Entry - *116952 - CELL DIVISION CYCLE 42; CDC42 - OMIM
* 116952

CELL DIVISION CYCLE 42; CDC42


Alternative titles; symbols

GTP-BINDING PROTEIN, 25-KD; G25K


HGNC Approved Gene Symbol: CDC42

Cytogenetic location: 1p36.12     Genomic coordinates (GRCh38): 1:22,052,709-22,101,360 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
1p36.12 Takenouchi-Kosaki syndrome 616737 AD 3

TEXT

Description

CDC42 is a Ras (see 190020)-related GTP-binding protein. It is implicated in a variety of biologic activities, including establishment of cell polarity in yeast, regulation of cell morphology, motility, and cell cycle progression in mammalian cells, and induction of malignant transformation (summary by Wu et al., 2000).


Cloning and Expression

Shinjo et al. (1990) isolated cDNA clones that code for cdc42, a low molecular weight GTP-binding protein originally designated G(p) and also called G25K, from a human placenta library. The predicted amino acid sequence of the protein was very similar to those of various members of the RAS superfamily of low molecular weight GTP-binding proteins, including NRAS, KRAS, HRAS, and the RHO proteins. The highest degree of sequence identity (80%) was found with the Saccharomyces cerevisiae cell division cycle protein CDC42. The human placental gene complemented a cdc42 mutation in S. cerevisiae. Munemitsu et al. (1990) presented further evidence that G25K is the human homolog of the CDC42 gene product.

Marks and Kwiatkowski (1996) identified 2 isoforms of mouse Cdc42. They demonstrated that the 2 murine isoforms arise from a single gene by alternative splicing. Although one is expressed in a wide variety of tissues, the second isoform appeared to be expressed exclusively in brain.


Gene Function

Erickson et al. (1996) used cell fractionation and immunofluorescence to show that cdc42 is localized to the Golgi apparatus of mammalian cells. It colocalizes with the nonclathrin coat proteins ARF3 (103190) and COPB (600959) and its Golgi localization is disrupted by the drug brefeldin A. Based on their findings, Erickson et al. (1996) suggested that cdc42 may be involved in the delivery of newly synthesized proteins and lipids to the plasma membrane and that the GTP-binding/GTPase cycle may dictate its subcellular localization.

By screening rat brain cytosol for proteins that interacted with Ras-related GTPases, or p21 proteins, of the Rho (RHOA; 165390) subfamily, Manser et al. (1994) identified 3 proteins, designated PAKs (see PAK1; 602590) that interacted with the GTP-bound forms of human CDC42 and RAC1 (602048), but not RHOA. Brown et al. (1996) found that activity of human PAK1 was induced by coexpression with RAC1 or CDC42.

Zheng et al. (1996) reported that the FGD1 protein (305400) acts as a cdc42-specific GDP-GTP exchange factor. Cells expressing a fragment of the FGD1 protein encompassing the pleckstrin and Dbl homology domains activated 2 elements downstream of cdc42, namely, Jun kinase (165160) and p70 S6 kinase.

Manser et al. (1993) identified ACK1 (606994) as a binding partner and inhibitor of the GTP-bound form of CDC42. Interaction between GTP-CDC42 and ACK1 inhibited both the intrinsic and GAP-stimulated GTPase activity of CDC42.

CDC42 can regulate the actin cytoskeleton through activation of WASP family members (see 301000). Activation relieves an autoinhibitory contact between the GTPase-binding domain and the C-terminal region of WASP proteins. Kim et al. (2000) reported the autoinhibited structure of the GTPase-binding domain of WASP, which can be induced by the C-terminal region or by organic cosolvents. In the autoinhibited complex, intramolecular interactions with the GTPase-binding domain occlude residues of the C terminus that regulate the Arp2/3 actin-nucleating complex (see 604221). Binding of CDC42 to the GTPase-binding domain causes a dramatic conformational change, resulting in disruption of the hydrophobic core and release of the C terminus, enabling its interaction with the actin regulatory machinery.

Wu et al. (2000) identified a CDC42 mutant, Cdc42F28L, that binds GTP in the absence of a guanine nucleotide exchange factor, but still hydrolyzes GTP with a turnover number identical to that for wildtype CDC42. Expression of this mutant in fibroblasts causes cellular transformation, mimicking many of the characteristics of cells transformed by the DBL oncoprotein (311030), a known guanine nucleotide exchange factor for CDC42. Wu et al. (2000) searched for new CDC42 targets in an effort to understand how CDC42 mediates cellular transformation. They identified the gamma-subunit of the coatomer complex (gamma-COP; 604355) as a specific binding partner for activated CDC42. The binding of CDC42 to gamma-COP is essential for a transforming signal distinct from those elicited by Ras.

Dendritic cells (DCs) developmentally regulate antigen uptake by controlling their endocytic capacity. Immature DCs actively internalize antigen. Mature DCs, however, are poorly endocytic, functioning instead to present antigens to T cells. Garrett et al. (2000) found that endocytic downregulation reflects a decrease in endocytic activity controlled by RHO family GTPases, especially CDC42. Blocking CDC42 function by toxin B treatment or injection of dominant-negative inhibitors of CDC42 abrogated endocytosis in immature DCs. In mature DCs, injection of constitutively active CDC42 or microbial delivery of a CDC42 nucleotide exchange factor reactivated endocytosis. DCs regulated endogenous levels of CDC42-GTP with activated CDC42 detectable only in immature cells. Garrett et al. (2000) concluded that DCs developmentally regulate endocytosis at least in part by controlling levels of activated CDC42.

Using Madin Darby canine kidney (MDCK) cells expressing Cdc42 mutants defective in nucleotide binding or hydrolysis, Musch et al. (2001) showed that Cdc42 differentially regulated the exit of apical and basolateral proteins from the trans-Golgi network (TGN). GTPase-deficient Cdc42 accelerated the exit of an apical marker from the TGN and inhibited the release of basolateral proteins. Basolateral protein transport by Cdc42 with an activating mutation was accompanied by changes in the organization of the actin cytoskeleton.

Sin et al. (2002) used in vivo time-lapse imaging of optic tectal cells in Xenopus laevis tadpoles to demonstrate that enhanced visual activity driven by a light stimulus promotes dendritic arbor growth. The stimulus-induced dendritic arbor growth requires glutamate receptor (see 138249)-mediated synaptic transmission, decreased RhoA (165390) activity, and increased RAC and CDC42 activity. Sin et al. (2002) concluded that their results delineated a role for Rho GTPases in the structural plasticity driven by visual stimulation in vivo.

Morphologic changes in dendritic spines are believed to be caused by dynamic regulation of actin polymerization. Irie and Yamaguchi (2002) found that the EphB2 receptor tyrosine kinase (600997) physically associates with the guanine nucleotide exchange factor intersectin-1 (602442) in cooperation with the actin-regulating protein N-WASP (605056), which in turn activates Cdc42 and spine morphogenesis.

In higher eukaryotes, the small GTPase CDC42, acting through a PAR6-atypical protein kinase C (see PKC-zeta, 176982) complex, is required to establish cellular asymmetry during epithelial morphogenesis, asymmetric cell division, and directed cell migration. Etienne-Manneville and Hall (2003) used primary rat astrocytes in a cell migration assay to demonstrate that PAR6-PKC-zeta interacts directly with and regulates glycogen synthase kinase-3-beta (GSK3-beta; 605004) to promote polarization of the centrosome and to control the direction of cell protrusion. CDC42-dependent phosphorylation of GSK3-beta occurs specifically at the leading edge of migrating cells, and induces the interaction of APC (611731) protein with the plus ends of microtubules. The association of APC with microtubules is essential for cell polarization. Etienne-Manneville and Hall (2003) concluded that CDC42 regulates cell polarity through the spatial regulation of GSK3-beta and APC.

Wu et al. (2003) presented evidence that activation of CDC42 protects EGF receptor (EGFR; 131550) from the negative regulatory activity of CBL (165360), a ubiquitin ligase.

Yasuda et al. (2004) demonstrated that Cdc42 and mDia3 (DIAPH2; 300108) regulate microtubule attachment to kinetochores.

Nalbant et al. (2004) reported the development of a biosensor capable of visualizing the changing activation of endogenous unlabeled Cdc42 in living cells. With the use of a dye that reports protein interactions, the biosensor revealed localized activation in the trans-Golgi apparatus, microtubule-dependent Cdc42 activation at the cell periphery, and activation kinetics precisely coordinated with cell extension and retraction.

By electroporating genes into chicken presomitic mesenchymal cells, Nakaya et al. (2004) demonstrated that Cdc42 and Rac1 play different roles in mesenchymal-epithelial transition. Different levels of Cdc42 appeared to affect the binary decision between epithelial and mesenchymal states. Proper levels of Rac1 were also necessary for somitic epithelialization, since cells with either activated or inhibited Rac1 failed to undergo correct epithelialization.

Using functional and proteomic screens to identify regulators of Cdc42, Wells et al. (2006) identified a network of proteins that centered on Rich1 (ARHGAP17; 608293) and organized apical polarity in canine kidney epithelial cells. Rich1 bound the coiled-coil domain of Amot (300410) and was thereby targeted to a complex at tight junctions containing the PDZ domain-containing proteins Pals1 (MPP5; 606958), Patj (INADL; 603199), and Par3 (PARD3; 606745). Regulation of Cdc42 by Rich1 was required for maintenance of tight junctions. The coiled-coil domain of Amot was required for its localization to apical membranes and for Amot to relocalize Pals1 and Par3 to internal puncta. Wells et al. (2006) proposed that RICH1 and AMOT maintain tight junction integrity by coordinated regulation of CDC42 and by linking specific components of the tight junction to intracellular protein trafficking.

Formation of the apical surface and lumen is a fundamental step in epithelial organ development. Martin-Belmonte et al. (2007) showed that Pten (601728) localized to the apical plasma membrane during epithelial morphogenesis to mediate enrichment of phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) at this domain during cyst development in a 3-dimensional Madin-Darby canine kidney cell system. Ectopic PtdIns(4,5)P2 at the basolateral surface caused apical proteins to relocalize to the basolateral surface. Annexin-2 (ANX2; 151740) bound PtdIns(4,5)P2 and was recruited to the apical surface. Anx2 bound Cdc42 and recruited it to the apical surface, and Cdc42 in turn recruited the Par6 (607484)/atypical protein kinase C (aPKC; see 176982) complex to the apical surface. Loss of function of Pten, Anx2, Cdc42, or aPKC prevented normal development of the apical surface and lumen. Martin-Belmonte et al. (2007) concluded that PTEN, PtdIns(4,5)P2, ANX2, CDC42, and aPKC control apical plasma membrane and lumen formation.

Hamann et al. (2007) found that both ASEF1 and ASEF2 were guanine nucleotide exchange factors (GEFs) for CDC42, but not for RAC1 or RHOA. ASEF2 required the lipid-modified form of CDC42. Using deletion mutants, Hamann et al. (2007) showed that the tandem N-terminal ABR and SH3 domain (ABRSH3) of the ASEF proteins was required to bind the armadillo repeat region of APC. ABRSH3 also functioned in an autoinhibitory reaction by binding the C-terminal tails of ASEF1 and ASEF2 and inhibiting their GEF activities. Deletion of ABRSH3 or coexpression of the APC armadillo repeat sequence with full-length ASEF2 stimulated filopodia formation in transfected HeLa cells. Hamann et al. (2007) concluded that activation of ASEF1 and ASEF2 involves binding of APC to ABRSH3, which disrupts the autoinhibitory interaction of ABRSH3 with the ASEF C-terminal tail and allows GDP/GTP exchange on CDC42.

Kawasaki et al. (2007) found that ASEF2 was a GEF for both CDC42 and RAC1 in MDCK and HeLa cells. Overexpression of ASEF2 increased membrane ruffling and CDC42-mediated filopodia formation in HeLa cells.

Shen et al. (2008) showed that Nudel (NDEL1; 607538) colocalized with Cdc42gap (ARHGAP1; 602732) at the leading edge of migrating NIH3T3 mouse fibroblasts. This localization of Nudel required its phosphorylation by Erk1 (MAPK3; 601795)/Erk2 (MAPK1; 176948). Shen et al. (2008) found that Nudel competed with Cdc42 for binding Cdc42gap. Consequently, Nudel inhibited Cdc42gap-mediated inactivation of Cdc42 in a dose-dependent manner. Depletion of Nudel by RNA interference or overexpression of a nonphosphorylatable Nudel mutant abolished Cdc42 activation and cell migration. Shen et al. (2008) concluded that NUDEL facilitates cell migration by sequestering CDC42GAP at the leading edge to stabilize active CDC42 in response to extracellular stimuli.

