Sei sulla pagina 1di 8

European Journal of Cell Biology 93 (2014) 98105

Contents lists available at ScienceDirect

European Journal of Cell Biology


journal homepage: www.elsevier.com/locate/ejcb

Mini review

Emerging roles of MCPH1: Expedition from primary


microcephaly to cancer
Thejaswini Venkatesh a, , Padmanaban S. Suresh b
a
Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, Karnataka, India
b
Centre for Biomedical Research, Vellore Institute of Technology University, Vellore, Tamil Nadu, India

a r t i c l e i n f o a b s t r a c t

Article history: Genetic mutations in microcephalin1 (MCPH1) cause primary autosomal recessive microcephaly which
Received 19 October 2013 is characterized by a marked reduction in brain size. MCPH1 encodes a centrosomal protein with three
Received in revised form 9 January 2014 BRCT (BRCA1 C-terminal) domains. Also, it is a key regulator of DNA repair pathway and cell cycle check-
Accepted 15 January 2014
points. Interestingly, in the past few years, many research studies have explored the role of MCPH1, a
neurodevelopmental gene in several cancers and its tumor suppressor functions have been elucidated.
Keywords:
Given the diverse new emerging roles, it becomes critical to review and summarize the multiple roles of
MCPH1
MCPH1 that is currently lacking in the literature. In this review after systematic analysis of literature, we
BRIT1
Autosomal recessive primary microcephaly
summarise the multiple functional roles of MCPH1 in centrosomal, DNA repair and apoptotic pathways.
DNA repair Additionally, we discuss the considerable efforts taken to understand the implications of MCPH1 in dis-
Cancer eases such as primary microcephaly and its other emerging association with cancer and otitis media. The
Otitis media promising view is that MCPH1 has distinct roles and its clinical associations in various diseases makes it
Apoptosis an attractive therapeutic target.
Glioblastoma 2014 Elsevier GmbH. All rights reserved.
OSCC
miR-27a

Introduction protein 2), CASC5 (cancer susceptibility candidate 5), CEP152 (cen-
trosomal protein 152 kDa), ASPM (asp [abnormal spindle] homolog,
The rare genetically heterogenous autosomal recessive pri- microcephaly associated [Drosophila]), CENPJ (centromeric protein
mary microcephaly, also called as MCPH (Micro-Cephaly Primary J), STIL (SCL/TAL1-interrupting locus), CEP135 (centrosomal pro-
Hereditary), is characterized by the clinical features such as OFC tein 135 kDa), CEP63 (centrosomal protein 63 kDa), ZNF335 (zinc
(occipital frontal circumference) of less than 3 SDs below the nger protein 335), PHC1 (polyhomeotic homolog 1 [Drosophila])
mean for age and sex, mild to moderate mental retardation and, and MKL2 (MKL/myocardin-like 2) (Bond et al., 2002; Jackson et al.,
with no further neurological ndings except for mild seizures 2002; Bond et al., 2005; Kumar et al., 2009; Guernsey et al., 2010;
(Passemard et al., 2009; Kaindl et al., 2010). However, the true Nicholas et al., 2010; Sir et al., 2011; Hussain et al., 2013; Yang et al.,
phenotype spectrum of MCPH patients is wider than indicated pre- 2012; Awad et al., 2013; Ramos et al., 2013). Among these genes,
viously and now, includes a simplied gyral pattern, periventricular MCPH1 was the rst gene to be identied as a causative gene for
neuronal heterotopias (abnormal positioning of the neurons in MCPH (Jackson et al., 1998; Jackson et al., 2002). It was mapped to
the brain), polymicrogyria (development of numerous microgyri), chromosome 8p22-pter using autozygosity mapping in two con-
speech delay, hyperactivity and attention decit, aggressiveness, sanguineous Pakistani families. Jackson et al. (1998) identied a
focal or generalized seizures, and delay of developmental mile- minimal region of 13.2 cM, dened by the markers D8S1824 and
stones and pyramidal signs (Kaindl et al., 2010). Totally, mutations D8S1825D8S and D8S1825 and subsequently, identied MCPH1 as
in thirteen genes have been identied to cause MCPH. They the causative gene. In this study, both the consanguineous families
include MCPH1 (microcephalin 1), WDR62 (WD repeat domain showed a single homozygous mutation c.74C>G(p.Ser25X) in the
62), CDK5RAP2 (cyclin-dependent kinase 5 regulatory associated second exon of MCPH1(Jackson et al., 2002). Also, another homozy-
gous missense mutation c.80C>G(p.Tyr27Arg) in a MCPH patient
of Caucasian origin was identied (Trimborn et al., 2005). Eight
mutations have been described for MCPH1 in a study consisting
Corresponding author at: Department of Molecular Reproduction, Development
of 112 Iranian families with MCPH (Darvish et al., 2010). Addi-
and Genetics, Indian Institute of Science, Bangalore 560012, India.
Tel.: +91 9663068681.
tionally, three more mutations in MCPH1 have been reported in
E-mail address: thejaswinivenkatesh5@gmail.com (T. Venkatesh). MCPH patients (Farooq et al., 2010; Ghani-Kakhki et al., 2012;

0171-9335/$ see front matter 2014 Elsevier GmbH. All rights reserved.
http://dx.doi.org/10.1016/j.ejcb.2014.01.005
T. Venkatesh, P.S. Suresh / European Journal of Cell Biology 93 (2014) 98105 99

