 Mb on chromosome 4 (40,631,47658,702,130; encoding the 135 kDa centrosomal protein (CEP135
hg19), and a candidate region of 8.8 Mb on chromosome [MIM 611423]) at HAS 4p14-q12 (Figure 1C).
9 (92,274,161101,122,314; hg19). Altogether these All exons and the intron-exon boundaries of CEP135
regions include more than 200 annotated known and and CDC14B were sequenced in the two affected individ-
predicted coding genes (UCSC Genome Bioinformatics, uals. The PCR products (primers are listed in Table S1)

872 The American Journal of Human Genetics 90, 871878, May 4, 2012
were sequenced bidirectionally with a BigDye Terminator Project, Seattle) (Table S3). Only three other homozygous
v1.1 cycle sequencing kit on an ABI3730xl automated variants resisted our filter criteria in addition to CEP135
DNA sequencer. Sequences were analyzed with DNASTAR c.970delC. None of these could be assumed to be relevant
(Lasergene) and Mutation Surveyor (SoftGenetics). We for the phenotype (Table S4). Likewise, we could not find
found no mutation in CDC14B but a homozygous single deleterious mutations in any of the seven known MCPH-
base-pair (bp) deletion (c.970delC) in exon 8 of CEP135. associated genes, even when allowing for compound
We found the homozygous 1 bp deletion in both affected heterozygosity during filtering.
children, but not in the unaffected child, whereas the The deletion c.970delC of CEP135 results in a frameshift,
father was heterozygous for this mutation (Figure 1D), changing glutamine at position 324 into serine, immedi-
which is compatible with recessive inheritance. Moreover, ately followed by a premature termination codon
the mutation is unlikely to be a polymorphism, as it is not (p.Gln324Serfs*2). This truncating mutation is obviously
listed in dbSNP, and we could not find it when testing 384 incompatible with a normal function of CEP135 if one
healthy Pakistani controls with pyrosequencing. For this considers the size of the protein and the position of
purpose, PCR primers (listed in Table S2) were designed the mutation (Figures 1E and 1F). CEP135 consists of 26
by the PSQ Assay Design program v.1.0.6 (QIAGEN, exons, and its open reading frame codes for a polypeptide
Hilden, Germany). Pyrosequencing was done according of 1,140 amino acids.
to the manufacturers instructions on a PSQ HS96A instru- CEP135 is a conserved a-helical protein which is present
ment (QIAGEN) with the use of PyroMark Gold Q96 at the centrosome throughout the cell cycle. Electron-
Reagents (QIAGEN). The data were analyzed by Pyro microscopic studies showed its association with the
Q-CpG v.1.0.9 analysis software (QIAGEN). pericentriolar material, an electron-dense material sur-
In an attempt to identify a second independent muta- rounding the centrioles. Reducing CEP135 amounts in
tion of CEP135, we sequenced CEP135 in patients from cells via RNA interference caused a disorganization of
seven other families affected with MCPH from northern interphase and mitotic spindles, leading to the hypothesis
Pakistan in which all known MCPH-associated genes that CEP135 has a role in maintaining the structure and
were previously excluded. No CEP135 mutation was found organization of the centrosome and of microtubules.15
in any of these families. Therefore, we decided to perform More recently the protein was identified as a centriolar
whole-exome sequencing of the affected boy (individual component; it functions in centriole biogenesis and
V-1) of the family presented in Figure 1B in order to presumably has a scaffolding role.16 Centrioles are core
demonstrate that there were no other mutations in rele- components of animal centrosomes and act as basal bodies
vant genes that might also explain the phenotype. We to assemble cilia and flagella.17
fragmented 1 mg of DNA using sonification technology To unravel the effect of the mutation on the cellular
(Covaris, Woburn, MA, USA). The fragments were end re- level, we analyzed control and patient fibroblasts. Biopsies
paired and adaptor ligated. After size selection, the library were taken from the affected individual V-1 (Figure 1B) and
was subjected to the enrichment process. We chose the Seq- healthy individuals. Tissues were cleaned with antiseptic
Cap EZ Human Exome Library v2.0 kit from NimbleGen agent (Betaisodona, Mundipharma) and incubated over-
(Roche NimbleGen, Madison, WI, USA) and analyzed the night with Dispase II (1.5 U/ml, Roche) diluted in PBS,
sample on an Illumina HiSeq 2000 sequencing instrument. pH 6.8, at 4 C to separate the intact epidermis from the
About 10 Gb of sequence were produced for this sample by dermis. The dermis was incubated in Dulbeccos modified
loading it individually on one lane of a flow cell and gener- Eagles medium (DMEM) at 37 C. After one week, fibro-
ating paired-end reads of 2 3 100 bp. This resulted in a very blasts were detected. The pool of fibroblasts was increased
high coverage; i.e., > 303 for nearly 92% of the target by additional culturing. Primary fibroblasts established
sequences, which comprised about 44 Mb. Primary data from the patient grew very slowly. To enhance growth,
were filtered according to signal purity by the Illumina DMEM with 15% fetal bovine serum was used. Patient
Realtime Analysis (RTA) software v1.8. Subsequently, the and wild-type primary fibroblasts were then cultured on
reads were mapped to the human genome reference build 12 mm coverslips and fixed with 3% paraformaldehyde.
hg19 via the ELANDv2 alignment algorithm on a multi- For staining of microtubules, cells were incubated for
node compute cluster. With the use of CASAVA v1.8, PCR 15 min in tubulin stabilization buffer, which is composed
duplicates were filtered out, and the output was converted of Hanks buffer (137 mM NaCl, 5 mM KCl, 1.1 mM
into BAM format. Variant calling was performed with the Na2HPO4, 0.4 mM KH2PO4, 5 mM Glucose, 4 mM
use of SAMtools (version 0.1.7) for indel detection.14 NaHCO3) containing 1 mM MES, pH 6.8, 2 mM EGTA,
Scripts developed in-house at the Cologne Center for and 2 mM MgCl2. Permeabilization was done by 0.5%
Genomics were applied to detect protein changes, affected Triton X-100 in 13 tubulin stabilization buffer for 4 min
splice sites, and overlaps with known variants. In partic- at room temperature. For g-tubulin detection, the cells
ular, we filtered the variants for high-quality unknown were fixed with prechilled methanol for 10 min at 20 C.
variants in the linkage intervals (dbSNP build 132 or the Subsequently, the fixed cells were treated three times with
1000 Genomes database; in-house variation database; and tubulin stabilization buffer. Blocking was done for 15 min
public Exome Variant Server, NHLBI Exome Sequencing with blocking buffer (13 PBG: PBS containing 5% BSA and