Kang et al. (2008) performed a global characterization of rat neural palmitoyl proteomes and identified most of the known neural palmitoyl proteins, 68 in total, plus more than 200 novel palmitoyl protein candidates, with further testing confirming palmitoylation for 21 of these candidates. The novel palmitoyl proteins included neurotransmitter receptors, transporters, adhesion molecules, scaffolding proteins, as well as SNAREs and other vesicular trafficking proteins. Of particular interest was the finding of palmitoylation for a brain-specific Cdc42 splice variant. The palmitoylated Cdc42 isoform (Cdc42-palm) differs from the canonical, prenylated form (Cdc42-prenyl), with regard to both localization and function: Cdc42-palm concentrates in dendritic spines and has a special role in inducing these postsynaptic structures. Furthermore, assessing palmitoylation dynamics in drug-induced activity models identified rapidly induced changes for Cdc42 as well as for other synaptic palmitoyl proteins, suggesting that palmitoylation may participate broadly in the activity-driven changes that shape synapse morphology and brain function.

Machacek et al. (2009) examined GTPase coordination in mouse embryonic fibroblasts both through simultaneous visualization of 2 GTPase biosensors and using a 'computational multiplexing' approach capable of defining the relationships between multiple protein activities visualized in separate experiments. They found that RhoA (165390) is activated at the cell edge synchronous with edge advancement, whereas Cdc42 and Rac1 are activated 2 microns behind the edge with a delay of 40 seconds. This indicates that Rac1 and RhoA operate antagonistically through spatial separation and precise timing, and that RhoA has a role in the initial events of protrusion, whereas Rac1 and Cdc42 activate pathways implicated in reinforcement and stabilization of newly expanded protrusions.

Frank et al. (2009) found that Cdc42 was involved in a signaling complex that modulated presynaptic Cav2.1 (CACNA1A; 601011) calcium channels in the Drosophila neuromuscular junction. This signaling system involved the Rho-type GEF ephexin (NGEF; 605991), which appeared to act with Cdc42 to activate the Eph receptor (see EPHA1; 179610) for modulation of Cav2.1 channel activity.

Murakoshi et al. (2011) used 2-photon fluorescence lifetime imaging microscopy to monitor the activity of 2 Rho GTPases, RhoA and Cdc42, in single dendritic spines undergoing structural plasticity associated with long-term potentiation in CA1 pyramidal neurons in cultured slices of rat hippocampus. When long-term volume increase was induced in a single spine using 2-photon glutamate uncaging, RhoA and Cdc42 were rapidly activated in the stimulated spine. These activities decayed over about 5 minutes, and were then followed by a phase of persistent activation lasting more than half an hour. Although active RhoA and Cdc42 were similarly mobile, their activity patterns were different. RhoA activation diffused out of the stimulated spine and spread over about 5 microns along the dendrite. In contrast, Cdc42 activation was restricted to the stimulated spine, and exhibited a steep gradient at the spine necks. Inhibition of the Rho-Rock pathway preferentially inhibited the initial spine growth, whereas the inhibition of the Cdc42-Pak pathway blocked the maintenance of sustained structural plasticity. RhoA and Cdc42 activation depended on calcium ion/calmodulin-dependent kinase (CaMKII). Thus, Murakoshi et al. (2011) concluded that RhoA and Cdc42 relay transient CaMKII activation to synapse-specific, long-term signaling required for spine structural plasticity.

Using synthetic derivatives of the enteropathogenic Escherichia coli guanine-nucleotide exchange factor Map, Orchard et al. (2012) found that CDC42 GTPase signal transduction was controlled by interaction between Map and the PDZ domains of EBP50 (SLC9A3R1; 604990) and the induction of clusters of actin-rich membrane protrusions.

Keestra et al. (2013) demonstrated that NOD1 (605980) senses cytosolic microbial products by monitoring the activation state of small Rho GTPases. Activation of RAC1 (602048) and CDC42 by bacterial delivery or ectopic expression of SopE, a virulence factor of the enteric pathogen Salmonella, triggered the NOD1 signaling pathway with consequent RIP2 (603455)-mediated induction of NF-kappa-B (see 164011)-dependent inflammatory responses. Similarly, activation of the NOD1 signaling pathway by peptidoglycan required RAC1 activity. Furthermore, Keestra et al. (2013) showed that constitutively active forms of RAC1, CDC42, and RHOA (165390) activated the NOD1 signaling pathway.

Florian et al. (2013) reported an unexpected shift from canonical to noncanonical Wnt signaling in mice due to elevated expression of Wnt5a (164975) in aged hematopoietic stem cells (HSCs), which causes stem cell aging. Wnt5a treatment of young HSCs induced aging-associated stem cell apolarity, reduction of regenerative capacity, and an aging-like myeloid-lymphoid differentiation skewing via activation of the small Rho GTPase Cdc42. Conversely, Wnt5a haploinsufficiency attenuated HSC aging, whereas stem cell-intrinsic reduction of Wnt5a expression resulted in functionally rejuvenated aged HSCs. Florian et al. (2013) concluded that the data demonstrated a critical role for stem cell-intrinsic noncanonical Wnt5a signaling in HSC aging.

Park et al. (2015) showed that the coat protein I complex (COPI; see 601924) sorts anterograde cargoes into tubules that connect the Golgi cisternae in human cells. Moreover, the small GTPase CDC42 regulates bidirectional Golgi transport by targeting the dual functions of COPI in cargo sorting and carrier formation. CDC42 also directly imparts membrane curvature to promote COPI tubule formation. Park et al. (2015) concluded that their findings further revealed that COPI tubular transport complements cisternal maturation in explaining how anterograde Golgi transport is achieved, and that bidirectional COPI transport is modulated by environmental cues through CDC42.

By transfecting MDCK canine kidney cell with wildtype or mutant human constructs, and by knockdown of endogenous MDCK proteins, Hayase et al. (2013) studied development of apical-basolateral polarity. Their results suggested that cytoplasmic MORG1 (WDR83; 616850) interacted directly with PAR6 (607484) and mediated apical translocation of a dimer made up of PAR6 and an atypical PKC (aPKC, e.g., PRKCI, 600539). MORG1 also interacted with the apical transmembrane protein CRB3 (609737) and facilitated binding between PAR6 and CRB3, thereby anchoring the PAR6-aPKC dimer to the apical membrane. The small GTPase CDC42 displaced MORG1 from the complex at the apical membrane and strengthened the association between PAR6 and CRB3. Knockdown of any complex component interfered with development of polarity in MDCK cells; however, overexpression of a CRB3-aPKC construct reversed the polarity defect in MORG1-deficient MDCK cells and restored apical identity.

In female meiosis, selfish elements drive by preferentially attaching to the egg side of the spindle. This implies some asymmetry between the 2 sides of the spindle. Akera et al. (2017) found that CDC42 signaling from the cell cortex regulated microtubule tyrosination to induce spindle asymmetry and that non-Mendelian segregation depended on this asymmetry. Cortical CDC42 depends on polarization directed by chromosomes, which are positioned near the cortex to allow the asymmetric cell division. Thus, Akera et al. (2017) selfish meiotic drivers exploit the asymmetry inherent in female meiosis to bias their transmission.

Hedrick et al. (2016) described a 3-molecule model of structural long-term potentiation of murine dendritic spines, implicating the localized, coincident activation of Rac1, RhoA, and Cdc42 as a causal signal of structural long-term potentiation. This model posited that complete tripartite signal overlap in spines confers structural long-term potentiation, but that partial overlap primes spines for structural plasticity. By monitoring the spatiotemporal activation patterns of these GTPases during structural long-term potentiation, Hedrick et al. (2016) found that such spatiotemporal signal complementation simultaneously explains 3 integral features of plasticity: the facilitation of plasticity by BDNF (113505), the postsynaptic source of which activates Cdc42 and Rac1, but not RhoA; heterosynaptic facilitation of structural long-term potentiation, which is conveyed by diffusive Rac1 and RhoA activity; and input specificity, which is afforded by spine-restricted Cdc42 activity.


Gene Structure

Nicole et al. (1999) determined the organization of the CDC42 gene and found that the gene encodes the placental and brain isoforms generated by alternative splicing.

Moats-Staats and Stiles (1995) showed that the 5-prime end of another gene, called BB1 by them, overlaps the 3-prime end of G25K.


Biochemical Features

Crystal Structure

Through a structural analysis of DOCK9 (607325)-CDC42 complexes, Yang et al. (2009) identified a nucleotide sensor within the alpha-10 helix of the DHR2 domain that contributes to release of guanine diphosphate (GDP) and then to discharge of the activated GTP-bound Cdc42. Magnesium exclusion, a critical factor in promoting GDP release, is mediated by a conserved valine residue within this sensor, whereas binding of GTP-magnesium ion to the nucleotide-free complex results in magnesium-inducing displacement of the sensor to stimulate discharge of Cdc42-GTP. Yang et al. (2009) concluded that their studies identified an unusual mechanism of GDP release and defined the complete guanine nucleotide exchange factor catalytic cycle from GDP dissociation followed by GTP binding and discharge of the activated GTPase.


Mapping

Using SSCP analysis of a mouse backcross panel, Marks and Kwiatkowski (1996) demonstrated that the gene encoding cdc42 is localized to the distal portion of mouse chromosome 4 between Ephb2 proximally and Cappb (601572) distally. The human homologs of both of the 2 flanking genes were mapped to human chromosome 1p36.1 by Barron-Casella et al. (1995), thus indicating that this is the likely site of the human CDC42 gene. The CDC42 gene was mapped to the 1p36-p35 region by radiation hybrid analysis (Schuler et al., 1996; Jensen et al., 1997; Deloukas et al., 1998).

Nicole et al. (1999) demonstrated a CDC42-like transcript on chromosome 4 that does not contain introns and is similar to the placental isoform, suggesting that it is a processed pseudogene.

Nicole et al. (1999) excluded the CDC42 gene as the site of mutation in the Schwartz-Jampel syndrome type 1 (255800).


Molecular Genetics

Takenouchi et al. (2015) and Takenouchi et al. (2016) each reported a patient with macrothrombocytopenia, lymphedema, developmental delay, and similar distinctive facial features, known as Takenouchi-Kosaki syndrome (TKS; 616737). Both patients were heterozygous for the same de novo mutation in the CDC42 gene (116952.0001).

In 15 patients from 13 unrelated families with a heterogeneous developmental disorder consistent with TKS, Martinelli et al. (2018) identified 9 different heterozygous missense mutations in the CDC42 gene (see, e.g., 116952.0001-116952.0006). The mutations occurred de novo in 12 unrelated patients; there was 1 family (family 30153) in which 3 affected individuals carried the same mutation. The mutations in most patients were identified by whole-exome sequencing; some mutations were identified by direct sequencing of the CDC42 gene. None of the mutations were found in the ExAC/gnomAD database, and all were predicted to be pathogenic according to ACMG criteria. Based on molecular modeling, predicted functional impact, and in vitro functional studies, the mutations were categorized into 3 main groups, all of which were determined to be pathogenic (see GENOTYPE/PHENOTYPE CORRELATIONS).


Genotype/Phenotype Correlations

Based on molecular modeling, predicted functional impact, and in vitro functional studies, the mutations identified by Martinelli et al. (2018) were categorized into 3 main groups. Group I mutations (Y64C; R66G, 116952.0002; and R68Q) occurred in the switch II domain, which mediates CDC42 binding to effectors and regulators, and were predicted to interfere with the catalytic activity of the GTPase and/or its capability to transduce signaling. Group I mutations were associated with a syndromic form of thrombocytopenia. Group II mutations (C81F, 116952.0003; S83P, 116952.0004; and A159V) were located within or close to the nucleotide-binding pocket, and were predicted to promote fast GDP/GTP cycling, favoring a hyperactive GTP-bound state. Group II mutations were associated with a variable developmental disorder characterized by striking dysmorphic features resembling a RASopathy. Group III mutations (I21T, 116952.0005; Y23C; and E171K, 116952.0006), located in residues predicted to disrupt interactions with effectors containing a CRIB (Cdc42, Rac interactive binding) motif, were associated with a milder phenotype resembling Noonan syndrome. In vitro studies of selected mutations using recombinant proteins showed variable effects on CDC42 function, including altering the switch between the active and inactive states of the GTPase and/or affecting CDC42 interaction with effectors. Group I mutations had defective interaction with tested partner proteins, group II mutations showed variable hyperactive behavior, and group III mutations showed perturbed binding to effectors. In addition, a wound healing assay indicated that the mutations resulted in dysregulated and disturbed cell polarization and proliferation. One specific mutation (E171K), which affects only 1 of the 2 CDC42 isoforms and specifically impaired binding to WASP (300392), resulted in an overall milder clinical phenotype that phenocopied Noonan syndrome. Studies in C. elegans showed that the mutations caused variable disruption of developmental processes, with some mutations acting as a gain of function and others acting as hypomorphs. In general, the group II mutations upregulated multiple signaling pathways. Overall, the findings indicated that CDC42 functions in a large array of developmental processes.