Hussain et al., 2013). In addition to MCPH, it is interesting to note MCPH1 localizes to centrosomes in U2OS (osteosarcoma) and
that mutations in MCPH1 have been associated with other disease DT-40 (chicken B) cells (Zhong et al., 2006; Jeffers et al., 2008). It also
conditions where microcephaly is one of the distinct features. A localizes to DNA repair foci in response to DNA damage in U2OS,
homozygous mutation c.427 428insA(p.Thr143fsX5) in MCPH1 has HeLa (cervical carcinoma), and microcephalic patient derived lym-
been reported in PCC (premature chromosome condensation) syn- phoblastoid cells (Rai et al., 2006; Wood et al., 2007; Gavvovidis
drome which is characterized by microcephaly and postnatal short et al., 2010). In addition to cell lines, the localization of MCPH1
stature. PCC is a phenomenon where mitotic chromosomes con- has been studied in human tissue sections. A study has shown that
dense at the interphase stage itself (Neitzel et al., 2002; Trimborn MCPH1 localizes to nuclear foci, the nucleus and the cytoplasm in
et al., 2004). Moreover, a deletion of 150200 kb encompassing primary cultures of normal ovarian tissues (Brning-Richardson
the promoter and rst 6 exons of MCPH1 has been reported in an et al., 2011). A study of IHC (immunohistochemistry) reported
Iranian consanguineous family with autosomal recessive mental that MCPH1 localization was conned to the nucleus and nuclear
retardation (ARMR) and mild microcephaly (Garshasbi et al., 2006). foci of normal breast tissues (Richardson et al., 2011). On a more
Although it is widely studied in relation to primary microcephaly, physiological scenario, localization of MCPH1 has been studied in
recent studies suggest correlation of MCPH1 mutations and its dys- Drosophila. MCPH1 localizes to the interphase DNA in syncytial
functions with cancer and otitis media (OM), and this has further embryos of Drosophila, and it is absent on the DNA of cells in the
strengthened its implications in different tissues. Its identication prometaphase to telophase stages of Drosophila syncytial embryos
and clinical correlation has also suggested that they have diverse (Brunk et al., 2007).
roles apart from neurodevelopment. Given the important roles of
MCPH1 and its emerging correlations with cancer biology and other
Phenomics of MCPH1 model organisms
diseases, a greater understanding of its role is undeniably needed.
In this review, considering the recent advances, we highlight its
Drosophila as a model organism
structure, expression and localization, targets and functions, and
recent insights into its role in cancer development and OM.
The full-length MCPH1 gene in Drosophila is designated as
MCPH1 (L). The MCPH1 (L) protein contains an N-terminal and two
Structure tandem C-terminal BRCT domains. MCPH1 (L) undergoes alterna-
tive splicing to give three more isoforms. The short isoform, MCPH1
The MCPH1 gene has 14 coding exons (Jackson et al., 2002). (S) contains the N-terminal BRCT domain. The other two isoforms
Recently, four alternative transcripts of human MCPH1 have been are short of 47 amino acids at their N-terminal ends and are sim-
identied in 562T broblast cells. The isoforms include: full- ilar to MCPH1(L) and MCPH1(S) isoforms (Brunk et al., 2007). The
length (FL) and the alternative transcripts 914 (914 exons are MCPH1 transcripts (S and L forms) show highest expression at the
deleted), 13 (13 exons are deleted), and 8 (exon 8 is deleted) syncitial and gastrula stages and, lowest expression at the blas-
(Gavvovidis et al., 2012). The human MCPH1 protein is 835 amino tula stage of the y development. MCPH1 protein is expressed at
acids long and shows 57% sequence identity with the mouse pro- a higher level in ovaries and a lower level in the brain of the lar-
tein of 822 amino acids (Jackson et al., 2002; Kumar et al., 2002). It vae (Brunk et al., 2007). Furthermore, two Drosophila lines with
consists of three BRCT (BRCA1 C-terminal) domains which extend the homozygous deletion of either 13 or 14 exons have been
from amino acids 7 to 83 (BRCT1), 642 to 720 (BRCT2) and 753 to generated (Brunk et al., 2007). These mutant ies were viable and
823 (BRCT3) (Jackson et al., 2002). BRCT2 and BRCT3 constitute the showed no abnormalities of the brain. In contrast to this study, Rick-
C-terminal twin BRCT domains. BRCT domains are a new class of myre et al. (2007) reported that the MCPH1 mutant homozygous
phosphoprotein binding modules that are found in the components male ies showed defective brain structures ranging from mal-
of DNA repair pathways (Huyton et al., 2000; Glover et al., 2004). formed lobes to complete absence of the lobes, though the sizes
MCPH1 protein has a NLS (nuclear localization signal) from 355 to of the brain were unaffected. But, the MCPH1 mutant homozy-
375 amino acids, and deletion of amino acid residues 301400 of gous female ies showed no defects in the size or the structures
MCPH1 leads to the detainment of MCPH1 within the cytoplasm of the brain (Rickmyre et al., 2007). Also, the mutant ies showed
of HEK293T (human embryonic kidney cells) (Wood et al., 2007). no morphological or behavioural defects in adults (Brunk et al.,
The evolutionary signicance of the MCPH1 protein has been stud- 2007). However, the embyros laid by the MCPH1 null female ies
ied. The MCPH1 protein from 11 species (7 primates and 4 other lacked cellularization and showed defective synticial embryogen-
mammalian species) were aligned to gain insights into the evolu- esis (Brunk et al., 2007).
tionary signicance of the protein (Shi et al., 2013). The amino acid
residues (M96, S101, V310, H314, T377, Y425, L442, R485 and P835)
Mouse as a model organism
of the human protein were conserved across the species. However,
the amino acid residues (I161, E167, A510 and S841) occurred in
Few groups have elucidated the functions of MCPH1 in mouse
the ancestor of hominidae and hence, these are great-ape specic
models. A mouse model with impaired Mcph1 function (referred
residues (Shi et al.., 2013).
as Mcph1gt/gt mouse) was generated using the gene trap method
(Trimborn et al., 2010). Trimborn et al. (2010) introduced a gene
Expression and localization trap in intron 12 of Mcph1, which led to the deletion of C-
terminal BRCT domain. In this study, the body and brain sizes
MCPH1 is ubiquitously expressed in adult human tissues, and of Mcph1gt/gt mice were within the range of wild type controls.
its highest expression is observed in the brain, testes, pancreas and However, the overall survival rates of Mcph1gt/gt mice were sig-
liver (Lin et al., 2010). Its transcripts are highly expressed in human nicantly reduced compared to wild type and heterozygous mice.
fetal brain, kidney and liver, and expressed less in human fetal heart No signs of premature malignant disease or impaired fertility were
and lungs (Jackson et al., 2002). Mcph1 has been detected in the reported in Mcph1gt/gt mice (Trimborn et al., 2010). Surprisingly,
lateral ventricles of the developing forebrain of the fetal mouse the cell cultures derived from th Mcph1gt/gt mice appeared to be
brain (Jackson et al., 2002). Additionally, MCPH1 transcripts have largely normal after irradiation and showed no signs of defective
been detected at all developmental stages from embryos to adults DNA damage responses (Trimborn et al., 2010). However, the pri-
in Drosophila (Brunk et al., 2007). mary broblast cultures of the homozygous Mcph1 mutant mice
100 T. Venkatesh, P.S. Suresh / European Journal of Cell Biology 93 (2014) 98105