The American Journal of Human Genetics 90, 871878, May 4, 2012 873
Figure 2. Centrosome, Microtubule, and
Nuclear Shape Defects in CEP135 Microce-
phalic Patient Cells
(A) Immunofluorescence staining and
confocal microscopy images of wild-type
primary fibroblasts with well-organized
microtubules and a single centrosome de-
tected by g-tubulin. g-tubulin (turquoise)
and a-tubulin (green) antibodies were
used. DAPI (blue) was used for DNA stain-
ing. Scale bar, 5 mm.
(B) Abnormality of centrosome number
in CEP135 patient primary fibroblasts.
Interphase cell showing supernumerary
centrosomes. In this figure three centro-
somes were observed as detected by
g-tubulin (turquoise). Scale bar, 5 mm.
(C) Disorganized microtubule network in
patient fibroblasts. Scale bar, 20 mm.
(D) Control primary fibroblast showing
a well-shaped ellipsoid nucleus. Scale bar,
5 mm.
(E) Mutant fibroblast with a dysmorphic
nucleus. Scale bar, 10 mm.
(F) Graphical representation showing
the number of CEP135 mutant primary
fibroblasts with dysmorphic nuclei. About
20% of mutant fibroblasts harbored
misshapen nuclei whereas in control fibro-
blasts this number was only ~3%. Three
hundred cells of each, wild-type and
mutant, were counted. Error bars represent
SEM, p 3.44 3 1003 (Students t test).