Animal Model

Wu et al. (2006) stated that constitutive knockout of Cdc42 in mice results in death around implantation. In order to examine the role of Cdc42 in the differentiation of skin stem cells into hair follicles, they targeted Cdc42 deletion to keratinocytes. Mutant mice were born without obvious defects but showed impaired hair formation and growth retardation. Within 4 weeks, all hairs were lost and did not grow again in older animals. In the absence of Cdc42, degradation of beta-catenin (CTNNB1; 116806) increased corresponding to decreased phosphorylation of Gsk3-beta and increased phosphorylation of axin (603816), which is required for binding of beta-catenin to the degradation machinery. Wu et al. (2006) concluded that Cdc42 regulation of beta-catenin turnover is required for terminal differentiation of hair follicle progenitor cells.

By targeted deletion of Cdc42 in telencephalic neural progenitors in mouse embryos, Chen et al. (2006) found that Cdc42 was essential for establishment of apical-basal polarity of the telencephalic neuroepithelium, a necessity for expansion and bifurcation of cerebral hemispheres.

Pleines et al. (2010) found that conditional knockout of Cdc42 in mice results in mild thrombocytopenia and increased platelet size (i.e., macrothrombocytopenia).


ALLELIC VARIANTS ( 6 Selected Examples):

.0001 TAKENOUCHI-KOSAKI SYNDROME

CDC42, TYR64CYS (SCV000577577.2)
  
RCV000203307...

In 2 unrelated females, one of Japanese-Iranian descent and the other of Japanese descent, with a syndrome of macrothrombocytopenia, developmental delay, and distinctive facial features (TKS; 616737), Takenouchi et al. (2015) and Takenouchi et al. (2016) identified a heterozygous c.191A-G transition (c.191A-G, NM_001039802) in exon 3 of the CDC42 gene, resulting in a tyrosine-to-cystine substitution at codon 64 (Y64C). The mutation was confirmed in both patients by Sanger sequencing and was not identified in either parent.

In a 15-year-old girl (patient LR17-420) with TKS, Martinelli et al. (2018) identified a de novo heterozygous Y64C mutation in the CDC42 gene. The mutation, which was found by whole-exome sequencing, was not found in the ExAC/gnomAD database and met the ACMG criteria for pathogenicity.


.0002 TAKENOUCHI-KOSAKI SYNDROME

CDC42, ARG66GLY (SCV000244118.3)
  
RCV000190678...

In 2 unrelated patients (LR14-352 and PCGC 1-04248) with Takenouchi-Kosaki syndrome (TKS; 616737), Martinelli et al. (2018) identified a de novo heterozygous c.196A-G transition (c.196A-G, NM_001791.3) in exon 3 of the CDC42 gene, resulting in an arg66-to-gly (R66G) substitution at a conserved residue in the switch II domain. The mutation, which was found by whole-exome sequencing, was not found in the ExAC/gnomAD database and met the ACMG criteria for pathogenicity.


.0003 TAKENOUCHI-KOSAKI SYNDROME

CDC42, CYS81PHE (SCV000589746.1)
  
RCV000497401...

In a 4-year-old boy (LR17-032), born of unrelated parents, with Takenouchi-Kosaki syndrome (TKS; 616737), Martinelli et al. (2018) identified a de novo heterozygous c.242G-T transversion (c.242G-T, NM_001791.3) in exon 3 of the CDC42 gene, resulting in a cys81-to-phe (C81F) substitution at a conserved residue in the beta-4 domain. The mutation, which was found by whole-exome sequencing, was not found in the ExAC/gnomAD database and met the ACMG criteria for pathogenicity.


.0004 TAKENOUCHI-KOSAKI SYNDROME

CDC42, SER83PRO (SCV000678255)
  
RCV000577866...

In a boy (LR10-046), conceived by in vitro fertilization, with Takenouchi-Kosaki syndrome (TKS; 616737), Martinelli et al. (2018) identified a de novo heterozygous c.247T-C transition (c.247T-C, NM_001791.3) in exon 3 of the CDC42 gene, resulting in a ser83-to-pro (S83P) substitution in the beta-4 domain. The mutation, which was found by whole-exome sequencing, was not found in the ExAC/gnomAD database and met the ACMG criteria for pathogenicity.


.0005 TAKENOUCHI-KOSAKI SYNDROME

CDC42, ILE21THR (SCV000572034.2)
  
RCV000481008...

In a patient (LR16-483) with Takenouchi-Kosaki syndrome (TKS; 616737), Martinelli et al. (2018) identified a de novo heterozygous c.62T-C transition (c.62T-C, NM_001791.3) in exon 1 of the CDC42 gene, resulting in an ile21-to-thr (I21T) substitution at a conserved residue in the alpha-1 domain. The mutation, which was found by whole-exome sequencing, was not found in the ExAC/gnomAD database and met the ACMG criteria for pathogenicity.


.0006 TAKENOUCHI-KOSAKI SYNDROME

CDC42, GLU171LYS (SCV000678257)
  
RCV000577848...

In 3 members of a family (family 30153) with Takenouchi-Kosaki syndrome (TKS; 616737), Martinelli et al. (2018) identified a heterozygous c.511G-A transition (c.511G-A, NM_001791.3) in exon 5 of the CDC42 gene, resulting in a glu171-to-lys (E171K) substitution at a conserved residue in the CBR domain. An unrelated patient (M060721) with a similar disorder carried this mutation in the de novo state. The mutation only affected transcript variant 1 and isoform 1 of CDC42. The mutation was not found in the ExAC/gnomAD database and met the ACMG criteria for pathogenicity.


REFERENCES

  1. Akera, T., Chmatal, L., Trimm, E., Yang, K., Aonbangkhen, C., Chenoweth, D. M., Janke, C., Schultz, R. M., Lampson, M. A. Spindle asymmetry drives non-Mendelian chromosome segregation. Science 358: 668-672, 2017. [PubMed: 29097549, images, related citations] [Full Text]

  2. Barron-Casella, E. A., Torres, M. A., Scherer, S. W., Heng, H. H., Tsui, L. C., Casella, J. F. Sequence analysis and chromosomal localization of human Cap Z: conserved residues within the actin-binding domain may link Cap Z to gelsolin/severin and profilin protein families. J. Biol. Chem. 270: 21472-21479, 1995. [PubMed: 7665558, related citations] [Full Text]

  3. Brown, J. L., Stowers, L., Baer, M., Trejo, J., Coughlin, S., Chant, J. Human Ste20 homologue hPAK1 links GTPases to the JNK MAP kinase pathway. Curr. Biol. 6: 598-605, 1996. [PubMed: 8805275, related citations] [Full Text]

  4. Chen, L., Liao, G., Yang, L., Campbell, K., Nakafuku, M., Kuan, C.-Y., Zheng, Y. Cdc42 deficiency causes Sonic hedgehog-independent holoprosencephaly. Proc. Nat. Acad. Sci. 103: 16520-16525, 2006. [PubMed: 17050694, images, related citations] [Full Text]

  5. Deloukas, P., Schuler, G. D., Gyapay, G., Beasley, E. M., Soderlund, C., Rodriguez-Tome, P., Hui, L., Matise, T. C., McKusick, K. B., Beckmann, J. S., Bentolila, S., Bihoreau, M.-T., and 53 others. A physical map of 30,000 human genes. Science 282: 744-746, 1998. [PubMed: 9784132, related citations] [Full Text]

  6. Erickson, J. W., Zhang, C., Kahn, R. A., Evans, T., Cerione, R. A. Mammalian cdc42 is a brefeldin A-sensitive component of the Golgi apparatus. J. Biol. Chem. 271: 26850-26854, 1996. [PubMed: 8900167, related citations] [Full Text]

  7. Etienne-Manneville, S., Hall, A. Cdc42 regulates GSK-3-beta and adenomatous polyposis coli to control cell polarity. Nature 421: 753-756, 2003. [PubMed: 12610628, related citations] [Full Text]

  8. Florian, M. C., Nattamai, K. J., Dorr, K., Marka, G., Uberle, B., Vas, V., Eckl, C., Andra, I., Schiemann, M., Oostendorp, R. A. J., Scharffetter-Kochanek, K., Kestler, H. A., Zheng, Y., Geiger, H. A canonical to non-canonical Wnt signalling switch in haematopoietic stem-cell ageing. Nature 503: 392-396, 2013. [PubMed: 24141946, images, related citations] [Full Text]

  9. Frank, C. A., Pielage, J., Davis, G. W. A presynaptic homeostatic signaling system composed of the Eph receptor, Ephexin, Cdc42, and Cav2.1 calcium channels. Neuron 61: 556-569, 2009. [PubMed: 19249276, images, related citations] [Full Text]

  10. Garrett, W. S., Chen, L.-M., Kroschewski, R., Ebersold, M., Turley, S., Trombetta, S., Galan, J. E., Mellman, I. Developmental control of endocytosis in dendritic cells by Cdc42. Cell 102: 325-334, 2000. [PubMed: 10975523, related citations] [Full Text]

  11. Hamann, M. J., Lubking, C. M., Luchini, D. N., Billadeau, D. D. Asef2 functions as a Cdc42 exchange factor and is stimulated by the release of an autoinhibitory module from a concealed C-terminal activation element. Molec. Cell. Biol. 27: 1380-1393, 2007. [PubMed: 17145773, images, related citations] [Full Text]

  12. Hayase, J., Kamakura, S., Iwakiri, Y., Yamaguchi, Y., Izaki, T., Ito, T., Sumimoto, H. The WD40 protein Morg1 facilitates Par6-aPKC binding to Crb3 for apical identity in epithelial cells. J. Cell Biol. 200: 635-650, 2013. [PubMed: 23439680, images, related citations] [Full Text]

  13. Hedrick, N. G., Harward, S. C., Hall, C. E., Murakoshi, H., McNamara, J. O., Yasuda, R. Rho GTPase complementation underlies BDNF-dependent homo- and heterosynaptic plasticity. Nature 538: 104-108, 2016. [PubMed: 27680697, images, related citations] [Full Text]

  14. Irie, F., Yamaguchi, Y. EphB receptors regulate dendritic spine development via intersectin, Cdc42 and N-WASP. Nature Neurosci. 5: 1117-1118, 2002. [PubMed: 12389031, related citations] [Full Text]

  15. Jensen, S. J., Sulman, E. P., Maris, J. M., Matise, T. C., Vojta, P. J., Barrett, J. C., Brodeur, G. M., White, P. S. An integrated transcript map of human chromosome 1p35-p36. Genomics 42: 126-136, 1997. [PubMed: 9177782, related citations] [Full Text]

  16. Kang, R., Wan, J., Arstikaitis, P., Takahashi, H., Huang, K., Bailey, A. O., Thompson, J. X., Roth, A. F., Drisdel, R. C., Mastro, R., Green, W. N., Yates, J. R., III, Davis, N. G., El-Husseini, A. Neural palmitoyl-proteomics reveals dynamic synaptic palmitoylation. Nature 456: 904-909, 2008. [PubMed: 19092927, images, related citations] [Full Text]

  17. Kawasaki, Y., Sagara, M., Shibata, Y., Shirouzu, M., Yokoyama, S., Akiyama, T. Identification and characterization of Asef2, a guanine-nucleotide exchange factor specific for Rac1 and Cdc42. Oncogene 26: 7620-7627, 2007. [PubMed: 17599059, related citations] [Full Text]