exhibited PCC. Also, the authors claim that these discrepancies of abrogated with the disruption of Pcnt (Wang et al., 2013). MCPH1
the Mcph1 functions in Mcph1gt/gt mice is due to the residual wild null blood lymphocytes derived from patients with MCPH and PCC
type Mcph1 expression which was indeed detected by the quan- syndrome exhibit PCC (Trimborn et al., 2004; Trimborn et al., 2005).
titative real-time RT-PCR in the spleen, brain and liver (Trimborn PCC is caused by the lack of a functional N-terminal BRCT domain of
et al., 2010). Liang et al. (2010) generated a Mcph1 global knock- MCPH1 (Wood et al., 2008; Richards et al., 2010; Yamashita et al.,
out mouse by deleting the exon 2 using the cre/lox system. These 2011). Also, centrosomal function of MCPH1 has been associated
Mcph1/ mice survived to adulthood, but were growth-retarded with PCC. MCPH1 interacts with PCNT (pericentrin) and directs
and infertile (Liang et al., 2010). However, the Mcph1/ mice did CHEK1 (checkpoint kinase 1) to centrosomes (Tibelius et al., 2009).
not develop any tumors over a period of one and a half years. Mouse CHEK1 mediates the inhibitory phosphorylation of centrosomal
embryonic broblasts (MEFs) and T lymphocytes derived from CDK1 (cyclin-dependent kinase 1) at Y15. CDK1 and CCNB1 (cyclin
these Mcph1/ mice were hypersensitive to -irradiation (Liang B1) accumulate in the centrosome during interphase, and activa-
et al., 2010). Furthermore, a failure to carry out meiotic recombi- tion of the CDK1-CCNB1 complex is the key event to enter into
nation was observed in spermatocytes, and this failure has been mitosis (Nurse, 1990; Bailly et al., 1992). The centrosomal activa-
attributed to the phenomenon of infertility seen in these knockout tion of CDK1 and CCNB1 occurs in the late prophase (Jackman et al.,
mice (Liang et al., 2010). Another Mcph1/ mice model was gener- 2003). Regulation of centrosomal CDK1 and CCNB1 is very critical
ated by the disruption of the Mcph1 gene using the cre/lox system to synchronise the cytoplasmic and nuclear mitotic events. In the
(Gruber et al., 2011). Their study demonstrated that the Mcph1- absence of MCPH1 in MCPH1 patients, the recruitment of CHEK1
del (Mcph1 knockout) mice showed reduced brain size and less to the centrosomes by MCPH1 is restricted and hence, CDK1 is
neuroprogenitor cell proliferation. Gruber et al. (2011) developed not inhibited by CHEK1. The MCPH1 mutant PCC cells exhibit stri-
neurospheres from both the Mcph1-del and Mcph1-ctr (littermate kingly low levels of pY15-CDK1 (Alderton et al., 2006). This leads
controls) mice and reported that the number and size of the neu- to the early activation of the CCNB1-CDK1 complex and hence,
rospheres were not affected in Mcph1-del mice as compared to mitosis starts prematurely leading to PCC (Tibelius et al., 2009).
that of the Mcph1-ctr mice. However, the tendency to form sec- Additionally, CDC25 phosphatases regulate CDK1-CCNB1 activity
ondary neurospheres was decreased in the Mcph1-del mice when by reversing inhibitory Y15 phosphorylation on CDK1, and the low
compared to the neurospheres derived from the Mcph1-ctr mice. levels of pY15-CDK1 levels were attributed to lack of CHEK1 medi-
Cells of the neurospheres derived from Mcph1-del mice underwent ated proteosomal degradation of CDC25 phosphatases (Alderton
premature mitotic entry, which eventually lead to the uncoupling et al., 2006). Thus, MCPH1 regulates mitotic entry by regulating the
of the mitosis and the centrosomal cycle, leading to misorienta- activities of CDC25A and CDK1-CCNB1 (Alderton et al., 2006).
tion of the mitotic spindles (Gruber et al., 2011). Misorientation of Additionally, another non-centrosomal mechanism has been
mitotic spindles alters the division plane, leading to a premature proposed to account for the feature of PCC seen in the MCPH1
shift from symmetric to asymmetric divisions in the neuropro- depleted condition. It has been demonstrated that the N-terminal
genitor cells (Gruber et al., 2011). Thus, the increased asymmetric BRCT domain of MCPH1 interacts with the SET (SET nuclear onco-
divisions in neuroprogenitor cells led to the reduction in the num- gene) protein and MCPH1/ broblast cells depleted of SET by
ber of neurons and microcephaly. Also, these mice did not exhibit siRNA exhibited profound PCC (Leung et al., 2011). But, it is still not
tumor development over a span of 2 years (Gruber et al., 2011). known how the MCPH1-SET complex generates PCC. However, it
Additionally, Zhou et al. (2013) have examined whether the Mcph1 is known that SET prevents histone acetylation and regulates chro-
deletion would affect DNA damage response in neural cells of matin compaction (Miyamoto et al., 2003). Hence, MCPH1 could be
Mcph1-del mice. The absence of Mcph1 compromised HR in Mcph1- aiding chromatin compaction functions of SET protein.
del neuroprogenitors (Zhou et al., 2013). The authors discuss that
the inefcient HR could cause DNA damage accumulation in cells DNA repair functions of MCPH1
leading to genomic instability and nally apoptosis. Corroborating
this concept, increased apoptosis has been observed in Mcph1-del The new DNA repair functional roles of MCPH1 are reshaping
neuroprogenitors (Gruber et al., 2011; Zhou et al., 2013). Further- the current understanding of the DNA repair pathways. MCPH1
more, increased apoptosis may also contribute to neuronal loss in functions as a proximal factor for the DNA damage repair path-
addition to shift in asymmetric divisions during the development way and regulates the ATM (ataxia telangiectasia mutated) and ATR
of microcephaly. Hence, the studies on MCPH1 in model organisms (ataxia telangiectasia and Rad3 related) pathways (Rai et al., 2006).
have lead to a better understanding of its functional roles in the In response to DNA damage, MCPH1 co-localizes with numerous
development of MCPH. However, there are no studies that report proteins such as -H2AX (H2A histone family, member X), MDC1
Mcph1 overexpressing mouse models. There is a need for future (mediator of DNA-damage checkpoint 1), TP53BP1 (tumor protein
studies on MCPH1 transgenic mice to enhance our understanding p53 binding protein 1), NLRP2 (NLR family, pyrin domain contain-
of the protein function. ing 2), p-ATM, ATR and p-RAD17 [RAD17 homolog (S. pombe)] to
DNA damage repair foci in U2OS cells (Fig. 1) (Rai et al., 2006). H2AX
is the proximal signalling protein, and upon DNA damage, it gets
MCPH1 targets and functions phosphorylated at Y142 and S139 by WSTF (Williams-Beuren syn-
drome transcription factor), and ATM or ATR kinases, respectively.
Centrosomal functions of MCPH1 MCPH1 functions as a versatile sensor of H2AX phosphorylation
marks by recognizing both of these phosphorylation tags (Wood
MCPH1 localizes to centrosomes in U2OS and DT-40 cells, and et al., 2007; Shao et al., 2012; Singh et al., 2012a). Also, the C-
the N-terminal BRCT domain mediates its centrosome localization terminal BRCT domain of MCPH1 interacts with the N-terminal
(Zhong et al., 2006; Jeffers et al., 2008; Rai et al., 2008). MCPH1 region of BRCA2 (breast cancer 2, early onset) and positively regu-
depleted DT-40 cells displayed normal barrel structures of the cen- lates the localization of BRCA2 to DNA damage repair foci (Wu et al.,
trioles (Brown et al., 2010). However, MCPH1 deciency lead to 2009). Moreover, MCPH1 interacts with ATM/ATR phosphorylated
IR induced centrosome amplication in DT-40, U2OS and human SMARCC2 (SWI/SNF related, matrix associated, actin dependent
MCPH1 mutant lymphoblastoid cells (Alderton et al., 2006; Rai et al., regulator of chromatin, subfamily c, member 2), which is a sub-
2008; Brown et al., 2010). It is surprising to note that the centro- unit of the SWI-SNF complex upon DNA damage. This interaction
some amplication seen in DT-40 cells after exposure to IR was of MCPH1 and SWI/SNF complex leads to chromatin relaxation
T. Venkatesh, P.S. Suresh / European Journal of Cell Biology 93 (2014) 98105 101

Fig. 1. Functions of MCPH1. MCPH1 is represented by the central green box. The protein/complex interacting with MCPH1 is shown as boxes/other shapes in contact with
it. The transcriptional activation and inactivation are indicated by vertical red and green arrows respectively. The six major functions of MCPH1 such as apoptosis, DNA
repair, centrosomal integrity, telomere maintenance, chromatin relaxation and cell cycle checkpoints are represented in each purple box. (A) MCPH1 mediates apoptosis
by upregulation of pro-apoptotic genes in E2F1 dependent manner. (B) MCPH1 interacts with -H2AX and BRCA2 for the formation of ionizing radiation induced foci (IRIF)
indicated within the dashed boxes. (C) MCPH1 maintains the centrosome number and spindle alignment. (D) It interacts with the telosome and maintains telomere integrity.
It also transcriptionally represses the activity of hTERT. (E) MCPH1 interacts with SWI/SNF complex, relaxes the chromatin and increases the accessibility to DNA repair
proteins. (F) It regulates G1/S and G2/M cell cycle checkpoints by transcriptional activation of BRCA1 and CHEK1. The diagram is based on various studies cited appropriately in
the text.(For interpretation of the references to colour in this gure legend, the reader is referred to the web version of this article.)

(opening of the DNA) and allows the recruitment of DNA repair with MCPH1 using its [Y/F] XL domain and maintains the telomere
proteins to the sites of DNA damage (Peng et al., 2009). Further- integrity in HTC75 (human brosarcoma) cells (Kim et al., 2009).
rmore, U2OS cells depleted of MCPH1 by siRNAs are defective in
HR (homologous recombination) and NHEJ (non-homologous end
MCPH1 represses hTERT
joining) pathways (Peng et al., 2009). Two more studies have con-
rmed the role of MCPH1 in HR using DR-GFP/I-sceI system in
MCPH1 was initially identied as a repressor of hTERT in an
MCPH1/ MEFs and Mcph1-del ES cells (Wood et al., 2008; Zhou
ERM (enhanced retroviral mutagens) screen in HeLa cells (Lin
et al., 2013). Furthermore, MCPH1 interacts with Condensin II, a
and Elledge, 2003). In support of the hTERT repressing activity of
protein complex required for chromosome condensation and seg-
MCPH1, it has been shown that MCPH1 binds to the proximal region
regation. But surprisingly, it has been shown that the interaction of
of the hTERT promoter and represses its transcription in HeLa cells
MCPH1 with Condensin II mediates HR and does not have any chro-
(Shi and Su, 2012).
mosome related function (Wood et al., 2008). Though a number of
studies have demonstrated the role of MCPH1 in HR, there exists a
lack of follow up studies to prove the role of MCPH1 in NHEJ. MCPH1 mediates G1/S and G2/M checkpoints

MCPH1 positively regulates the expression of BRCA1 and CHEK1


MCPH1 maintains telomeres in U2OS cells (Xu et al., 2004; Lin et al., 2005; Passemard et al.,
2011)(Fig. 1). BRCA1 (breast cancer 1, early onset) and CHEK1 are
In mammalian cells, telomeres are protected by telosome com- required for IR-induced S phase and G2/M cell cycle checkpoints
plexes (Blasco, 2003). A telosome complex constitutes proteins (Xu et al., 2001; Yarden et al., 2002). However, transcriptional
such as TERF1 (telomere repeat factor-1), TERF2 (telomere repeat inactivation of BRCA1 and CHEK1 was not observed in human lym-
factor-2), POT1 (protection of telomere-1), RAP1 (human analogue phoblastoid cell lines (LBLs) with different truncating mutations
of yeast telomeric protein rag1), TIN2 (TERF1 interacting protein) in MCPH1 (MCPH1C74G and MCPH1427insA ) (Alderton et al., 2006).
and TPP1 (TINT1/PTOP/P1P1). Additionally, it is a platform for var- These cells exhibited normal levels of phosphorylated CHEK1 and
ious proteins/signalling pathways to interact with telomeres (Xin an active ATR signalling, suggesting that clinical features of MCPH
et al., 2008). Within the telosome complexes, TERF2 protein plays syndrome patient cells cannot be attributed to MCPH1-dependent
an important role as it recruits many proteins, such as TINF2 transcriptional regulation of CHEK1 or BRCA1 (Alderton et al., 2006).
(TERF1 [TRF1]-interacting nuclear factor 2), DCLRE1B (DNA cross- Knockdown of MCPH1 increased the radio-resistant DNA synthesis
link repair 1B) and PPP1R10 (protein phosphatase 1, regulatory (RDS) in U2OS cells (Xu et al., 2004). RDS is a phenomenon where
subunit 10), for the maintenance of the telomere ends (Kim et al., cells fail to inhibit DNA synthesis in response to IR (Rowley et al.,
2009). In addition to recruitment of these proteins, TERF2 interacts 1999). Moreover, depletion of MCPH1 lead to a higher content of
102 T. Venkatesh, P.S. Suresh / European Journal of Cell Biology 93 (2014) 98105