the vicinity of the nucleus during


interphase (Figure 2A). In the
patients primary fibroblasts, the
centrosome number was increased in
more than 18% of the cells (Table
S5). In such cells we found 3, 4, or
5 centrosomes per cell (Figures 2B
and S2). Furthermore, centrosomes
0.45% fish gelatin). Primary antibodies were diluted in appeared fragmented (Figure S2). The microtubule network
blocking buffer and incubated overnight at 4 C. The was frequently disorganized (~55% of the cells), which was
following antibodies were used: mouse monoclonal anti accompanied by cell shape changes (Figures 2C and S3;
g-tubulin (Sigma-Aldrich, GTU-88; 1:300), rabbit poly- Table S5). We also observed misshapen and fragmented
clonal anti-pericentrin (Abcam, ab4448; 1:300), and rat nuclei (Figure 2E and S4). Statistical analysis showed
monoclonal anti a-tubulin (YL 1/2 1:20).18 After incuba- that ~20% of the mutant cells harbored misshapen nuclei
tion, samples were treated with 13 PBS three times for as compared to ~3% in control fibroblasts (Figure 2F; Table
5 min followed by secondary antibody incubation S5). Another prominent aspect of the patients primary
(1:1,000 diluted in blocking buffer) for one hour at room fibroblasts was the complete loss of centrosomes. Approx-
temperature. Alexa Fluor 568 goat anti-mouse IgG (Invitro- imately 22% of mutant primary fibroblasts were without
gen, A11004), Alexa Fluor 647 donkey anti-mouse IgG centrosomes as detected with g-tubulin, whereas this was
(Invitrogen, A31571), and Alexa Fluor 488 goat anti-rat never observed in control cells (Table S5; Figure S3). Most
IgG (Invitrogen, A11006) were used as secondary anti- cells with misshapen nuclei were also devoid of centro-
bodies. DNA was detected with DAPI (Sigma-Aldrich, somes (Figure S4).
D9564). Finally, the cells were mounted on glass slides For ectopic expression of wild-type and mutant
with Gelvatol. Images were taken with a confocal micro- (c.970delC) CEP135 fused to green fluorescent protein
scope (Leica, LSM TCS SP5). (GFP), we used COS-7 cells. Gateway Technology (Invitro-
Control primary fibroblasts had oval nuclei surrounded gen) was employed to clone wild-type (NM_025009.3)
by organized microtubules and a single centrosome in CEP135 cDNA. The CEP135 mutation (c.970delC) was

874 The American Journal of Human Genetics 90, 871878, May 4, 2012
Figure 3. Distribution of GFP-Tagged
Wild-Type and Mutant CEP135
(A) N-terminally GFP-tagged human
CEP135 (GFP-CEP135) was transiently
expressed in COS-7 cells. GFP-CEP135
is present in dots of variable size and
number. A single centrosome was detected
by pericentrin-specific antibodies (tur-
quoise). Scale bar, 5 mm.
(B) COS-7 cell transfected with GFP-tagged
mutant CEP135 (GFP-CEP135-mut), where
diffuse staining was detected. Scale bar,
5 mm.
(C) Abnormal microtubule network in
COS-7 expressing wild-type CEP135. Scale
bar, 5 mm.
(D) COS-7 cells expressing GFP-CEP135-
mut have a disorganized microtubule
network. Microtubules appeared broken
and concentrated near the nucleus. Scale
bar, 5 mm.