  18. Keestra, A. M., Winter, M. G., Auburger, J. J., Frassle, S. P., Xavier, M. N., Winter, S. E., Kim, A., Poon, V., Ravesloot, M. M., Waldenmaier, J. F. T., Tsolis, R. M., Eigenheer, R. A., Baumler, A. J. Manipulation of small Rho GTPases is a pathogen-induced process detected by NOD1. Nature 496: 233-237, 2013. [PubMed: 23542589, images, related citations] [Full Text]

  19. Kim, A. S., Kakalis, L. T., Abdul-Manan, N., Liu, G. A., Rosen, M. K. Autoinhibition and activation mechanisms of the Wiskott-Aldrich syndrome protein. Nature 404: 151-158, 2000. [PubMed: 10724160, related citations] [Full Text]

  20. Machacek, M., Hodgson, L., Welch, C., Elliott, H., Pertz, O., Nalbant, P., Abell, A., Johnson, G. L., Hahn, K. M., Danuser, G. Coordination of Rho GTPase activities during cell protrusion. Nature 461: 99-103, 2009. [PubMed: 19693013, related citations] [Full Text]

  21. Manser, E., Leung, T., Salihuddin, H., Tan, L., Lim, L. A non-receptor tyrosine kinase that inhibits the GTPase activity of p21cdc42. Nature 363: 364-367, 1993. [PubMed: 8497321, related citations] [Full Text]

  22. Manser, E., Leung, T., Salihuddin, H., Zhao, Z., Lim, L. A brain serine/threonine protein kinase activated by Cdc42 and Rac1. Nature 367: 40-46, 1994. [PubMed: 8107774, related citations] [Full Text]

  23. Marks, P. W., Kwiatkowski, D. J. Genomic organization and chromosomal location of murine Cdc42. Genomics 38: 13-18, 1996. [PubMed: 8954774, related citations] [Full Text]

  24. Martin-Belmonte, F., Gassama, A., Datta, A., Yu, W., Rescher, U., Gerke, V., Mostov, K. PTEN-mediated apical segregation of phosphoinositides controls epithelial morphogenesis through Cdc42. Cell 128: 383-397, 2007. [PubMed: 17254974, images, related citations] [Full Text]

  25. Martinelli, S., Krumbach, O. H. F., Pantaleoni, F., Coppola, S., Amin, E., Pannone, L., Nouri, K., Farina, L., Dvorsky, R., Lepri, F., Buchholzer, M., Konopatzki, R, and 37 others. Functional dysregulation of CDC42 causes diverse developmental phenotypes. Am. J. Hum. Genet. 102: 309-320, 2018. [PubMed: 29394990, images, related citations] [Full Text]

  26. Moats-Staats, B. M., Stiles, A. D. Southern hybridization analyses of somatic cell hybrids reveal that human BB1 is a member of a multigene family dispersed throughout the human genome and appears to be linked to the human G25K genes. DNA Cell Biol. 14: 465-474, 1995. [PubMed: 7598801, related citations] [Full Text]

  27. Munemitsu, S., Innis, M. A., Clark, R., McCormick, F., Ullrich, A., Polakis, P. Molecular cloning and expression of a G25K cDNA, the human homolog of the yeast cell cycle gene CDC42. Molec. Cell. Biol. 10: 5977-5982, 1990. [PubMed: 2122236, related citations] [Full Text]

  28. Murakoshi, H., Wang, H., Yasuda, R. Local, persistent activation of Rho GTPases during plasticity of single dendritic spines. Nature 472: 100-104, 2011. [PubMed: 21423166, images, related citations] [Full Text]

  29. Musch, A., Cohen, D., Kreitzer, G., Rodriguez-Boulan, E. cdc42 regulates the exit of apical and basolateral proteins from the trans-Golgi network. EMBO J. 20: 2171-2179, 2001. [PubMed: 11331583, images, related citations] [Full Text]

  30. Nakaya, Y., Kuroda, S., Katagiri, Y. T., Kaibuchi, K., Takahashi, Y. Mesenchymal-epithelial transition during somitic segmentation is regulated by differential roles of Cdc42 and Rac1. Dev. Cell 7: 425-438, 2004. [PubMed: 15363416, related citations] [Full Text]

  31. Nalbant, P., Hodgson, L., Kraynov, V., Toutchkine, A., Hahn, K. M. Activation of endogenous Cdc42 visualized in living cells. Science 305: 1615-1619, 2004. [PubMed: 15361624, related citations] [Full Text]

  32. Nicole, S., White, P. S., Topaloglu, H., Beigthon, P., Salih, M., Hentati, F., Fontaine, B. The human CDC42 gene: genomic organization, evidence for the existence of a putative pseudogene and exclusion as a SJS1 candidate gene. Hum. Genet. 105: 98-103, 1999. [PubMed: 10480361, related citations] [Full Text]

  33. Orchard, R. C., Kittisopikul, M., Altschuler, S. J., Wu, L. F., Suel, G. M., Alto, N. M. Identification of F-actin as the dynamic hub in a microbial-induced GTPase polarity circuit. Cell 148: 803-815, 2012. [PubMed: 22341450, images, related citations] [Full Text]

  34. Park, S.-Y., Yang, J.-S., Schmider, A. B., Soberman, R. J., Hsu, V. W. Coordinated regulation of bidirectional COPI transport at the Golgi by CDC42. Nature 521: 529-532, 2015. [PubMed: 25945738, images, related citations] [Full Text]

  35. Pleines, I., Eckly, A., Elvers, M., Hagedorn, I., Eliautou, S., Bender, M., Wu, X., Lanza, F., Gachet, C., Brakebusch, C., Nieswandt, B. Multiple alterations of platelet functions dominated by increased secretion in mice lacking Cdc42 in platelets. Blood 115: 3364-3373, 2010. [PubMed: 20139097, related citations] [Full Text]

  36. Schuler, G. D., Boguski, M. S., Stewart, E. A., Stein, L. D., Gyapay, G., Rice, K., White, R. E., Rodriguez-Tome, P., Aggarwal, A., Bajorek, E., Bentolila, S., Birren, B. B., and 92 others. A gene map of the human genome. Science 274: 540-546, 1996. [PubMed: 8849440, related citations]

  37. Shen, Y., Li, N., Wu, S., Zhou, Y., Shan, Y., Zhang, Q., Ding, C., Yuan, Q., Zhao, F., Zeng, R., Zhu, X. Nudel binds Cdc42GAP to modulate Cdc42 activity at the leading edge of migrating cells. Dev. Cell 14: 342-353, 2008. [PubMed: 18331715, related citations] [Full Text]

  38. Shinjo, K., Koland, J. G., Hart, M. J., Narasimhan, V., Johnson, D. I., Evans, T., Cerione, R. A. Molecular cloning of the gene for the human placental GTP-binding protein G(p) (G25K): identification of this GTP-binding protein as the human homolog of the yeast cell-division-cycle protein CDC42. Proc. Nat. Acad. Sci. 87: 9853-9857, 1990. [PubMed: 2124704, related citations] [Full Text]

  39. Sin, W. C., Haas, K., Ruthazer, E. S., Cline, H. T. Dendrite growth increased by visual activity requires NMDA receptor and Rho GTPases. Nature 419: 475-480, 2002. [PubMed: 12368855, related citations] [Full Text]

  40. Takenouchi, T., Kosaki, R., Niizuma, T., Hata, K., Kosaki, K. Macrothrombocytopenia and developmental delay with a de novo CDC42 mutation: yet another locus for thrombocytopenia and developmental delay. Am. J. Med. Genet. 167A: 2822-2825, 2015. [PubMed: 26386261, related citations] [Full Text]

  41. Takenouchi, T., Okamoto, N., Ida, S., Uehara, T., Kosaki, K. Further evidence of a mutation in CDC42 as a cause of a recognizable syndromic form of thrombocytopenia. Am. J. Med. Genet. 170A: 852-855, 2016. [PubMed: 26708094, related citations] [Full Text]

  42. Wells, C. D., Fawcett, J. P., Traweger, A., Yamanaka, Y., Goudreault, M., Elder, K., Kulkarni, S., Gish, G., Virag, C., Lim, C., Colwill, K., Starostine, A., Metalnikov, P., Pawson, T. A Rich1/Amot complex regulates the Cdc42 GTPase and apical-polarity proteins in epithelial cells. Cell 125: 535-548, 2006. [PubMed: 16678097, related citations] [Full Text]

  43. Wu, W. J., Erickson, J. W., Lin, R., Cerione, R. A. The gamma-subunit of the coatomer complex binds Cdc42 to mediate transformation. Nature 405: 800-804, 2000. [PubMed: 10866202, related citations] [Full Text]

  44. Wu, W. J., Tu, S., Cerione, R. A. Activated Cdc42 sequesters c-Cbl and prevents EGF receptor degradation. Cell 114: 715-725, 2003. [PubMed: 14505571, related citations] [Full Text]

  45. Wu, X., Quondamatteo, F., Lefever, T., Czuchra, A., Meyer, H., Chrostek, A., Paus, R., Langbein, L., Brakebusch, C. Cdc42 controls progenitor cell differentiation and beta-catenin turnover in skin. Genes Dev. 20: 571-585, 2006. [PubMed: 16510873, images, related citations] [Full Text]

  46. Yang, J., Zhang, Z., Roe, S. M., Marshall, C. J., Barford, D. Activation of Rho GTPases by DOCK exchange factors is mediated by a nucleotide sensor. Science 325: 1398-1402, 2009. [PubMed: 19745154, related citations] [Full Text]

  47. Yasuda, S., Oceguera-Yanez, F., Kato, T., Okamoto, M., Yonemura, S., Terada, Y., Ishizaki, T., Narumiya, S. Cdc42 and mDia3 regulate microtubule attachment to kinetochores. Nature 428: 767-771, 2004. [PubMed: 15085137, related citations] [Full Text]

  48. Zheng, Y., Fischer, D. J., Santos, M. F., Tigyi, G., Pasteris, N. G., Gorski, J. L., Xu, Y. The faciogenital dysplasia gene product FGD1 functions as a Cdc42Hs-specific guanine-nucleotide exchange factor. J. Biol. Chem. 271: 33169-33172, 1996. [PubMed: 8969170, related citations] [Full Text]


Ada Hamosh - updated : 09/18/2019
Cassandra L. Kniffin - updated : 04/04/2018
Ada Hamosh - updated : 02/21/2018
Patricia A. Hartz - updated : 3/8/2016
Ada Hamosh - updated : 1/8/2016
Ada Hamosh - updated : 6/24/2015
Ada Hamosh - updated : 12/9/2013
Ada Hamosh - updated : 5/6/2013
Paul J. Converse - updated : 10/26/2012
Matthew B. Gross - updated : 5/10/2011
Ada Hamosh - updated : 5/9/2011
Patricia A. Hartz - updated : 1/6/2011
Matthew B. Gross - updated : 5/11/2010
Ada Hamosh - updated : 1/8/2010
Ada Hamosh - updated : 10/13/2009
Ada Hamosh - updated : 2/18/2009
Patricia A. Hartz - updated : 4/28/2008
Patricia A. Hartz - updated : 8/30/2007
Patricia A. Hartz - updated : 1/19/2007
Patricia A. Hartz - updated : 5/3/2006
Patricia A. Hartz - updated : 3/28/2006
Patricia A. Hartz - updated : 10/7/2004
Ada Hamosh - updated : 9/28/2004
Ada Hamosh - updated : 4/16/2004
Cassandra L. Kniffin - updated : 3/5/2003
Ada Hamosh - updated : 1/29/2003
Patricia A. Hartz - updated : 6/5/2002
Stylianos E. Antonarakis - updated : 9/7/2000
Ada Hamosh - updated : 7/20/2000
Ada Hamosh - updated : 3/10/2000
Victor A. McKusick - updated : 8/23/1999
Jennifer P. Macke - updated : 4/8/1998
Jennifer P. Macke - updated : 5/28/1997
Alan F. Scott - updated : 3/6/1996
Creation Date:
Victor A. McKusick : 1/17/1991
alopez : 03/21/2023
carol : 08/04/2020
alopez : 09/18/2019
carol : 04/09/2018
carol : 04/06/2018
ckniffin : 04/04/2018
alopez : 02/21/2018
carol : 02/22/2017
carol : 08/08/2016
carol : 03/18/2016
carol : 3/9/2016
alopez : 3/8/2016
alopez : 1/13/2016
alopez : 1/8/2016
alopez : 1/8/2016
alopez : 6/24/2015
alopez : 12/9/2013
alopez : 5/6/2013
mgross : 11/20/2012
terry : 10/26/2012
mgross : 5/10/2011
alopez : 5/10/2011
terry : 5/9/2011
mgross : 1/24/2011
terry : 1/6/2011
wwang : 5/17/2010
mgross : 5/11/2010
mgross : 5/11/2010
alopez : 1/11/2010
alopez : 1/11/2010
terry : 1/8/2010
alopez : 10/22/2009
terry : 10/13/2009
carol : 7/7/2009
alopez : 2/20/2009
terry : 2/18/2009
mgross : 4/28/2008
ckniffin : 2/5/2008
mgross : 10/4/2007
terry : 8/30/2007
mgross : 1/19/2007
mgross : 6/7/2006
terry : 5/3/2006
wwang : 4/3/2006
terry : 3/28/2006
mgross : 10/7/2004
alopez : 10/4/2004
tkritzer : 9/28/2004
alopez : 4/19/2004
terry : 4/16/2004
cwells : 11/10/2003
tkritzer : 3/14/2003
ckniffin : 3/5/2003
alopez : 3/3/2003
alopez : 1/29/2003
terry : 1/29/2003
alopez : 11/19/2002
terry : 11/18/2002
carol : 6/5/2002
terry : 11/15/2001
mgross : 9/7/2000
alopez : 7/20/2000
alopez : 3/10/2000
mcapotos : 12/7/1999
psherman : 11/3/1999
psherman : 10/18/1999
jlewis : 9/3/1999
terry : 8/23/1999
kayiaros : 7/13/1999
psherman : 3/18/1999
psherman : 4/21/1998
dholmes : 4/8/1998
dholmes : 4/8/1998
alopez : 8/1/1997
alopez : 7/23/1997
alopez : 7/23/1997
mark : 12/16/1996
terry : 12/10/1996
terry : 4/17/1996
mark : 3/6/1996
carol : 4/1/1994
supermim : 3/16/1992
carol : 1/2/1992
carol : 3/4/1991
carol : 1/17/1991