phospho-histone H3 (Ser 10), a mitotic marker in U2OS cells after of dysregulation of gene expressions of oncogenes and tumor
exposure to radiation (Xu et al., 2004). A higher staining of phospho- suppressors. The dysregulation of gene expressions are either
histone H3 (Ser 10) indicates the presence of defective G2/M arrest. a pre-transcriptional or post-transcriptional phenomenon. Pres-
Alderton et al. (2006) have studied the G2/M checkpoints in Lym- ence of gene amplications/deletions, somatic mutation and DNA
phoblastoid cell lines (LBLs) from control (WT), an ATM-defective hypo/hyper methylation are some of the mechanisms that result
ataxia telangiectasia (A-T), an ATR-Seckel and MCPH patients after in dysregulated gene expressions. Several DNA repair regulators
exposure to UV, and MCPH1 mutant cells were defective in G2/M have been associated with human cancer such as BRCA1 and BRCA2
checkpoint. Also, MCPH1 depleted broblast cells derived from (Domchek et al., 2010). Hence, this opens up a novel perspective
patients show a delayed release from G2/M cell cycle checkpoints that MCPH1 could have a functional role in cancer development or
following exposure to IR (Gavvovidis et al., 2010). Peripheral blood its progression. But, there are no reports of signs of cancer predis-
cells of chronic myeloid leukemia (CML) patients show downreg- position in the MCPH affected humans or knockout mice models. A
ulated MCPH1 expression and exhibit defective G2/M arrest upon hint about the functional role for MCPH1 in cancer is provided by the
exposure to hydroxyurea (HU) (Giallongo et al., 2011). Hence, the presence of 37 mutations in MCPH1 that has been recorded in COS-
effects of MCPH1 are different for IR and HU induced DNA damage MIC (Catalogue of somatic mutations in cancer) database (Forbes
responses. et al., 2011; http://www.sanger.ac.uk/cosmic). Over a decadeof its
discovery, several studies have addressed the genetic, epigenetic
MCPH1 mediates apoptosis and functional roles of MCPH1 in various cancers. These studies
conducted over the past years have lead to a new and growing con-
It is well established that E2F1 upregulates genes such as cept that the story of MCPH1 does not end at primary microcephaly
TP53 (tumor protein p53), TP73 (tumor protein p73), CASP3 (cas- and that it extends to the fascinating world of cancer too. Here, we
pase 3, apoptosis-related cysteine peptidase), CASP7 (caspase 7, summarise the studies conducted to elucidate the role of MCPH1
apoptosis-related cysteine peptidase) and APAF1 (apoptotic pepti- in various human cancer.
dase activating factor 1) and hence, mediates apoptosis (Ginsberg,
2002). Knockdown of MCPH1 in HEK293 cells, results in decreased
expression of E2F1 target genes such as TP53, CASP3, CASP7, BRCA1
MCPH1 in breast cancer
and CHEK1. Moreover, the second and the third BRCT domains
of MCPH1 interact with the N-terminal of E2F1 (Yang et al.,
MCPH1 contains three BRCT domains, and it is well known that
2008). These observations indicate that MCPH1 is involved in E2F1
breast cancer is inuenced by BRCT containing proteins like BRCA1
mediated apoptosis. Also, the ChIP and ChIP-re-ChIP assays have
and BRCA2 (di Masi et al., 2011). Decreased copy number of MCPH1
conrmed the presence of E2F1-MCPH1 complex on the promoters
has been reported in 38/54 breast cancer cell lines by CGH (com-
of CHEK1, BRCA1, TP73 and CASP7 in U2OS and HEK293 cells (Yang
parative genomic hybridization) (Rai et al., 2006). MCPH1 is down
et al., 2008). Interestingly, the E2F1 mediated regulatory effects of
regulated in 70% (7/10) of breast cancer tissues by IHC (Rai et al.,
human MCPH1 and rhesus macaque MCPH1 are different for p73,
2008). Also, another IHC study has reported down regulation of
Cyclin E and p14Arf (Shi et al., 2013). This indicates a functional
MCPH1 in 93/319 (29%) breast cancer samples (Richardson et al.,
divergence of MCPH1 between human and non-human primates.
2011) (Table 1). Reduced expression of MCPH1 has been demon-
strated in high grade, BRCA1 negative tumors and is a prognostic
Effects of MCPH1 on mitotic phenotypes indicator in breast cancer (Richardson et al., 2011). Also, presence
of T allele in the SNPs of MCPH1 (rs2912010 and rs1057090) is
The functions of MCPH1 have been an evolving area and a associated with increasing tumor grade in breast cancer (Jo et al.,
new role for MCPH1 in the regulation of mitosis has been exam- 2013). A 38 bp deletion in exon 10 of MCPH1 has been identied in
ined (Singh et al., 2012b). Singh et al. (2012b) have shown that a breast cancer sample (Rai et al., 2006). Bhattacharya et al. (2013)
CDC27 interacts with MCPH1 using crystallization and ITC (isother- reported that reduced expression and alteration of ATM and MCPH1
mal titration calorimetry) based studies. CDC27 is a component of were seen in 96% (121/ 126) of breast cancer samples and also,
anaphase promoting complex (APC). APC is very essential for the patients with alterations in MCPH1 show poor outcome after treat-
regulation of mitosis and it is interesting to note that the knock- ment with radiation or DNA interacting drugs (Bhattacharya et al.,
down of CDC27 or MCPH1 exhibit similar mitotic phenotypes such 2013). Knockdown of MCPH1 lead to increased cellular prolifera-
as presence of smaller nuclei, chromosomal segregation defects and tion and growth in soft agar in MCF10A (breast cancer) cells (Zhang
PCC (Singh et al., 2012b). et al., 2013). In corroboration with these experiments, the ectopic
expression of MCPH1 in MCF7 (breast cancer) cells suppressed cell
Insights into the functions of MCPH1 in cancer development proliferation and colony formation in vitro and tumor growth in
nude mice (Zhang et al., 2013). The mechanism of tumor suppres-
Cancer is a multifactorial disease caused by both genetic and sion of MCPH1 is attributed to the increased stabilisation of the
non-genetic factors. It is a major public concern and continues master tumor suppressor TP53 (tumor protein p53) in MCF10A
to be the second common cause of death following heart dis- cells (Zhang et al., 2013). MCPH1 induces auto-ubiquitination of
eases in America (Valdivieso et al., 2012). Cancer is a phenomenon MDM2 (murine double minute 2), the negative regulator of TP53

Table 1
Summary of studies on MCPH1 in human cancer.