expression of both wild-type and


mutant GFP-tagged CEP135 in HaCaT
cells led to the presence of abnormal
microtubule networks. The severity
of the disorganization was more
pronounced in cells expressing the
mutant protein (Figure 3C and 3D).
Cells transfected with GFP-tagged
mutant CEP135 also harbored mul-
tiple centrosomes (up to 5), which
then resulted in multipolar spindle
formation. This phenotype was not
observed in the cells that overex-
pressed the wild-type protein. Inter-
estingly, the GFP-tagged CEP135 was
detected on microtubules (Figure 4).
These findings of a disorganized
introduced into the full-length cDNA with the use of the microtubule network and multiple centrosomes in cells
QuikChange II Site-Directed Mutagenesis Kit (Stratagene) transfected with GFP-tagged CEP135 resembled those
(primers are listed in Table S6) in an attempt to reproduce observed in mutant primary fibroblast cells.
the patients situation. Entry clones in pENTR/TEV/ The CEP135 mutation c.970delC is thought to lead to a
D-TOPO were transformed into Gateway destination truncation of the protein due to the introduction of a
vector pcDNA-DEST53, which has an N-terminal cycle-3 premature stop codon (p.Gln324Serfs*2). Alternatively,
GFP tag. Both wild-type and mutant plasmids (10 mg/ml) it might also trigger nonsense-mediated mRNA decay
were used for transfection of COS-7 cells. The Gene Pulser (NMD).19 We designed RT-PCR experiments to investigate
II (Bio-Rad) device was used for electroporation. 72 hr this (Table S7). The PAXgene Blood RNA system (QIAGEN)
after transfection, the cells were subjected to immuno- was used to extract RNA from patient and control blood
fluorescence. samples, and cDNA was synthesized with SuperScript III
When we analyzed the localization of the proteins and reverse transcriptase enzyme (Invitrogen). When primers
their effect on centrosomes and microtubule organization, were used to amplify a nearly full-length CEP135 cDNA
we observed wild-type GFP-tagged CEP135 at the centro- of ~3.4 kb, no PCR product was obtained with the patients
some where it colocalized with the centrosomal protein RNA, but only with the control sample, suggesting
pericentrin. We also detected it in some spots outside of pronounced degradation of mutant CEP135 mRNA
the centrosome that were not positive for pericentrin (Figure S5A). In contrast, a smaller PCR product of only
(Figure 3A). By contrast, we did not detect the mutant 385 bp, generated with primers just flanking the site of
protein on the centrosome; instead, in a few cells we mutation, was easily obtained from both control and
observed a diffuse cytoplasmic staining (Figure 3B). Over- mutant RNA (Figure S5B). A contamination of the mutant

The American Journal of Human Genetics 90, 871878, May 4, 2012 875
Figure 4. Centrosomal and Spindle Abnormalities in COS-7 Cells Transiently Expressing Wild-Type and Mutant CEP135
(A) A GFP-CEP135-expressing cell at metaphase showing bipolar spindles with two centrosomes.
(B) GFP-CEP135-mut-expressing cell with supernumerary centrosomes, which result in multipolar spindle formation. Centrosomes
were detected by pericentrin-specific antibodies (turquoise). Scale bar, 5 mm.

sample with wild-type RNA was excluded by sequencing a reduced growth rate.15 In mutant primary fibroblast cells,
of the PCR product. One explanation to reconcile these we have observed strongly reduced growth as well, which
apparently contradictory results might be an incomplete did not allow the acquisition of further data. The two
decay of the mutant mRNA. To verify this hypothesis, we known interaction partners of CEP135, the 50 kDa subunit
performed quantitative real-time PCR with cDNAs from of the dynactin complex (p50) and C-Nap1, have their
patient V-1 and his mother (IV-2), along with two different binding sites in the C-terminal domain of CEP135. A trun-
control samples. GAPDH and CEP135 gene-specific primers cation of CEP135 releases p50 and C-Nap1 and causes
(Table S8), 10 mM each, were mixed with SYBR Green decomposition of functional centrosomes and premature
PCR Master Mix (QIAGEN) and the respective cDNA. An centrosome splitting, respectively.23,24 Knockdown of
ANXA7 plasmid was used for calibration. Thermal cycler CEP135 leads to disorganized interphase and mitotic
conditions were 95 C for 15 min, 41 cycles of 94 C for spindle microtubules with bipolar and multipolar orienta-
30 s, 57 C for 45 s, and 68 C for 40 s. A DNA Engine tion.15 A role in procentriole formation has been uncov-
Opticon 2 (MJ Research, Bio-Rad) was used. The data ered wherein CEP135 and CPAP, also known as CENPJ
were analyzed with MJ Opticon Monitor software (version (MCPH6 protein), form a core structure within the prox-
3.1). We found a dramatic reduction in the CEP135 mRNA imal lumen of both parental and nascent centrioles.16
level in the patient as compared to the controls (Figure S6). The phenotype of multiple centrosomes has also been
The mothers CEP135 mRNA level was twice as high as that described in different patient and animal cells carrying
of the affected son but still significantly reduced when mutations in genes responsible for MCPH. Approximately
compared to the controls. These data corroborate the 25% of the cells of lymphoblastoid cell lines carrying
notion of a partial NMD effect caused by the mutation a mutation in MCPH1 harbored supernumerary centro-
c.970delC. somes.25 An aberrant centrosome number was also
CEP135 was identified as a centrosomal component observed in Mcph1 null DT40 cells after ionizing radiation
by proteomic analysis and shown to be present in the treatment.26 Mutant centrosomal protein CEP152 is the
pericentriolar matrix, around the centriolar surface, and cause of primary microcephaly MCPH4 and of Seckel
within the proximal lumen of the centrioles.16,20 In syndrome.6,27 Fibroblast cells of patients with Seckel syn-
Chlamydomonas, the CEP135 ortholog Bld10p localizes drome associated with CEP152 mutations also harbored
to the cartwheel, a 9-fold symmetrical structure that multiple centrosomes of variable size.27 Cdk5rap2 mutant
presumably functions as the scaffold for the centriole- mouse embryonic fibroblasts had supernumerary centro-
microtubule assembly and is responsible for achieving somes resulting in bipolar and multipolar spindles.28
the 9-fold symmetry.21 Bld10p mutants (bld10) show aber- A similar phenotype was seen in mouse neuronal progen-
rant interphase microtubules and mitotic spindles, defects itors mutated in the MCPH3-associated gene ortholog
in cell division, and a significant reduction of the growth Cdk5rap2.29 We have identified multiple centrosomes
rate.22 Knockdown of CEP135 in CHO cells also led to in primary CEP135 patient fibroblasts as well as in cells