* 116952

CELL DIVISION CYCLE 42; CDC42


Alternative titles; symbols

GTP-BINDING PROTEIN, 25-KD; G25K


HGNC Approved Gene Symbol: CDC42

SNOMEDCT: 1172685001;  


Cytogenetic location: 1p36.12     Genomic coordinates (GRCh38): 1:22,052,709-22,101,360 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
1p36.12 Takenouchi-Kosaki syndrome 616737 Autosomal dominant 3

TEXT

Description

CDC42 is a Ras (see 190020)-related GTP-binding protein. It is implicated in a variety of biologic activities, including establishment of cell polarity in yeast, regulation of cell morphology, motility, and cell cycle progression in mammalian cells, and induction of malignant transformation (summary by Wu et al., 2000).


Cloning and Expression

Shinjo et al. (1990) isolated cDNA clones that code for cdc42, a low molecular weight GTP-binding protein originally designated G(p) and also called G25K, from a human placenta library. The predicted amino acid sequence of the protein was very similar to those of various members of the RAS superfamily of low molecular weight GTP-binding proteins, including NRAS, KRAS, HRAS, and the RHO proteins. The highest degree of sequence identity (80%) was found with the Saccharomyces cerevisiae cell division cycle protein CDC42. The human placental gene complemented a cdc42 mutation in S. cerevisiae. Munemitsu et al. (1990) presented further evidence that G25K is the human homolog of the CDC42 gene product.

Marks and Kwiatkowski (1996) identified 2 isoforms of mouse Cdc42. They demonstrated that the 2 murine isoforms arise from a single gene by alternative splicing. Although one is expressed in a wide variety of tissues, the second isoform appeared to be expressed exclusively in brain.


Gene Function

Erickson et al. (1996) used cell fractionation and immunofluorescence to show that cdc42 is localized to the Golgi apparatus of mammalian cells. It colocalizes with the nonclathrin coat proteins ARF3 (103190) and COPB (600959) and its Golgi localization is disrupted by the drug brefeldin A. Based on their findings, Erickson et al. (1996) suggested that cdc42 may be involved in the delivery of newly synthesized proteins and lipids to the plasma membrane and that the GTP-binding/GTPase cycle may dictate its subcellular localization.

By screening rat brain cytosol for proteins that interacted with Ras-related GTPases, or p21 proteins, of the Rho (RHOA; 165390) subfamily, Manser et al. (1994) identified 3 proteins, designated PAKs (see PAK1; 602590) that interacted with the GTP-bound forms of human CDC42 and RAC1 (602048), but not RHOA. Brown et al. (1996) found that activity of human PAK1 was induced by coexpression with RAC1 or CDC42.

Zheng et al. (1996) reported that the FGD1 protein (305400) acts as a cdc42-specific GDP-GTP exchange factor. Cells expressing a fragment of the FGD1 protein encompassing the pleckstrin and Dbl homology domains activated 2 elements downstream of cdc42, namely, Jun kinase (165160) and p70 S6 kinase.

Manser et al. (1993) identified ACK1 (606994) as a binding partner and inhibitor of the GTP-bound form of CDC42. Interaction between GTP-CDC42 and ACK1 inhibited both the intrinsic and GAP-stimulated GTPase activity of CDC42.

CDC42 can regulate the actin cytoskeleton through activation of WASP family members (see 301000). Activation relieves an autoinhibitory contact between the GTPase-binding domain and the C-terminal region of WASP proteins. Kim et al. (2000) reported the autoinhibited structure of the GTPase-binding domain of WASP, which can be induced by the C-terminal region or by organic cosolvents. In the autoinhibited complex, intramolecular interactions with the GTPase-binding domain occlude residues of the C terminus that regulate the Arp2/3 actin-nucleating complex (see 604221). Binding of CDC42 to the GTPase-binding domain causes a dramatic conformational change, resulting in disruption of the hydrophobic core and release of the C terminus, enabling its interaction with the actin regulatory machinery.

Wu et al. (2000) identified a CDC42 mutant, Cdc42F28L, that binds GTP in the absence of a guanine nucleotide exchange factor, but still hydrolyzes GTP with a turnover number identical to that for wildtype CDC42. Expression of this mutant in fibroblasts causes cellular transformation, mimicking many of the characteristics of cells transformed by the DBL oncoprotein (311030), a known guanine nucleotide exchange factor for CDC42. Wu et al. (2000) searched for new CDC42 targets in an effort to understand how CDC42 mediates cellular transformation. They identified the gamma-subunit of the coatomer complex (gamma-COP; 604355) as a specific binding partner for activated CDC42. The binding of CDC42 to gamma-COP is essential for a transforming signal distinct from those elicited by Ras.

Dendritic cells (DCs) developmentally regulate antigen uptake by controlling their endocytic capacity. Immature DCs actively internalize antigen. Mature DCs, however, are poorly endocytic, functioning instead to present antigens to T cells. Garrett et al. (2000) found that endocytic downregulation reflects a decrease in endocytic activity controlled by RHO family GTPases, especially CDC42. Blocking CDC42 function by toxin B treatment or injection of dominant-negative inhibitors of CDC42 abrogated endocytosis in immature DCs. In mature DCs, injection of constitutively active CDC42 or microbial delivery of a CDC42 nucleotide exchange factor reactivated endocytosis. DCs regulated endogenous levels of CDC42-GTP with activated CDC42 detectable only in immature cells. Garrett et al. (2000) concluded that DCs developmentally regulate endocytosis at least in part by controlling levels of activated CDC42.

Using Madin Darby canine kidney (MDCK) cells expressing Cdc42 mutants defective in nucleotide binding or hydrolysis, Musch et al. (2001) showed that Cdc42 differentially regulated the exit of apical and basolateral proteins from the trans-Golgi network (TGN). GTPase-deficient Cdc42 accelerated the exit of an apical marker from the TGN and inhibited the release of basolateral proteins. Basolateral protein transport by Cdc42 with an activating mutation was accompanied by changes in the organization of the actin cytoskeleton.

Sin et al. (2002) used in vivo time-lapse imaging of optic tectal cells in Xenopus laevis tadpoles to demonstrate that enhanced visual activity driven by a light stimulus promotes dendritic arbor growth. The stimulus-induced dendritic arbor growth requires glutamate receptor (see 138249)-mediated synaptic transmission, decreased RhoA (165390) activity, and increased RAC and CDC42 activity. Sin et al. (2002) concluded that their results delineated a role for Rho GTPases in the structural plasticity driven by visual stimulation in vivo.

Morphologic changes in dendritic spines are believed to be caused by dynamic regulation of actin polymerization. Irie and Yamaguchi (2002) found that the EphB2 receptor tyrosine kinase (600997) physically associates with the guanine nucleotide exchange factor intersectin-1 (602442) in cooperation with the actin-regulating protein N-WASP (605056), which in turn activates Cdc42 and spine morphogenesis.

In higher eukaryotes, the small GTPase CDC42, acting through a PAR6-atypical protein kinase C (see PKC-zeta, 176982) complex, is required to establish cellular asymmetry during epithelial morphogenesis, asymmetric cell division, and directed cell migration. Etienne-Manneville and Hall (2003) used primary rat astrocytes in a cell migration assay to demonstrate that PAR6-PKC-zeta interacts directly with and regulates glycogen synthase kinase-3-beta (GSK3-beta; 605004) to promote polarization of the centrosome and to control the direction of cell protrusion. CDC42-dependent phosphorylation of GSK3-beta occurs specifically at the leading edge of migrating cells, and induces the interaction of APC (611731) protein with the plus ends of microtubules. The association of APC with microtubules is essential for cell polarization. Etienne-Manneville and Hall (2003) concluded that CDC42 regulates cell polarity through the spatial regulation of GSK3-beta and APC.

Wu et al. (2003) presented evidence that activation of CDC42 protects EGF receptor (EGFR; 131550) from the negative regulatory activity of CBL (165360), a ubiquitin ligase.

Yasuda et al. (2004) demonstrated that Cdc42 and mDia3 (DIAPH2; 300108) regulate microtubule attachment to kinetochores.

Nalbant et al. (2004) reported the development of a biosensor capable of visualizing the changing activation of endogenous unlabeled Cdc42 in living cells. With the use of a dye that reports protein interactions, the biosensor revealed localized activation in the trans-Golgi apparatus, microtubule-dependent Cdc42 activation at the cell periphery, and activation kinetics precisely coordinated with cell extension and retraction.

By electroporating genes into chicken presomitic mesenchymal cells, Nakaya et al. (2004) demonstrated that Cdc42 and Rac1 play different roles in mesenchymal-epithelial transition. Different levels of Cdc42 appeared to affect the binary decision between epithelial and mesenchymal states. Proper levels of Rac1 were also necessary for somitic epithelialization, since cells with either activated or inhibited Rac1 failed to undergo correct epithelialization.

Using functional and proteomic screens to identify regulators of Cdc42, Wells et al. (2006) identified a network of proteins that centered on Rich1 (ARHGAP17; 608293) and organized apical polarity in canine kidney epithelial cells. Rich1 bound the coiled-coil domain of Amot (300410) and was thereby targeted to a complex at tight junctions containing the PDZ domain-containing proteins Pals1 (MPP5; 606958), Patj (INADL; 603199), and Par3 (PARD3; 606745). Regulation of Cdc42 by Rich1 was required for maintenance of tight junctions. The coiled-coil domain of Amot was required for its localization to apical membranes and for Amot to relocalize Pals1 and Par3 to internal puncta. Wells et al. (2006) proposed that RICH1 and AMOT maintain tight junction integrity by coordinated regulation of CDC42 and by linking specific components of the tight junction to intracellular protein trafficking.

Formation of the apical surface and lumen is a fundamental step in epithelial organ development. Martin-Belmonte et al. (2007) showed that Pten (601728) localized to the apical plasma membrane during epithelial morphogenesis to mediate enrichment of phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) at this domain during cyst development in a 3-dimensional Madin-Darby canine kidney cell system. Ectopic PtdIns(4,5)P2 at the basolateral surface caused apical proteins to relocalize to the basolateral surface. Annexin-2 (ANX2; 151740) bound PtdIns(4,5)P2 and was recruited to the apical surface. Anx2 bound Cdc42 and recruited it to the apical surface, and Cdc42 in turn recruited the Par6 (607484)/atypical protein kinase C (aPKC; see 176982) complex to the apical surface. Loss of function of Pten, Anx2, Cdc42, or aPKC prevented normal development of the apical surface and lumen. Martin-Belmonte et al. (2007) concluded that PTEN, PtdIns(4,5)P2, ANX2, CDC42, and aPKC control apical plasma membrane and lumen formation.