Status of MCPH1 Type of tumor References

M, D, Me, LOH Breast Rai et al., 2006; Richardson et al., 2011; Bhattacharya et al., 2012.
M Endometrial Bilbao et al., 2010
D Primary ovarian cancer cultures, ovarian Rai et al., 2006; Brning-Richardson et al., 2011
D Glioblastoma Hagemann et al., 2008
D, M, Me, LOH, miR Oral squamous cell carcinoma Venkatesh et al., 2013

Abbreviation: M, mutated; D, down regulated; Me, methylated; LOH; loss of heterozygosity; miR, miRNA regulated.
T. Venkatesh, P.S. Suresh / European Journal of Cell Biology 93 (2014) 98105 103

which eventually leads to the proteosomal degradation of MDM2 Implication of MCPH1 in Otitis media (OM)
and thus, stabilizes TP53 (Zhang et al., 2013).
Recently, a novel role for MCPH1 in the development of otitis
MCPH1 in endometrial cancer media has been identied. Otitis media is a multifactorial disease
characterized by the inammation of the middle ear. Environmen-
Endometrial cancer is the most common malignancy of the tal factors such as smoking and type of child care, anatomical
female genital tract (Siegel et al., 2012). It is estrogen dependent and dysmorphology, Eustachian-tube function, viral and bacterial load,
patients with advanced stage cancers have a much less favorable and, genetic predisposition are some of the factors responsible
prognosis (Karaayvaz et al., 2012). Mononucleotide deletions in the for the development and persistence of OM (Chen et al., 2013).
A tracts of exons 4 (2/41 samples), 5 (1/41 samples) and 8 (2/41 Chen et al. (2013) generated a Mcph1 decient mice and examined
samples) of the MCPH1 gene have been reported in microsatellite the hearing ability in these mice using ABR (auditory brainstem
instability (MSI) positive endometrial cancer tissues, accounting to response) analysis. Homozygous mutant mice exhibited elevated
a total mutation rate of 12% (5/41) (Bilbao et al., 2010). All these ABR thresholds as early as 3 weeks of age. The heterozygous mice
mutations were monoallelic and 62 microsatellite stable (MSS) showed no hearing impairment in consistent with the recessive
endometrial cancer tissues were normal for MCPH1 (Bilbao et al., model of inheritance for patients with MCPH1 mutations (Chen
2010). et al., 2013). Furthermore, anatomical and histological studies in
these Mcph1 decient mice revealed OM. Several defects in the mid-
dle ear such as thickened and white bone forming the bulla instead
MCPH1 in ovarian cancer
of the normal thin and transparent bone, retracted tympanic mem-
brane, bubbles present underneath the tympanic membrane, or
Ovarian cancer is the fth-leading cause of cancer-related
middle ear cavities lled with clear or cloudy uid were detected
death in the United States, and is the most lethal of all gyne-
(Chen et al., 2013). Chen et al. (2013) performed immunohisto-
cological malignancies (Zhan et al., 2013). Rai et al. (2006) have
chemical studies and revealed that Mcph1 is expressed in the
shown decreased MCPH1 genomic DNA copy number in 35/87
middle ear of the wildtype mice. Still, question remains regarding
(40.22%) advanced epithelial ovarian cancers using CGH (Compar-
the MCPH1 signalling pathways that lead to OM.
ative genomic hybridization). Also, decreased MCPH1 level was
seen in 19/30 ovarian cancer specimens in comparison to seven
benign ovarian specimens (Rai et al., 2006)(Table 1). Additionally, Conclusions
ICC (immunocytochemistry) of 36 primary cultures from ovarian
cancer samples showed a strong/moderate staining of MCPH1 in Studies so far have proved that MCPH1 has roles in the develop-
the nucleus and out of these, a weak cytoplasmic staining was seen ment of brain, cancer and OM. Though several interacting proteins
in 25% of the samples (Brning-Richardson et al., 2011). and pathways have been elucidated for MCPH1, there are sev-
eral interesting aspects of MCPH1 which are yet to be unravelled.
MCPH1 in glioblastoma They include elucidation of a more direct centrosomal function for
MCPH1 or to identify whether MCPH1 interacts with any other
Glioblastoma multiforme (GBM) is the most prevalent malig- MCPH proteins. Our growing familiarity with MCPH proteins has
nant brain tumor in adults. These tumors may occur as de novo proved their signalling to be highly complex in the pathogenesis of
or progress from a low grade astrocytoma (LGA, WHO grade II) MCPH. However, a possibility of the existence of a master regula-
or anaplastic astrocytoma (WHO grade III) (Biernat et al., 1997). tor for all these MCPH proteins cannot be ruled out. Also, the area
Hagemann et al. (2008) analysed the transcript level of MCPH1 in less visited is the post translation modication of MCPH1, and how
15 LGA and 15 GBM tumors. They reported that all the normal brain these could inuence the functions of MCPH1 in primary micro-
samples expressed MCPH1, but all at low levels. MCPH1 expression cephaly, cancer and OM. Wide scale systems biology, knock out
was reduced to 20% and 27% in LGA and GBM tumors, respec- studies and networking based approaches will enhance our knowl-
tively, when compared to the normal brain tissues (Hagemann et al., edge of using MCPH1 as a therapeutic target for cancer. As, MCPH1
2008). is a negative regulator of MDM2 in breast cancer, molecular effec-
tors that can up regulate MCPH1 can be an attractive anticancer
therapeutic approach.
MCPH1 in oral squamous cell carcinoma

Oral squamous cell carcinoma (OSCC) is a subgroup of HNSCC Conict of interest


(Supic et al., 2009). It accounts for 24% of all malignancies in the
west. However, in Indian subcontinent, OSCC accounts to 4050% The authors declare no conict of interest.
of all malignancies (Mehrotra et al., 2006). Microsatellite analy-
sis has shown LOH (loss of heterozygosity) at the D8S518 and Acknowledgement
D8S277 markers anking the MCPH1 locus in 1/21 oral tumors
(Wu et al., 1997). Venkatesh et al. (2013) have shown LOH We would like to thank Professor Arun Kumar, Indian Institute
of MCPH1 in 14/71 (19.72%) informative samples. Also, a pro- of Science, Bangalore for his help and support and also the study
tein truncating mutation c.1561G.T(p.Glu521X) was identied in institutes. We thank Kavita Bommasamudram, Stony Brook Uni-
1/15 OSCC samples (Venkatesh et al., 2013). Furthermore, their versity, New York for critical reading of the manuscript. We also
study has reported that MCPH1 was down regulated at both thank the reviewers for their valuable suggestions to improve the
the transcript and protein levels in 21/41 (51.22%) and 19/25 manuscript.
(76%) OSCC samples, respectively (Table 1). Overexpression of
MCPH1 in KB cells decreased cellular proliferation, anchorage-
independent growth in soft agar, cell invasion and tumor size in References
nude mice, indicating its tumor suppressive function. Additionally,
Alderton, G.K., Galbiati, L., Grifth, E., Surinya, K.H., Neitzel, H., Jackson, A.P., Jeggo,
it is intriguing to note that MCPH1 is regulated by miR-27a in OSCC P.A., ODriscoll, M., 2006. Regulation of mitotic entry by microcephalin and its
(Venkatesh et al., 2013). overlap with ATR signalling. Nat. Cell Biol. 8, 725733.
104 T. Venkatesh, P.S. Suresh / European Journal of Cell Biology 93 (2014) 98105