876 The American Journal of Human Genetics 90, 871878, May 4, 2012
transfected with a plasmid encoding the mutant protein. Online Mendelian Inheritance in Man (OMIM), http://www.
The abnormal centrosome number supports CEP135s omim.org
role in centriole biogenesis, whereas a disorganization of SMART database, http://smart.embl-heidelberg.de/
the microtubule network points to its role at the centro- UCSC Genome Browser, http://genome.ucsc.edu
some as microtubule-organizing center.
In summary, we have shown a truncating mutation
of CEP135 to cause autosomal-recessive primary micro- References
cephaly in a Pakistani family. We suggest designation of 1. Kaindl, A.M., Passemard, S., Kumar, P., Kraemer, N., Issa, L.,
the corresponding locus at 4q12 MCPH8. The correspond- Zwirner, A., Gerard, B., Verloes, A., Mani, S., and Gressens, P.
ing protein, CEP135, is a centrosomal component and (2010). Many roads lead to primary autosomal recessive
further strengthens the role of centrosomes in the cause microcephaly. Prog. Neurobiol. 90, 363383.
of MCPH. In 18% and 22% of mutant fibroblasts, we 2. Jackson, A.P., Eastwood, H., Bell, S.M., Adu, J., Toomes, C.,
observed either the presence of multiple centrosomes or Carr, I.M., Roberts, E., Hampshire, D.J., Crow, Y.J., Mighell,
a complete loss of centrosomes, respectively. Centrosome A.J., et al. (2002). Identification of microcephalin, a protein
amplification most likely affects mitotic progression and implicated in determining the size of the human brain. Am.
J. Hum. Genet. 71, 136142.
mitotic spindle orientation and leads to multipolar spin-
3. Nicholas, A.K., Khurshid, M., Desir, J., Carvalho, O.P., Cox, J.J.,
dles, which result in the loss of progenitor cells. This has
Thornton, G., Kausar, R., Ansar, M., Ahmad, W., Verloes, A.,
been observed in MCPH1 and CEP152 mutant human cells et al. (2010). WDR62 is associated with the spindle pole and
as well as in Cdk5rap2 mutant mouse cells.2528 These is mutated in human microcephaly. Nat. Genet. 42, 1010
events can also affect the polarity of cell division in the 1014.
neural progenitor cells, leading to an altered number of 4. Yu, T.W., Mochida, G.H., Tischfield, D.J., Sgaier, S.K., Flores-
neural progenitor cells or an altered fate, which may be Sarnat, L., Sergi, C.M., Topcu, M., McDonald, M.T., Barry,
the cause of reduced neuron production. B.J., Felie, J.M., et al. (2010). Mutations in WDR62, encoding
a centrosome-associated protein, cause microcephaly with
simplified gyri and abnormal cortical architecture. Nat. Genet.
Supplemental Data 42, 10151020.
Supplemental Data include six figures and eight tables and can be 5. Bond, J., Roberts, E., Springell, K., Lizarraga, S.B., Scott, S.,
found with this article online at http://www.cell.com/AJHG/. Higgins, J., Hampshire, D.J., Morrison, E.E., Leal, G.F., Silva,