Hamann et al. (2007) found that both ASEF1 and ASEF2 were guanine nucleotide exchange factors (GEFs) for CDC42, but not for RAC1 or RHOA. ASEF2 required the lipid-modified form of CDC42. Using deletion mutants, Hamann et al. (2007) showed that the tandem N-terminal ABR and SH3 domain (ABRSH3) of the ASEF proteins was required to bind the armadillo repeat region of APC. ABRSH3 also functioned in an autoinhibitory reaction by binding the C-terminal tails of ASEF1 and ASEF2 and inhibiting their GEF activities. Deletion of ABRSH3 or coexpression of the APC armadillo repeat sequence with full-length ASEF2 stimulated filopodia formation in transfected HeLa cells. Hamann et al. (2007) concluded that activation of ASEF1 and ASEF2 involves binding of APC to ABRSH3, which disrupts the autoinhibitory interaction of ABRSH3 with the ASEF C-terminal tail and allows GDP/GTP exchange on CDC42.

Kawasaki et al. (2007) found that ASEF2 was a GEF for both CDC42 and RAC1 in MDCK and HeLa cells. Overexpression of ASEF2 increased membrane ruffling and CDC42-mediated filopodia formation in HeLa cells.

Shen et al. (2008) showed that Nudel (NDEL1; 607538) colocalized with Cdc42gap (ARHGAP1; 602732) at the leading edge of migrating NIH3T3 mouse fibroblasts. This localization of Nudel required its phosphorylation by Erk1 (MAPK3; 601795)/Erk2 (MAPK1; 176948). Shen et al. (2008) found that Nudel competed with Cdc42 for binding Cdc42gap. Consequently, Nudel inhibited Cdc42gap-mediated inactivation of Cdc42 in a dose-dependent manner. Depletion of Nudel by RNA interference or overexpression of a nonphosphorylatable Nudel mutant abolished Cdc42 activation and cell migration. Shen et al. (2008) concluded that NUDEL facilitates cell migration by sequestering CDC42GAP at the leading edge to stabilize active CDC42 in response to extracellular stimuli.

Kang et al. (2008) performed a global characterization of rat neural palmitoyl proteomes and identified most of the known neural palmitoyl proteins, 68 in total, plus more than 200 novel palmitoyl protein candidates, with further testing confirming palmitoylation for 21 of these candidates. The novel palmitoyl proteins included neurotransmitter receptors, transporters, adhesion molecules, scaffolding proteins, as well as SNAREs and other vesicular trafficking proteins. Of particular interest was the finding of palmitoylation for a brain-specific Cdc42 splice variant. The palmitoylated Cdc42 isoform (Cdc42-palm) differs from the canonical, prenylated form (Cdc42-prenyl), with regard to both localization and function: Cdc42-palm concentrates in dendritic spines and has a special role in inducing these postsynaptic structures. Furthermore, assessing palmitoylation dynamics in drug-induced activity models identified rapidly induced changes for Cdc42 as well as for other synaptic palmitoyl proteins, suggesting that palmitoylation may participate broadly in the activity-driven changes that shape synapse morphology and brain function.

Machacek et al. (2009) examined GTPase coordination in mouse embryonic fibroblasts both through simultaneous visualization of 2 GTPase biosensors and using a 'computational multiplexing' approach capable of defining the relationships between multiple protein activities visualized in separate experiments. They found that RhoA (165390) is activated at the cell edge synchronous with edge advancement, whereas Cdc42 and Rac1 are activated 2 microns behind the edge with a delay of 40 seconds. This indicates that Rac1 and RhoA operate antagonistically through spatial separation and precise timing, and that RhoA has a role in the initial events of protrusion, whereas Rac1 and Cdc42 activate pathways implicated in reinforcement and stabilization of newly expanded protrusions.

Frank et al. (2009) found that Cdc42 was involved in a signaling complex that modulated presynaptic Cav2.1 (CACNA1A; 601011) calcium channels in the Drosophila neuromuscular junction. This signaling system involved the Rho-type GEF ephexin (NGEF; 605991), which appeared to act with Cdc42 to activate the Eph receptor (see EPHA1; 179610) for modulation of Cav2.1 channel activity.

Murakoshi et al. (2011) used 2-photon fluorescence lifetime imaging microscopy to monitor the activity of 2 Rho GTPases, RhoA and Cdc42, in single dendritic spines undergoing structural plasticity associated with long-term potentiation in CA1 pyramidal neurons in cultured slices of rat hippocampus. When long-term volume increase was induced in a single spine using 2-photon glutamate uncaging, RhoA and Cdc42 were rapidly activated in the stimulated spine. These activities decayed over about 5 minutes, and were then followed by a phase of persistent activation lasting more than half an hour. Although active RhoA and Cdc42 were similarly mobile, their activity patterns were different. RhoA activation diffused out of the stimulated spine and spread over about 5 microns along the dendrite. In contrast, Cdc42 activation was restricted to the stimulated spine, and exhibited a steep gradient at the spine necks. Inhibition of the Rho-Rock pathway preferentially inhibited the initial spine growth, whereas the inhibition of the Cdc42-Pak pathway blocked the maintenance of sustained structural plasticity. RhoA and Cdc42 activation depended on calcium ion/calmodulin-dependent kinase (CaMKII). Thus, Murakoshi et al. (2011) concluded that RhoA and Cdc42 relay transient CaMKII activation to synapse-specific, long-term signaling required for spine structural plasticity.

Using synthetic derivatives of the enteropathogenic Escherichia coli guanine-nucleotide exchange factor Map, Orchard et al. (2012) found that CDC42 GTPase signal transduction was controlled by interaction between Map and the PDZ domains of EBP50 (SLC9A3R1; 604990) and the induction of clusters of actin-rich membrane protrusions.

Keestra et al. (2013) demonstrated that NOD1 (605980) senses cytosolic microbial products by monitoring the activation state of small Rho GTPases. Activation of RAC1 (602048) and CDC42 by bacterial delivery or ectopic expression of SopE, a virulence factor of the enteric pathogen Salmonella, triggered the NOD1 signaling pathway with consequent RIP2 (603455)-mediated induction of NF-kappa-B (see 164011)-dependent inflammatory responses. Similarly, activation of the NOD1 signaling pathway by peptidoglycan required RAC1 activity. Furthermore, Keestra et al. (2013) showed that constitutively active forms of RAC1, CDC42, and RHOA (165390) activated the NOD1 signaling pathway.

Florian et al. (2013) reported an unexpected shift from canonical to noncanonical Wnt signaling in mice due to elevated expression of Wnt5a (164975) in aged hematopoietic stem cells (HSCs), which causes stem cell aging. Wnt5a treatment of young HSCs induced aging-associated stem cell apolarity, reduction of regenerative capacity, and an aging-like myeloid-lymphoid differentiation skewing via activation of the small Rho GTPase Cdc42. Conversely, Wnt5a haploinsufficiency attenuated HSC aging, whereas stem cell-intrinsic reduction of Wnt5a expression resulted in functionally rejuvenated aged HSCs. Florian et al. (2013) concluded that the data demonstrated a critical role for stem cell-intrinsic noncanonical Wnt5a signaling in HSC aging.

Park et al. (2015) showed that the coat protein I complex (COPI; see 601924) sorts anterograde cargoes into tubules that connect the Golgi cisternae in human cells. Moreover, the small GTPase CDC42 regulates bidirectional Golgi transport by targeting the dual functions of COPI in cargo sorting and carrier formation. CDC42 also directly imparts membrane curvature to promote COPI tubule formation. Park et al. (2015) concluded that their findings further revealed that COPI tubular transport complements cisternal maturation in explaining how anterograde Golgi transport is achieved, and that bidirectional COPI transport is modulated by environmental cues through CDC42.

By transfecting MDCK canine kidney cell with wildtype or mutant human constructs, and by knockdown of endogenous MDCK proteins, Hayase et al. (2013) studied development of apical-basolateral polarity. Their results suggested that cytoplasmic MORG1 (WDR83; 616850) interacted directly with PAR6 (607484) and mediated apical translocation of a dimer made up of PAR6 and an atypical PKC (aPKC, e.g., PRKCI, 600539). MORG1 also interacted with the apical transmembrane protein CRB3 (609737) and facilitated binding between PAR6 and CRB3, thereby anchoring the PAR6-aPKC dimer to the apical membrane. The small GTPase CDC42 displaced MORG1 from the complex at the apical membrane and strengthened the association between PAR6 and CRB3. Knockdown of any complex component interfered with development of polarity in MDCK cells; however, overexpression of a CRB3-aPKC construct reversed the polarity defect in MORG1-deficient MDCK cells and restored apical identity.

In female meiosis, selfish elements drive by preferentially attaching to the egg side of the spindle. This implies some asymmetry between the 2 sides of the spindle. Akera et al. (2017) found that CDC42 signaling from the cell cortex regulated microtubule tyrosination to induce spindle asymmetry and that non-Mendelian segregation depended on this asymmetry. Cortical CDC42 depends on polarization directed by chromosomes, which are positioned near the cortex to allow the asymmetric cell division. Thus, Akera et al. (2017) selfish meiotic drivers exploit the asymmetry inherent in female meiosis to bias their transmission.

Hedrick et al. (2016) described a 3-molecule model of structural long-term potentiation of murine dendritic spines, implicating the localized, coincident activation of Rac1, RhoA, and Cdc42 as a causal signal of structural long-term potentiation. This model posited that complete tripartite signal overlap in spines confers structural long-term potentiation, but that partial overlap primes spines for structural plasticity. By monitoring the spatiotemporal activation patterns of these GTPases during structural long-term potentiation, Hedrick et al. (2016) found that such spatiotemporal signal complementation simultaneously explains 3 integral features of plasticity: the facilitation of plasticity by BDNF (113505), the postsynaptic source of which activates Cdc42 and Rac1, but not RhoA; heterosynaptic facilitation of structural long-term potentiation, which is conveyed by diffusive Rac1 and RhoA activity; and input specificity, which is afforded by spine-restricted Cdc42 activity.


Gene Structure

Nicole et al. (1999) determined the organization of the CDC42 gene and found that the gene encodes the placental and brain isoforms generated by alternative splicing.

Moats-Staats and Stiles (1995) showed that the 5-prime end of another gene, called BB1 by them, overlaps the 3-prime end of G25K.


Biochemical Features

Crystal Structure

Through a structural analysis of DOCK9 (607325)-CDC42 complexes, Yang et al. (2009) identified a nucleotide sensor within the alpha-10 helix of the DHR2 domain that contributes to release of guanine diphosphate (GDP) and then to discharge of the activated GTP-bound Cdc42. Magnesium exclusion, a critical factor in promoting GDP release, is mediated by a conserved valine residue within this sensor, whereas binding of GTP-magnesium ion to the nucleotide-free complex results in magnesium-inducing displacement of the sensor to stimulate discharge of Cdc42-GTP. Yang et al. (2009) concluded that their studies identified an unusual mechanism of GDP release and defined the complete guanine nucleotide exchange factor catalytic cycle from GDP dissociation followed by GTP binding and discharge of the activated GTPase.


Mapping

Using SSCP analysis of a mouse backcross panel, Marks and Kwiatkowski (1996) demonstrated that the gene encoding cdc42 is localized to the distal portion of mouse chromosome 4 between Ephb2 proximally and Cappb (601572) distally. The human homologs of both of the 2 flanking genes were mapped to human chromosome 1p36.1 by Barron-Casella et al. (1995), thus indicating that this is the likely site of the human CDC42 gene. The CDC42 gene was mapped to the 1p36-p35 region by radiation hybrid analysis (Schuler et al., 1996; Jensen et al., 1997; Deloukas et al., 1998).

Nicole et al. (1999) demonstrated a CDC42-like transcript on chromosome 4 that does not contain introns and is similar to the placental isoform, suggesting that it is a processed pseudogene.

Nicole et al. (1999) excluded the CDC42 gene as the site of mutation in the Schwartz-Jampel syndrome type 1 (255800).


Molecular Genetics

Takenouchi et al. (2015) and Takenouchi et al. (2016) each reported a patient with macrothrombocytopenia, lymphedema, developmental delay, and similar distinctive facial features, known as Takenouchi-Kosaki syndrome (TKS; 616737). Both patients were heterozygous for the same de novo mutation in the CDC42 gene (116952.0001).