Awad, S., Al-Dosari, M.S., Al-Yacoub, N., Colak, D., Salih, M.A., Alkuraya, F.S., Poizat, Ginsberg, D., 2002. E2F1 pathways to apoptosis. FEBS Lett. 529, 122125.
C., 2013. Mutation in PHC1 implicates chromatin remodeling in primary micro- Glover, J.N., Williams, R.S., Lee, M.S., 2004. Interactions between BRCT repeats and
cephaly pathogenesis. Hum. Mol. Genet. 22, 22002213. phosphoproteins: tangled up in two. Trends Biochem. Sci. 29, 579585.
Bailly, E., Pines, J., Hunter, T., Bornens, M., 1992. Cytoplasmic accumulation of cyclin Gruber, R., Zhou, Z., Sukchev, M., Joerss, T., Frappart, P.O., Wang, Z.Q., 2011. MCPH1
B1 in human cells: association with a detergent-resistant compartment and with regulates the neuroprogenitor division mode by coupling the centrosomal
the centrosome. J. Cell Sci. 101, 529545. cycle with mitotic entry through the Chk1-Cdc25 pathway. Nat. Cell Biol. 13,
Bhattacharya, N., Mukherjee, N., Singh, R.K., Sinha, S., Alam, N., Roy, A., Roy- 13251334.
choudhury, S., Panda, C.K., 2012. Frequent Alterations of MCPH1 and ATM are Guernsey, D.L., Jiang, H., Hussin, J., Arnold, M., Bouyakdan, K., Perry, S., Babineau-
associated with primary breast carcinoma: clinical and prognostic implications. Sturk, T., Beis, J., Dumas, N., Evans, S.C., Ferguson, M., Matsuoka, M., Macgillivray,
Ann. Surg. Oncol. (Epub ahead of print). C., Nightingale, M., Patry, L., Rideout, A.L., Thomas, A., Orr, A., Hoffmann, I.,
Biernat, W., Tohma, Y., Yonekawa, Y., Kleihues, P., Ohgaki, H., 1997. Alterations of Michaud, J.L., Awadalla, P., Meek, D.C., Ludman, M., Samuels, M.E., 2010. Muta-
cell cycle regulatory genes in primary (de novo) and secondary glioblastomas. tions in centrosomal protein CEP152 in primary microcephaly families linked to
Acta Neuropathol. 94, 303309. MCPH4. Am. J. Hum. Genet. 87, 4051.
Bilbao, C., Ramrez, R., Rodrguez, G., Falcn, O., Len, L., Daz-Chico, N., Perucho, Hagemann, C., Anacker, J., Gerngras, S., Khnel, S., Said, H.M., Patel, R., Kmmerer, U.,
M., Daz-Chico, J.C., 2010. Double strand break repair components are frequent Vordermark, D., Roosen, K., Vince, G.H., 2008. Expression analysis of the autoso-
targets of microsatellite instability in endometrial cancer. Eur. J. Cancer 46, mal recessive primary microcephaly genes MCPH1 (microcephalin) and MCPH5
28212827. (ASPM, abnormal spindle-like, microcephaly associated) in human malignant
Blasco, M.A., 2003. Mammalian telomeres and telomerase: why they matter for gliomas. Oncol. Rep. 20, 301308.
cancer and aging. Eur. J. Cell Biol. 82, 441446. Hussain, S.M., Marriam Bakhtiar, S., Farooq, M., Anjum, I., Janzen, E., Reza Toliat, M.,
Bond, J., Roberts, E., Mochida, G.H., Hampshire, D.J., Scott, S., Askham, J.M., Springell, Eiberg, H., Kjaer, K., Tommerup, N., Noegel, A., Nrnberg, P., Baig, S., Hansen, L.,
K., Mahadevan, M., Crow, Y.J., Markham, A.F., Walsh, C.A., Woods, C.G., 2002. 2013. Genetic heterogeneity in Pakistani microcephaly families. Clin. Genet. 83,
ASPM is a major determinant of cerebral cortical size. Nat. Genet. 32, 316320. 446451.
Bond, J., Roberts, E., Springell, K., Lizarraga, S.B., Scott, S., Higgins, J., Hampshire, D.J., Huyton, T., Bates, P.A., Zhang, X., Sternberg, M.J., Freemont, P.S., 2000. The BRCA1
Morrison, E.E., 2005. A centrosomal mechanism involving CDK5RAP2 and CENPJ C-terminal domain: structure and function. Mutat. Res. 460, 319332.
controls brain size. Nat. Genet. 37, 353355. Jackman, M., Lindon, C., Nigg, E.A., Pines, J., 2003. Active cyclin B1-Cdk1 rst appears
Brown, J.A., Bourke, E., Liptrot, C., Dockery, P., Morrison, C.G., 2010. MCPH1/BRIT1 on centrosomes in prophase. Nat. Cell Biol. 5, 143148.
limits ionizing radiation-induced centrosome amplication. Oncogene 29, Jackson, A.P., Eastwood, H., Bell, S.M., Adu, J., Toomes, C., Carr, I.M., Roberts, E., Hamp-
55375544. shire, D.J., Crow, Y.J., Mighell, A.J., Karbani, G., Jafri, H., Rashid, Y., Mueller, R.F.,
Brning-Richardson, A., Bond, J., Alsiary, R., Richardson, J., Cairns, D.A., McCormack, Markham, A.F., Woods, C.G., 2002. Identication of microcephalin, a protein
L., Hutson, R., Burns, P., Wilkinson, N., Hall, G.D., Morrison, E.E., Bell, S.M., 2011. implicated in determining the size of the human brain. Am. J. Hum. Genet. 71,
ASPM and microcephalin expression in epithelial ovarian cancer correlates with 136142.
tumour grade and survival. Br. J. Cancer 104, 16021610. Jackson, A.P., McHale, D.P., Campbell, D.A., Jafri, H., Rashid, Y., Mannan, J., Karbani,
Brunk, K., Vernay, B., Grifth, E., Reynolds, N.L., Strutt, D., Ingham, P.W., Jackson, A.P., G., Corry, P., Levene, M.I., Mueller, R.F., Markham, A.F., Lench, N.J., Woods, C.G.,
2007. Microcephalin coordinates mitosis in the syncytial Drosophila embryo. J. 1998. Primary autosomal recessive microcephaly (MCPH1) maps to chromo-
Cell Sci. 120, 33883578. some 8p22-pter. Am. J. Hum. Genet. 63, 541546.
Chen, J., Ingham, N., Clare, S., Raisen, C., Vancollie, V.E., Ismail, O., McIntyre, R.E., Jeffers, L.J., Coull, B.J., Stack, S.J., Morrison, C.G., 2008. Distinct BRCT domains in
Tsang, S.H., Mahajan, V.B., Dougan, G., Adams, D.J., White, J.K., Steel, K.P., 2013. Mcph1/Brit1 mediate ionizing radiation-induced focus formation and centro-
Mcph1-decient mice reveal a role for MCPH1 in otitis media. PLoS One 8, somal localization. Oncogene 27, 139144.
e58156. Jo, Y.H., Kim, H.O., Lee, J., Lee, S.S., Cho, C.H., Kang, I.S., Choe, W.J., Baik, H.H., Yoon,
Darvish, H., Esmaeeli-Nieh, S., Monajemi, G.B., Mohseni, M., Ghasemi-Firouzabadi, K.S., 2013. MCPH1 protein expression and polymorphisms are associated with
S., Abedini, S.S., Bahman, I., Jamali, P., Azimi, S., Mojahedi, F., Dehghan, A., risk of breast cancer. Gene 517, 184190.
Shafeghati, Y., Jankhah, A., Falah, M., Soltani Banavandi, M.J., Ghani-Kakhi, M., Kaindl, A.M., Passemard, S., Kumar, P., Kraemer, N., Issa, L., Zwirner, A., Gerard, B.,
Garshasbi, M., Rakhshani, F., Naghavi, A., Tzschach, A., Neitzel, H., Ropers, H.H., Verloes, A., Mani, S., Gressens, P., 2010. Many roads lead to primary autosomal