E.O., et al. (2005). A centrosomal mechanism involving
CDK5RAP2 and CENPJ controls brain size. Nat. Genet. 37,
Acknowledgments 353355.
6. Guernsey, D.L., Jiang, H., Hussin, J., Arnold, M., Bouyakdan,
We are grateful to all family members for their participation in this
K., Perry, S., Babineau-Sturk, T., Beis, J., Dumas, N., Evans,
study. We wish to thank Ursula Euteneuer and Ludwig Eichinger
S.C., et al. (2010). Mutations in centrosomal protein CEP152
for helpful discussion and Ingelore Bamann, Martina Munck,
in primary microcephaly families linked to MCPH4. Am. J.
and Alexandra Herzog for technical assistance. P.N. is a founder,
Hum. Genet. 87, 4051.
CEO, and shareholder of ATLAS Biolabs GmbH. ATLAS Biolabs
7. Bond, J., Roberts, E., Mochida, G.H., Hampshire, D.J., Scott, S.,
GmbH is a service provider for genomic analyses. This work was
Askham, J.M., Springell, K., Mahadevan, M., Crow, Y.J., Mark-
supported by grants from the Higher Education Commission
ham, A.F., et al. (2002). ASPM is a major determinant of
(HEC) of Pakistan, the German Academic Exchange Service
cerebral cortical size. Nat. Genet. 32, 316320.
(DAAD), and the Center for Molecular Medicine Cologne
(CMMC). 8. Kumar, A., Girimaji, S.C., Duvvari, M.R., and Blanton, S.H.
(2009). Mutations in STIL, encoding a pericentriolar and cen-
Received: November 17, 2011 trosomal protein, cause primary microcephaly. Am. J. Hum.
Revised: March 7, 2012 Genet. 84, 286290.
Accepted: March 15, 2012 9. Marchal, J.A., Ghani, M., Schindler, D., Gavvovidis, I., Win-
Published online: April 19, 2012 kler, T., Esquitino, V., Sternberg, N., Busche, A., Krawitz, P.,
Hecht, J., et al. (2011). Misregulation of mitotic chromosome
segregation in a new type of autosomal recessive primary
Web Resources microcephaly. Cell Cycle 10, 29672977.
10. Thornton, G.K., and Woods, C.G. (2009). Primary micro-
The URLs for the data presented herein are as follows:
cephaly: do all roads lead to Rome? Trends Genet. 25, 501510.
Endeavour, http://homes.esat.kuleuven.be/~bioiuser/endeavour/ 11. Borck, G., Ur Rehman, A., Lee, K., Pogoda, H.M., Kakar, N.,
tool/endeavourweb.php von Ameln, S., Grillet, N., Hildebrand, M.S., Ahmed, Z.M.,
Ensembl Genome Browser, http://www.ensembl.org Nurnberg, G., et al. (2011). Loss-of-function mutations of
Exome Variant Server, http://snp.gs.washington.edu/EVS/ ILDR1 cause autosomal-recessive hearing impairment
GeneWanderer, http://compbio.charite.de/genewanderer/ DFNB42. Am. J. Hum. Genet. 88, 127137.
GeneWanderer 12. Aerts, S., Lambrechts, D., Maity, S., Van Loo, P., Coessens, B.,
Marshfield genetic map, http://www.bli.uzh.ch/BLI/Projects/ De Smet, F., Tranchevent, L.C., De Moor, B., Marynen, P., Has-
genetics/maps/marsh.html san, B., et al. (2006). Gene prioritization through genomic
NCBI Map Viewer, http://www.ncbi.nlm.nih.gov/mapview/ data fusion. Nat. Biotechnol. 24, 537544.