In 15 patients from 13 unrelated families with a heterogeneous developmental disorder consistent with TKS, Martinelli et al. (2018) identified 9 different heterozygous missense mutations in the CDC42 gene (see, e.g., 116952.0001-116952.0006). The mutations occurred de novo in 12 unrelated patients; there was 1 family (family 30153) in which 3 affected individuals carried the same mutation. The mutations in most patients were identified by whole-exome sequencing; some mutations were identified by direct sequencing of the CDC42 gene. None of the mutations were found in the ExAC/gnomAD database, and all were predicted to be pathogenic according to ACMG criteria. Based on molecular modeling, predicted functional impact, and in vitro functional studies, the mutations were categorized into 3 main groups, all of which were determined to be pathogenic (see GENOTYPE/PHENOTYPE CORRELATIONS).


Genotype/Phenotype Correlations

Based on molecular modeling, predicted functional impact, and in vitro functional studies, the mutations identified by Martinelli et al. (2018) were categorized into 3 main groups. Group I mutations (Y64C; R66G, 116952.0002; and R68Q) occurred in the switch II domain, which mediates CDC42 binding to effectors and regulators, and were predicted to interfere with the catalytic activity of the GTPase and/or its capability to transduce signaling. Group I mutations were associated with a syndromic form of thrombocytopenia. Group II mutations (C81F, 116952.0003; S83P, 116952.0004; and A159V) were located within or close to the nucleotide-binding pocket, and were predicted to promote fast GDP/GTP cycling, favoring a hyperactive GTP-bound state. Group II mutations were associated with a variable developmental disorder characterized by striking dysmorphic features resembling a RASopathy. Group III mutations (I21T, 116952.0005; Y23C; and E171K, 116952.0006), located in residues predicted to disrupt interactions with effectors containing a CRIB (Cdc42, Rac interactive binding) motif, were associated with a milder phenotype resembling Noonan syndrome. In vitro studies of selected mutations using recombinant proteins showed variable effects on CDC42 function, including altering the switch between the active and inactive states of the GTPase and/or affecting CDC42 interaction with effectors. Group I mutations had defective interaction with tested partner proteins, group II mutations showed variable hyperactive behavior, and group III mutations showed perturbed binding to effectors. In addition, a wound healing assay indicated that the mutations resulted in dysregulated and disturbed cell polarization and proliferation. One specific mutation (E171K), which affects only 1 of the 2 CDC42 isoforms and specifically impaired binding to WASP (300392), resulted in an overall milder clinical phenotype that phenocopied Noonan syndrome. Studies in C. elegans showed that the mutations caused variable disruption of developmental processes, with some mutations acting as a gain of function and others acting as hypomorphs. In general, the group II mutations upregulated multiple signaling pathways. Overall, the findings indicated that CDC42 functions in a large array of developmental processes.


Animal Model

Wu et al. (2006) stated that constitutive knockout of Cdc42 in mice results in death around implantation. In order to examine the role of Cdc42 in the differentiation of skin stem cells into hair follicles, they targeted Cdc42 deletion to keratinocytes. Mutant mice were born without obvious defects but showed impaired hair formation and growth retardation. Within 4 weeks, all hairs were lost and did not grow again in older animals. In the absence of Cdc42, degradation of beta-catenin (CTNNB1; 116806) increased corresponding to decreased phosphorylation of Gsk3-beta and increased phosphorylation of axin (603816), which is required for binding of beta-catenin to the degradation machinery. Wu et al. (2006) concluded that Cdc42 regulation of beta-catenin turnover is required for terminal differentiation of hair follicle progenitor cells.

By targeted deletion of Cdc42 in telencephalic neural progenitors in mouse embryos, Chen et al. (2006) found that Cdc42 was essential for establishment of apical-basal polarity of the telencephalic neuroepithelium, a necessity for expansion and bifurcation of cerebral hemispheres.

Pleines et al. (2010) found that conditional knockout of Cdc42 in mice results in mild thrombocytopenia and increased platelet size (i.e., macrothrombocytopenia).


ALLELIC VARIANTS 6 Selected Examples):

.0001   TAKENOUCHI-KOSAKI SYNDROME

CDC42, TYR64CYS ({dbSNP SCV000577577.2})
SNP: rs864309721, ClinVar: RCV000203307, RCV000489915, RCV001266021, RCV001291421

In 2 unrelated females, one of Japanese-Iranian descent and the other of Japanese descent, with a syndrome of macrothrombocytopenia, developmental delay, and distinctive facial features (TKS; 616737), Takenouchi et al. (2015) and Takenouchi et al. (2016) identified a heterozygous c.191A-G transition (c.191A-G, NM_001039802) in exon 3 of the CDC42 gene, resulting in a tyrosine-to-cystine substitution at codon 64 (Y64C). The mutation was confirmed in both patients by Sanger sequencing and was not identified in either parent.

In a 15-year-old girl (patient LR17-420) with TKS, Martinelli et al. (2018) identified a de novo heterozygous Y64C mutation in the CDC42 gene. The mutation, which was found by whole-exome sequencing, was not found in the ExAC/gnomAD database and met the ACMG criteria for pathogenicity.


.0002   TAKENOUCHI-KOSAKI SYNDROME

CDC42, ARG66GLY ({dbSNP SCV000244118.3})
SNP: rs797044870, ClinVar: RCV000190678, RCV000601199, RCV001291422

In 2 unrelated patients (LR14-352 and PCGC 1-04248) with Takenouchi-Kosaki syndrome (TKS; 616737), Martinelli et al. (2018) identified a de novo heterozygous c.196A-G transition (c.196A-G, NM_001791.3) in exon 3 of the CDC42 gene, resulting in an arg66-to-gly (R66G) substitution at a conserved residue in the switch II domain. The mutation, which was found by whole-exome sequencing, was not found in the ExAC/gnomAD database and met the ACMG criteria for pathogenicity.


.0003   TAKENOUCHI-KOSAKI SYNDROME

CDC42, CYS81PHE ({dbSNP SCV000589746.1})
SNP: rs1553196100, ClinVar: RCV000497401, RCV000604971, RCV001291424

In a 4-year-old boy (LR17-032), born of unrelated parents, with Takenouchi-Kosaki syndrome (TKS; 616737), Martinelli et al. (2018) identified a de novo heterozygous c.242G-T transversion (c.242G-T, NM_001791.3) in exon 3 of the CDC42 gene, resulting in a cys81-to-phe (C81F) substitution at a conserved residue in the beta-4 domain. The mutation, which was found by whole-exome sequencing, was not found in the ExAC/gnomAD database and met the ACMG criteria for pathogenicity.


.0004   TAKENOUCHI-KOSAKI SYNDROME

CDC42, SER83PRO ({dbSNP SCV000678255})
SNP: rs1553196101, ClinVar: RCV000577866, RCV000611080, RCV001291425

In a boy (LR10-046), conceived by in vitro fertilization, with Takenouchi-Kosaki syndrome (TKS; 616737), Martinelli et al. (2018) identified a de novo heterozygous c.247T-C transition (c.247T-C, NM_001791.3) in exon 3 of the CDC42 gene, resulting in a ser83-to-pro (S83P) substitution in the beta-4 domain. The mutation, which was found by whole-exome sequencing, was not found in the ExAC/gnomAD database and met the ACMG criteria for pathogenicity.


.0005   TAKENOUCHI-KOSAKI SYNDROME

CDC42, ILE21THR ({dbSNP SCV000572034.2})
SNP: rs1064795845, ClinVar: RCV000481008, RCV000601771, RCV001291419

In a patient (LR16-483) with Takenouchi-Kosaki syndrome (TKS; 616737), Martinelli et al. (2018) identified a de novo heterozygous c.62T-C transition (c.62T-C, NM_001791.3) in exon 1 of the CDC42 gene, resulting in an ile21-to-thr (I21T) substitution at a conserved residue in the alpha-1 domain. The mutation, which was found by whole-exome sequencing, was not found in the ExAC/gnomAD database and met the ACMG criteria for pathogenicity.


.0006   TAKENOUCHI-KOSAKI SYNDROME

CDC42, GLU171LYS ({dbSNP SCV000678257})
SNP: rs1553196539, ClinVar: RCV000577848, RCV000607619, RCV001291162

In 3 members of a family (family 30153) with Takenouchi-Kosaki syndrome (TKS; 616737), Martinelli et al. (2018) identified a heterozygous c.511G-A transition (c.511G-A, NM_001791.3) in exon 5 of the CDC42 gene, resulting in a glu171-to-lys (E171K) substitution at a conserved residue in the CBR domain. An unrelated patient (M060721) with a similar disorder carried this mutation in the de novo state. The mutation only affected transcript variant 1 and isoform 1 of CDC42. The mutation was not found in the ExAC/gnomAD database and met the ACMG criteria for pathogenicity.


REFERENCES

  1. Akera, T., Chmatal, L., Trimm, E., Yang, K., Aonbangkhen, C., Chenoweth, D. M., Janke, C., Schultz, R. M., Lampson, M. A. Spindle asymmetry drives non-Mendelian chromosome segregation. Science 358: 668-672, 2017. [PubMed: 29097549] [Full Text: https://doi.org/10.1126/science.aan0092]

  2. Barron-Casella, E. A., Torres, M. A., Scherer, S. W., Heng, H. H., Tsui, L. C., Casella, J. F. Sequence analysis and chromosomal localization of human Cap Z: conserved residues within the actin-binding domain may link Cap Z to gelsolin/severin and profilin protein families. J. Biol. Chem. 270: 21472-21479, 1995. [PubMed: 7665558] [Full Text: https://doi.org/10.1074/jbc.270.37.21472]

  3. Brown, J. L., Stowers, L., Baer, M., Trejo, J., Coughlin, S., Chant, J. Human Ste20 homologue hPAK1 links GTPases to the JNK MAP kinase pathway. Curr. Biol. 6: 598-605, 1996. [PubMed: 8805275] [Full Text: https://doi.org/10.1016/s0960-9822(02)00546-8]

  4. Chen, L., Liao, G., Yang, L., Campbell, K., Nakafuku, M., Kuan, C.-Y., Zheng, Y. Cdc42 deficiency causes Sonic hedgehog-independent holoprosencephaly. Proc. Nat. Acad. Sci. 103: 16520-16525, 2006. [PubMed: 17050694] [Full Text: https://doi.org/10.1073/pnas.0603533103]

  5. Deloukas, P., Schuler, G. D., Gyapay, G., Beasley, E. M., Soderlund, C., Rodriguez-Tome, P., Hui, L., Matise, T. C., McKusick, K. B., Beckmann, J. S., Bentolila, S., Bihoreau, M.-T., and 53 others. A physical map of 30,000 human genes. Science 282: 744-746, 1998. [PubMed: 9784132] [Full Text: https://doi.org/10.1126/science.282.5389.744]

  6. Erickson, J. W., Zhang, C., Kahn, R. A., Evans, T., Cerione, R. A. Mammalian cdc42 is a brefeldin A-sensitive component of the Golgi apparatus. J. Biol. Chem. 271: 26850-26854, 1996. [PubMed: 8900167] [Full Text: https://doi.org/10.1074/jbc.271.43.26850]

  7. Etienne-Manneville, S., Hall, A. Cdc42 regulates GSK-3-beta and adenomatous polyposis coli to control cell polarity. Nature 421: 753-756, 2003. [PubMed: 12610628] [Full Text: https://doi.org/10.1038/nature01423]

  8. Florian, M. C., Nattamai, K. J., Dorr, K., Marka, G., Uberle, B., Vas, V., Eckl, C., Andra, I., Schiemann, M., Oostendorp, R. A. J., Scharffetter-Kochanek, K., Kestler, H. A., Zheng, Y., Geiger, H. A canonical to non-canonical Wnt signalling switch in haematopoietic stem-cell ageing. Nature 503: 392-396, 2013. [PubMed: 24141946] [Full Text: https://doi.org/10.1038/nature12631]

  9. Frank, C. A., Pielage, J., Davis, G. W. A presynaptic homeostatic signaling system composed of the Eph receptor, Ephexin, Cdc42, and Cav2.1 calcium channels. Neuron 61: 556-569, 2009. [PubMed: 19249276] [Full Text: https://doi.org/10.1016/j.neuron.2008.12.028]