Kuss, A.W., Behjati, F., Kahrizi, K., Najmabadi, H., 2010. A clinical and molecular recessive microcephaly. Prog. Neurobiol. 90, 363383.
genetic study of 112 Iranian families with primary microcephaly. J. Med. Genet. Karaayvaz, M., Zhang, C., Liang, S., Shroyer, K.R., Ju, J., 2012. Prognostic signicance
47, 823828. of miR-205 in endometrial cancer. PLoS One 7, e35158.
di Masi, A., Gullotta, F., Cappadonna, V., Leboffe, L., Ascenzi, P., 2011. Cancer predis- Kim, H., Lee, O.H., Xin, H., Chen, L.Y., Qin, J., Chae, H.K., Lin, S.Y., Safari, A., Liu, D.,
posing mutations in BRCT domains. IUBMB Life 63, 503512. Songyang, Z., 2009. TRF2 functions as a protein hub and regulates telomere
Domchek, S.M., Friebel, T.M., Singer, C.F., Evans, D.G., Lynch, H.T., Isaacs, C., Garber, maintenance by recognizing specic peptide motifs. Nat. Struct. Mol. Biol. 16,
J.E., Neuhausen, S.L., Matloff, E., Eeles, R., Pichert, G., Van tveer, L., Tung, N., 372379.
Weitzel, J.N., Couch, F.J., Rubinstein, W.S., Ganz, P.A., Daly, M.B., Olopade, O.I., Kumar, A., Girimaji, S.C., Duvvari, M.R., Blanton, S.H., 2009. Mutations in STIL, encod-
Tomlinson, G., Schildkraut, J., Blum, J.L., Rebbeck, T.R., 2010. Association of risk- ing a pericentriolar and centrosomal protein, cause primary microcephaly. Am.
reducing surgery in BRCA1 or BRCA2 mutation carriers with cancer risk and J. Hum. Genet. 84, 286290.
mortality. JAMA 304, 967975. Kumar, A., Markandaya, M., Girimaji, S.C., 2002. Primary microcephaly: micro-
Farooq, M., Baig, S., Tommerup, N., Kjaer, K.W., 2010. Craniosynostosis-microcephaly cephalin and ASPM determine the size of the human brain. J. Biosci. 27,
with chromosomal breakage and other abnormalities is caused by a truncating 629632.
MCPH1 mutation and is allelic to premature chromosomal condensation syn- Leung, J.W., Leitch, A., Wood, J.L., Shaw-Smith, C., Metcalfe, K., Bicknell, L.S., Jackson,
drome and primary autosomal recessive microcephaly type 1. Am. J. Med. Genet. A.P., Chen, J., 2011. SET nuclear oncogene associates with microcephalin/MCPH1
A 152A, 495497. and regulates chromosome condensation. J. Biol. Chem. 286, 2139321400.
Forbes, S.A., Bindal, N., Bamford, S., Cole, C., Kok, C.Y., Beare, D., Jia, M., Shepherd, R., Liang, Y., Gao, H., Lin, S.Y., Peng, G., Huang, X., Zhang, P., Goss, J.A., Brunicardi, F.C.,
Leung, K., Menzies, A., Teague, J.W., Campbell, P.J., Stratton, M.R., Futreal, P.A., Multani, A.S., Chang, S., Li, K., 2010. BRIT1/MCPH1 is essential for mitotic and
2011. COSMIC: mining complete cancer genomes in the catalogue of somatic meiotic recombination DNA repair and maintaining genomic stability in mice.
mutations in cancer. Nucleic Acids Res. 39, D945D950. PLoS Genet. 6, e1000826.
Garshasbi, M., Motazacker, M.M., Kahrizi, K., Behjati, F., Abedini, S.S., Nieh, S.E., Lin, S.Y., Elledge, S.J., 2003. Multiple tumor suppressor pathways negatively regulate
Firouzabadi, S.G., Becker, C., Rschendorf, F., Nrnberg, P., Tzschach, A., Vaz- telomerase. Cell 113, 881889.
ifehmand, R., Erdogan, F., Ullmann, R., Lenzner, S., Kuss, A.W., Ropers, H.H., Lin, S.Y., Liang, Y., Li, K., 2010. Multiple roles of BRIT1/MCPH1 in DNA damage
Najmabadi, H., 2006. SNP array-based homozygosity mapping reveals MCPH1 response, DNA repair, and cancer suppression. Yonsei Med. J. 51, 295301.
deletion in family with autosomal recessive mental retardation and mild micro- Lin, S.Y., Rai, R., Li, K., Xu, Z.X., Elledge, S.J., 2005. BRIT1/MCPH1 is a DNA dam-
cephaly. Hum. Genet. 118, 708715. age responsive protein that regulates the Brca1-Chk1 pathway, implicating
Gavvovidis, I., Phlmann, C., Marchal, J.A., Stumm, M., Yamashita, D., Hirano, T., checkpoint dysfunction in microcephaly. Proc. Natl. Acad. Sci. U. S. A. 102,
Schindler, D., Neitzel, H., Trimborn, M., 2010. MCPH1 patient cells exhibit 1510515109.
delayed release from DNA damage-induced G2/M checkpoint arrest. Cell Cycle Mehrotra, R., Gupta, A., Singh, M., Ibrahim, R., 2006. Application of cytology and
9, 48934899. molecular biology in diagnosing premalignant or malignant oral lesions. Mol.
Gavvovidis, I., Rost, I., Trimborn, M., Kaiser, F.J., Purps, J., Wiek, C., Hanenberg, H., Cancer 5, 11.
Neitzel, H., Schindler, D., 2012. A novel MCPH1 isoform complements the defec- Miyamoto, S., Suzuki, T., Muto, S., Aizawa, K., Kimura, A., Mizuno, Y., Nagino, T., Imai,
tive chromosome condensation of human MCPH1-decient cells. PLoS One 7, Y., Adachi, N., Horikoshi, M., Nagai, R., 2003. Positive and negative regulation of
e40387. the cardiovascular transcription factor KLF5 by p300 and the oncogenic regulator
Ghani-Kakhki, M., Robinson, P.N., Morlot, S., Mitter, D., Trimborn, M., Albrecht, B., SET through interaction and acetylation on the DNA-binding domain. Mol. Cell
Varon, R., Sperling, K., Neitzel, H., 2012. Two missense mutations in the pri- Biol. 23, 85288541.
mary autosomal recessive microcephaly gene MCPH1 disrupt the function of the Neitzel, H., Neumann, L.M., Schindler, D., Wirges, A., Tnnies, H., Trimborn, M., Kreb-
highly conserved N-terminal BRCT domain of microcephalin. Mol. Syndromol. sova, A., Richter, R., Sperling, K., 2002. Premature chromosome condensation in
3, 613. humans associated with microcephaly and mental retardation: a novel autoso-
Giallongo, C., Tibullo, D., La Cava, P., Branca, A., Parrinello, N., Spina, P., Stagno, F., mal recessive condition. Am. J. Hum. Genet. 70, 10151022.
Conticello, C., Chiarenza, A., Vigneri, P., Palumbo, G.A., Di Raimondo, F., 2011. Nicholas, A.K., Khurshid, M., Dsir, J., Carvalho, O.P., Cox, J.J., Thornton, G., Kausar, R.,
BRIT1/MCPH1 expression in chronic myeloid leukemia and its regulation of the Ansar, M., Ahmad, W., Verloes, A., Passemard, S., Misson, J.P., Lindsay, S., Gergely,
G2/M checkpoint. Acta Haematol. 126, 205210. F., Dobyns, W.B., Roberts, E., Abramowicz, M., Woods, C.G., 2010. WDR62 is
T. Venkatesh, P.S. Suresh / European Journal of Cell Biology 93 (2014) 98105 105