The American Journal of Human Genetics 90, 871878, May 4, 2012 877
13. Kohler, S., Bauer, S., Horn, D., and Robinson, P.N. (2008). 22. Matsuura, K., Lefebvre, P.A., Kamiya, R., and Hirono, M.
Walking the interactome for prioritization of candidate (2004). Bld10p, a novel protein essential for basal body
disease genes. Am. J. Hum. Genet. 82, 949958. assembly in Chlamydomonas: localization to the cartwheel,
14. Li, H., Handsaker, B., Wysoker, A., Fennell, T., Ruan, J., Homer, the first ninefold symmetrical structure appearing during
N., Marth, G., Abecasis, G., and Durbin, R.; 1000 Genome assembly. J. Cell Biol. 165, 663671.
Project Data Processing Subgroup. (2009). The Sequence 23. Uetake, Y., Terada, Y., Matuliene, J., and Kuriyama, R. (2004).
Alignment/Map format and SAMtools. Bioinformatics 25, Interaction of Cep135 with a p50 dynactin subunit in
20782079. mammalian centrosomes. Cell Motil. Cytoskeleton 58, 5366.
15. Ohta, T., Essner, R., Ryu, J.H., Palazzo, R.E., Uetake, Y., and 24. Kim, K., Lee, S., Chang, J., and Rhee, K. (2008). A novel func-
Kuriyama, R. (2002). Characterization of Cep135, a novel tion of CEP135 as a platform protein of C-NAP1 for its centrio-
coiled-coil centrosomal protein involved in microtubule orga- lar localization. Exp. Cell Res. 314, 36923700.
nization in mammalian cells. J. Cell Biol. 156, 8799. 25. Alderton, G.K., Galbiati, L., Griffith, E., Surinya, K.H., Neitzel,
16. Kleylein-Sohn, J., Westendorf, J., Le Clech, M., Habedanck, R., H., Jackson, A.P., Jeggo, P.A., and ODriscoll, M. (2006). Regu-
Stierhof, Y.D., and Nigg, E.A. (2007). Plk4-induced centriole lation of mitotic entry by microcephalin and its overlap with
biogenesis in human cells. Dev. Cell 13, 190202. ATR signalling. Nat. Cell Biol. 8, 725733.
17. Azimzadeh, J., and Marshall, W.F. (2010). Building the 26. Brown, J.A.L., Bourke, E., Liptrot, C., Dockery, P., and Morri-
centriole. Curr. Biol. 20, R816R825. son, C.G. (2010). MCPH1/BRIT1 limits ionizing radiation-
18. Kilmartin, J.V., Wright, B., and Milstein, C. (1982). Rat mono- induced centrosome amplification. Oncogene 29, 55375544.
clonal antitubulin antibodies derived by using a new nonse- 27. Kalay, E., Yigit, G., Aslan, Y., Brown, K.E., Pohl, E., Bicknell,
creting rat cell line. J. Cell Biol. 93, 576582. L.S., Kayserili, H., Li, Y., Tuysuz, B., Nurnberg, G., et al.
19. Nicholson, P., and Muhlemann, O. (2010). Cutting the (2011). CEP152 is a genome maintenance protein disrupted
nonsense: the degradation of PTC-containing mRNAs. Bio- in Seckel syndrome. Nat. Genet. 43, 2326.
chem. Soc. Trans. 38, 16151620. 28. Barrera, J.A., Kao, L.R., Hammer, R.E., Seemann, J., Fuchs, J.L.,
20. Andersen, J.S., Wilkinson, C.J., Mayor, T., Mortensen, P., Nigg, and Megraw, T.L. (2010). CDK5RAP2 regulates centriole
E.A., and Mann, M. (2003). Proteomic characterization of the engagement and cohesion in mice. Dev. Cell 18, 913926.
human centrosome by protein correlation profiling. Nature 29. Lizarraga, S.B., Margossian, S.P., Harris, M.H., Campagna,
426, 570574. D.R., Han, A.P., Blevins, S., Mudbhary, R., Barker, J.E., Walsh,
21. Hiraki, M., Nakazawa, Y., Kamiya, R., and Hirono, M. (2007). C.A., and Fleming, M.D. (2010). Cdk5rap2 regulates centro-
Bld10p constitutes the cartwheel-spoke tip and stabilizes the some function and chromosome segregation in neuronal
9-fold symmetry of the centriole. Curr. Biol. 17, 17781783. progenitors. Development 137, 19071917.

878 The American Journal of Human Genetics 90, 871878, May 4, 2012

Você também pode gostar