associated with the spindle pole and is mutated in human microcephaly. Nat. Trimborn, M., Bell, S.M., Felix, C., Rashid, Y., Jafri, H., Grifths, P.D., Neumann, L.M.,
Genet. 42, 10101014. Krebs, A., Reis, A., Sperling, K., Neitzel, H., Jackson, A.P., 2004. Mutations in micro-
Nurse, P., 1990. Universal control mechanism regulating onset of M-phase. Nature cephalin cause aberrant regulation of chromosome condensation. Am. J. Hum.
344, 503508. Genet. 75, 261266.
Passemard, S., El Ghouzzi, V., Nasser, H., Verney, C., Vodjdani, G., Lacaud, A., Lebon, Trimborn, M., Ghani, M., Walther, D.J., Dopatka, M., Dutrannoy, V., Busche, A., Meyer,
S., Laburthe, M., Robberecht, P., Nardelli, J., Mani, S., Verloes, A., Gressens, P., F., Nowak, S., Zabel, C., Klose, J., Esquitino, V., Garshasbi, M., Kuss, A.W., Ropers,
Lelivre, V., 2011. VIP blockade leads to microcephaly in mice via disruption of H.H., Mueller, S., Poehlmann, C., Gavvovidis, I., Schindler, D., Sperling, K., Neitzel,
Mcph1-Chk1 signaling. J. Clin. Invest. 121, 30713087. H., 2010. Establishment of a mouse model with misregulated chromosome con-
Passemard, S., Titomanlio, L., Elmaleh, M., Afenjar, A., Alessandri, J.L., Andria, G., de densation due to defective Mcph1 function. PLoS One 5, e9242.
Villemeur, T.B., Boespug-Tanguy, O., Burglen, L., Del Giudice, E., Guimiot, F., Trimborn, M., Richter, R., Sternberg, N., Gavvovidis, I., Schindler, D., Jackson, A.P.,
Hyon, C., Isidor, B., Mgarban, A., Moog, U., Odent, S., Hernandez, K., Pouvreau, Prott, E.C., Sperling, K., Gillessen-Kaesbach, G., Neitzel, H., 2005. The rst mis-
N., Scala, I., Schaer, M., Gressens, P., Gerard, B., Verloes, A., 2009. Expanding the sense alteration in the MCPH1 gene causes autosomal recessive microcephaly
clinical and neuroradiologic phenotype of primary microcephaly due to ASPM with an extremely mild cellular and clinical phenotype. Hum. Mutat. 26, 496.
mutations. Neurology 73, 962969. Valdivieso, M., Kujawa, A.M., Jones, T., Baker, L.H., 2012. Cancer survivors in the
Peng, G., Yim, E.K., Dai, H., Jackson, A.P., Burgt, I., Pan, M.R., Hu, R., Li, K., Lin, S.Y., 2009. United States: a review of the literature and a call to action. Int. J. Med. Sci. 9,
BRIT1/MCPH1 links chromatin remodelling to DNA damage response. Nat. Cell 163173.
Biol. 11, 865872. Venkatesh, T., Nagashri, M.N., Swamy, S.S., Mohiyuddin, S.M., Gopinath, K.S., Kumar,
Rai, R., Dai, H., Multani, A.S., Li, K., Chin, K., Gray, J., Lahad, J.P., Liang, J., Mills, G.B., A., 2013. Primary microcephaly gene MCPH1 shows signatures of tumor sup-
Meric-Bernstam, F., Lin, S.Y., 2006. BRIT1 regulates early DNA damage response, pressors and is regulated by miR-27a in oral squamous cell carcinoma. PLoS
chromosomal integrity, and cancer. Cancer Cell 10, 145157. One 8, e54643.
Rai, R., Phadnis, A., Haralkar, S., Badwe, R.A., Dai, H., Li, K., Lin, S.Y., 2008. Differential Wang, Y., Dantas, T.J., Lalor, P., Dockery, P., Morrison, C.G., 2013. Promoter hijack
regulation of centrosome integrity by DNA damage response proteins. Cell Cycle reveals pericentrin functions in mitosis and the DNA damage response. Cell Cycle
7, 22252233. 12, 635646.
Ramos, E., Bien-Willner, G., Li, J., Hughes, A., Giacalone, J., Chasnoff, S., Kulkarni, S., Wood, J.L., Liang, Y., Li, K., Chen, J., 2008. Microcephalin/MCPH1 associates with the
Parmacek, M., Cole, F., Druley, T., 2013. Genetic variation in MKL2 and decreased condensin II complex to function in homologous recombination repair. J. Biol.
downstream PCTAIRE1 expression in extreme, fatal primary human micro- Chem. 283, 2958629592.
cephaly. Clin. Genet. (Epub ahead of print). Wood, J.L., Singh, N., Mer, G., Chen, J., 2007. MCPH1 functions in an H2AX-dependent
Richards, M.W., Leung, J.W., Roe, S.M., Li, K., Chen, J., Bayliss, R.A., 2010. A pocket but MDC1-independent pathway in response to DNA damage. J. Biol. Chem. 282,
on the surface of the N-terminal BRCT domain of Mcph1 is required to prevent 3541635423.
abnormal chromosome condensation. J. Mol. Biol. 395, 908915. Wu, C.L., Roz, L., Sloan, P., Read, A.P., Holland, S., Porter, S., Scully, C., Speight,
Richardson, J., Shaaban, A.M., Kamal, M., Alisary, R., Walker, C., Ellis, I.O., Speirs, V., P.M., Thakker, N., 1997. Deletion mapping denes three discrete areas of allelic
Green, A.R., Bell, S.M., 2011. Microcephalin is a new novel prognostic indicator imbalance on chromosome arm 8p in oral and oropharyngeal squamous cell
in breast cancer associated with BRCA1 inactivation. Breast Cancer Res. Treat. carcinomas. Genes Chromosomes Cancer 20, 347353.
127, 639648. Wu, X., Mondal, G., Wang, X., Wu, J., Yang, L., Pankratz, V.S., Rowley, M., Couch, F.J.,
Rickmyre, J.L., Dasgupta, S., Ooi, D.L., Keel, J., Lee, E., Kirschner, M.W., Waddell, S., 2009. Microcephalin regulates BRCA2 and Rad51-associated DNA double-strand
Lee, L.A., 2007. The Drosophila homolog of MCPH1, a human microcephaly gene, break repair. Cancer Res. 69, 55315536.
is required for genomic stability in the early embryo. J. Cell Sci. 120, 35653577. Xin, H., Liu, D., Songyang, Z., 2008. The telosome/shelterin complex and its functions.
Rowley, R., Phillips, E.N., Schroeder, A.L., 1999. The effects of ionizing radiation on Genome Biol. 9, 232.
DNA synthesis in eukaryotic cells. Int. J. Radiat. Biol. 75, 267283. Xu, B., Kim, S., Kastan, M.B., 2001. Involvement of Brca1 in S-phase and
Shao, Z., Li, F., Sy, S.M., Yan, W., Zhang, Z., Gong, D., Wen, B., Huen, M.S., Gong, Q., G(2)-phase checkpoints after ionizing irradiation. Mol. Cell Biol. 21,
Wu, J., Shi, Y., 2012. Specic recognition of phosphorylated tail of H2AX by the 34453450.
tandem BRCT domains of MCPH1 revealed by complex structure. J. Struct. Biol. Xu, X., Lee, J., Stern, D.F., 2004. Microcephalin is a DNA damage response
177, 459468. protein involved in regulation of CHK1 and BRCA1. J. Biol. Chem. 279,
Shi, L., Su, B., 2012. Identication and functional characterization of a primate- 3409134094.
specic E2F1 binding motif regulating MCPH1 expression. FEBS J. 279, 491503. Yamashita, D., Shintomi, K., Ono, T., Gavvovidis, I., Schindler, D., Neitzel, H., Trimborn,
Shi, L., Li, M., Lin, Q., Qi, X., Su, B., 2013. Functional divergence of the brain-size M., Hirano, T., 2011. MCPH1 regulates chromosome condensation and shaping
regulating gene MCPH1 during primate evolution and the origin of humans. as a composite modulator of condensin II. J. Cell Biol. 194, 841854.
BMC Biol. 11, 62. Yang, S.Z., Lin, F.T., Lin, W.C., 2008. MCPH1/BRIT1 cooperates with E2F1 in
Siegel, R., Naishadham, D., Jemal, A., 2012. Cancer statistics. CA. Cancer J. Clin. 62, the activation of checkpoint, DNA repair and apoptosis. EMBO Rep. 9,
1029. 907915.
Singh, N., Basnet, H., Wiltshire, T.D., Mohammad, D.H., Thompson, J.R., Hroux, A., Yang, Y.J., Baltus, A.E., Mathew, R.S., Murphy, E.A., Evrony, G.D., Gonzalez, D.M.,
Botuyan, M.V., Yaffe, M.B., Couch, F.J., Rosenfeld, M.G., Mer, G., 2012a. Dual Wang, E.P., Walker, Marshall-, Barry, C.A., Murn, B.J., Tatarakis, J., Mahajan, A.,
recognition of phosphoserine and phosphotyrosine in histone variant H2A. X Samuels, M.A., Shi, H.H., Golden, Y., Mahajnah, J.A., Shenhav, M., Walsh, R., C.A,
by DNA damage response protein MCPH1. Proc. Natl. Acad. Sci. U. S. A. 109, 2012. Microcephaly gene links trithorax and REST/NRSF to control neural stem
1438114386. cell proliferation and differentiation. Cell 151, 10971112.
Singh, N., Wiltshire, T.D., Thompson, J.R., Mer, G., Couch, F.J., 2012b. Molecular basis Yarden, R.I., Pardo-Reoyo, S., Sgagias, M., Cowan, K.H., Brody, L.C., 2002. BRCA1 reg-
for the association of microcephalin (MCPH1) protein with the cell division cycle ulates the G2/M checkpoint by activating Chk1 kinase upon DNA damage. Nat.
protein 27 (Cdc27) subunit of the anaphase-promoting complex. J. Biol. Chem. Genet. 30, 259285.
287, 28542862. Zhan, Q., Wang, C., Ngai, S., 2013. Ovarian cancer stem cells: a new target for cancer
Sir, J.H., Barr, A.R., Nicholas, A.K., Carvalho, O.P., Khurshid, M., Sossick, A., Reichelt, therapy. Biomed. Res. Int. (Epub ahead of print).
S., DSantos, C., Woods, C.G., Gergely, F., 2011. A primary microcephaly protein Zhang, B., Wang, E., Dai, H., Hu, R., Liang, Y., Li, K., Wang, G., Peng, G., Lin, S.Y., 2013.
complex forms a ring around parental centrioles. Nat. Genet. 43, 11471153. BRIT1 regulates p 53 stability and functions as a tumor suppressor in breast
Supic, G., Kozomara, R., Brankovic-Magic, M., Jovic, N., Magic, Z., 2009. Gene hyper- cancer. Carcinogenesis (Epub ahead of print).
methylation in tumor tissue of advanced oral squamous cell carcinoma patients. Zhong, X., Pfeifer, G.P., Xu, X., 2006. Microcephalin encodes a centrosomal protein.
Oral Oncol. 45, 10511057. Cell Cycle. 5, 457458.
Tibelius, A., Marhold, J., Zentgraf, H., Heilig, C.E., Neitzel, H., Ducommun, B., Rauch, Zhou, Z.W., Tapias, A., Bruhn, C., Gruber, R., Sukchev, M., Wang, Z.Q., 2013. DNA
A., Ho, A.D., Bartek, J., Krmer, A., 2009. Microcephalin and pericentrin regulate damage response in microcephaly development of MCPH1 mouse model. DNA
mitotic entry via centrosome-associated Chk1. J. Cell Biol. 185, 11491157. Repair (Amst.) 12, 645655 (Epub ahead of print).

Potrebbero piacerti anche