  10. Garrett, W. S., Chen, L.-M., Kroschewski, R., Ebersold, M., Turley, S., Trombetta, S., Galan, J. E., Mellman, I. Developmental control of endocytosis in dendritic cells by Cdc42. Cell 102: 325-334, 2000. [PubMed: 10975523] [Full Text: https://doi.org/10.1016/s0092-8674(00)00038-6]

  11. Hamann, M. J., Lubking, C. M., Luchini, D. N., Billadeau, D. D. Asef2 functions as a Cdc42 exchange factor and is stimulated by the release of an autoinhibitory module from a concealed C-terminal activation element. Molec. Cell. Biol. 27: 1380-1393, 2007. [PubMed: 17145773] [Full Text: https://doi.org/10.1128/MCB.01608-06]

  12. Hayase, J., Kamakura, S., Iwakiri, Y., Yamaguchi, Y., Izaki, T., Ito, T., Sumimoto, H. The WD40 protein Morg1 facilitates Par6-aPKC binding to Crb3 for apical identity in epithelial cells. J. Cell Biol. 200: 635-650, 2013. [PubMed: 23439680] [Full Text: https://doi.org/10.1083/jcb.201208150]

  13. Hedrick, N. G., Harward, S. C., Hall, C. E., Murakoshi, H., McNamara, J. O., Yasuda, R. Rho GTPase complementation underlies BDNF-dependent homo- and heterosynaptic plasticity. Nature 538: 104-108, 2016. [PubMed: 27680697] [Full Text: https://doi.org/10.1038/nature19784]

  14. Irie, F., Yamaguchi, Y. EphB receptors regulate dendritic spine development via intersectin, Cdc42 and N-WASP. Nature Neurosci. 5: 1117-1118, 2002. [PubMed: 12389031] [Full Text: https://doi.org/10.1038/nn964]

  15. Jensen, S. J., Sulman, E. P., Maris, J. M., Matise, T. C., Vojta, P. J., Barrett, J. C., Brodeur, G. M., White, P. S. An integrated transcript map of human chromosome 1p35-p36. Genomics 42: 126-136, 1997. [PubMed: 9177782] [Full Text: https://doi.org/10.1006/geno.1997.4714]

  16. Kang, R., Wan, J., Arstikaitis, P., Takahashi, H., Huang, K., Bailey, A. O., Thompson, J. X., Roth, A. F., Drisdel, R. C., Mastro, R., Green, W. N., Yates, J. R., III, Davis, N. G., El-Husseini, A. Neural palmitoyl-proteomics reveals dynamic synaptic palmitoylation. Nature 456: 904-909, 2008. [PubMed: 19092927] [Full Text: https://doi.org/10.1038/nature07605]

  17. Kawasaki, Y., Sagara, M., Shibata, Y., Shirouzu, M., Yokoyama, S., Akiyama, T. Identification and characterization of Asef2, a guanine-nucleotide exchange factor specific for Rac1 and Cdc42. Oncogene 26: 7620-7627, 2007. [PubMed: 17599059] [Full Text: https://doi.org/10.1038/sj.onc.1210574]

  18. Keestra, A. M., Winter, M. G., Auburger, J. J., Frassle, S. P., Xavier, M. N., Winter, S. E., Kim, A., Poon, V., Ravesloot, M. M., Waldenmaier, J. F. T., Tsolis, R. M., Eigenheer, R. A., Baumler, A. J. Manipulation of small Rho GTPases is a pathogen-induced process detected by NOD1. Nature 496: 233-237, 2013. [PubMed: 23542589] [Full Text: https://doi.org/10.1038/nature12025]

  19. Kim, A. S., Kakalis, L. T., Abdul-Manan, N., Liu, G. A., Rosen, M. K. Autoinhibition and activation mechanisms of the Wiskott-Aldrich syndrome protein. Nature 404: 151-158, 2000. [PubMed: 10724160] [Full Text: https://doi.org/10.1038/35004513]

  20. Machacek, M., Hodgson, L., Welch, C., Elliott, H., Pertz, O., Nalbant, P., Abell, A., Johnson, G. L., Hahn, K. M., Danuser, G. Coordination of Rho GTPase activities during cell protrusion. Nature 461: 99-103, 2009. [PubMed: 19693013] [Full Text: https://doi.org/10.1038/nature08242]

  21. Manser, E., Leung, T., Salihuddin, H., Tan, L., Lim, L. A non-receptor tyrosine kinase that inhibits the GTPase activity of p21cdc42. Nature 363: 364-367, 1993. [PubMed: 8497321] [Full Text: https://doi.org/10.1038/363364a0]

  22. Manser, E., Leung, T., Salihuddin, H., Zhao, Z., Lim, L. A brain serine/threonine protein kinase activated by Cdc42 and Rac1. Nature 367: 40-46, 1994. [PubMed: 8107774] [Full Text: https://doi.org/10.1038/367040a0]

  23. Marks, P. W., Kwiatkowski, D. J. Genomic organization and chromosomal location of murine Cdc42. Genomics 38: 13-18, 1996. [PubMed: 8954774] [Full Text: https://doi.org/10.1006/geno.1996.0586]

  24. Martin-Belmonte, F., Gassama, A., Datta, A., Yu, W., Rescher, U., Gerke, V., Mostov, K. PTEN-mediated apical segregation of phosphoinositides controls epithelial morphogenesis through Cdc42. Cell 128: 383-397, 2007. [PubMed: 17254974] [Full Text: https://doi.org/10.1016/j.cell.2006.11.051]

  25. Martinelli, S., Krumbach, O. H. F., Pantaleoni, F., Coppola, S., Amin, E., Pannone, L., Nouri, K., Farina, L., Dvorsky, R., Lepri, F., Buchholzer, M., Konopatzki, R, and 37 others. Functional dysregulation of CDC42 causes diverse developmental phenotypes. Am. J. Hum. Genet. 102: 309-320, 2018. [PubMed: 29394990] [Full Text: https://doi.org/10.1016/j.ajhg.2017.12.015]

  26. Moats-Staats, B. M., Stiles, A. D. Southern hybridization analyses of somatic cell hybrids reveal that human BB1 is a member of a multigene family dispersed throughout the human genome and appears to be linked to the human G25K genes. DNA Cell Biol. 14: 465-474, 1995. [PubMed: 7598801] [Full Text: https://doi.org/10.1089/dna.1995.14.465]

  27. Munemitsu, S., Innis, M. A., Clark, R., McCormick, F., Ullrich, A., Polakis, P. Molecular cloning and expression of a G25K cDNA, the human homolog of the yeast cell cycle gene CDC42. Molec. Cell. Biol. 10: 5977-5982, 1990. [PubMed: 2122236] [Full Text: https://doi.org/10.1128/mcb.10.11.5977-5982.1990]

  28. Murakoshi, H., Wang, H., Yasuda, R. Local, persistent activation of Rho GTPases during plasticity of single dendritic spines. Nature 472: 100-104, 2011. [PubMed: 21423166] [Full Text: https://doi.org/10.1038/nature09823]

  29. Musch, A., Cohen, D., Kreitzer, G., Rodriguez-Boulan, E. cdc42 regulates the exit of apical and basolateral proteins from the trans-Golgi network. EMBO J. 20: 2171-2179, 2001. [PubMed: 11331583] [Full Text: https://doi.org/10.1093/emboj/20.9.2171]

  30. Nakaya, Y., Kuroda, S., Katagiri, Y. T., Kaibuchi, K., Takahashi, Y. Mesenchymal-epithelial transition during somitic segmentation is regulated by differential roles of Cdc42 and Rac1. Dev. Cell 7: 425-438, 2004. [PubMed: 15363416] [Full Text: https://doi.org/10.1016/j.devcel.2004.08.003]

  31. Nalbant, P., Hodgson, L., Kraynov, V., Toutchkine, A., Hahn, K. M. Activation of endogenous Cdc42 visualized in living cells. Science 305: 1615-1619, 2004. [PubMed: 15361624] [Full Text: https://doi.org/10.1126/science.1100367]

  32. Nicole, S., White, P. S., Topaloglu, H., Beigthon, P., Salih, M., Hentati, F., Fontaine, B. The human CDC42 gene: genomic organization, evidence for the existence of a putative pseudogene and exclusion as a SJS1 candidate gene. Hum. Genet. 105: 98-103, 1999. [PubMed: 10480361] [Full Text: https://doi.org/10.1007/s004399900065]

  33. Orchard, R. C., Kittisopikul, M., Altschuler, S. J., Wu, L. F., Suel, G. M., Alto, N. M. Identification of F-actin as the dynamic hub in a microbial-induced GTPase polarity circuit. Cell 148: 803-815, 2012. [PubMed: 22341450] [Full Text: https://doi.org/10.1016/j.cell.2011.11.063]

  34. Park, S.-Y., Yang, J.-S., Schmider, A. B., Soberman, R. J., Hsu, V. W. Coordinated regulation of bidirectional COPI transport at the Golgi by CDC42. Nature 521: 529-532, 2015. [PubMed: 25945738] [Full Text: https://doi.org/10.1038/nature14457]

  35. Pleines, I., Eckly, A., Elvers, M., Hagedorn, I., Eliautou, S., Bender, M., Wu, X., Lanza, F., Gachet, C., Brakebusch, C., Nieswandt, B. Multiple alterations of platelet functions dominated by increased secretion in mice lacking Cdc42 in platelets. Blood 115: 3364-3373, 2010. [PubMed: 20139097] [Full Text: https://doi.org/10.1182/blood-2009-09-242271]

  36. Schuler, G. D., Boguski, M. S., Stewart, E. A., Stein, L. D., Gyapay, G., Rice, K., White, R. E., Rodriguez-Tome, P., Aggarwal, A., Bajorek, E., Bentolila, S., Birren, B. B., and 92 others. A gene map of the human genome. Science 274: 540-546, 1996. [PubMed: 8849440]

  37. Shen, Y., Li, N., Wu, S., Zhou, Y., Shan, Y., Zhang, Q., Ding, C., Yuan, Q., Zhao, F., Zeng, R., Zhu, X. Nudel binds Cdc42GAP to modulate Cdc42 activity at the leading edge of migrating cells. Dev. Cell 14: 342-353, 2008. [PubMed: 18331715] [Full Text: https://doi.org/10.1016/j.devcel.2008.01.001]

  38. Shinjo, K., Koland, J. G., Hart, M. J., Narasimhan, V., Johnson, D. I., Evans, T., Cerione, R. A. Molecular cloning of the gene for the human placental GTP-binding protein G(p) (G25K): identification of this GTP-binding protein as the human homolog of the yeast cell-division-cycle protein CDC42. Proc. Nat. Acad. Sci. 87: 9853-9857, 1990. [PubMed: 2124704] [Full Text: https://doi.org/10.1073/pnas.87.24.9853]

  39. Sin, W. C., Haas, K., Ruthazer, E. S., Cline, H. T. Dendrite growth increased by visual activity requires NMDA receptor and Rho GTPases. Nature 419: 475-480, 2002. [PubMed: 12368855] [Full Text: https://doi.org/10.1038/nature00987]

  40. Takenouchi, T., Kosaki, R., Niizuma, T., Hata, K., Kosaki, K. Macrothrombocytopenia and developmental delay with a de novo CDC42 mutation: yet another locus for thrombocytopenia and developmental delay. Am. J. Med. Genet. 167A: 2822-2825, 2015. [PubMed: 26386261] [Full Text: https://doi.org/10.1002/ajmg.a.37275]

  41. Takenouchi, T., Okamoto, N., Ida, S., Uehara, T., Kosaki, K. Further evidence of a mutation in CDC42 as a cause of a recognizable syndromic form of thrombocytopenia. Am. J. Med. Genet. 170A: 852-855, 2016. [PubMed: 26708094] [Full Text: https://doi.org/10.1002/ajmg.a.37526]

  42. Wells, C. D., Fawcett, J. P., Traweger, A., Yamanaka, Y., Goudreault, M., Elder, K., Kulkarni, S., Gish, G., Virag, C., Lim, C., Colwill, K., Starostine, A., Metalnikov, P., Pawson, T. A Rich1/Amot complex regulates the Cdc42 GTPase and apical-polarity proteins in epithelial cells. Cell 125: 535-548, 2006. [PubMed: 16678097] [Full Text: https://doi.org/10.1016/j.cell.2006.02.045]

  43. Wu, W. J., Erickson, J. W., Lin, R., Cerione, R. A. The gamma-subunit of the coatomer complex binds Cdc42 to mediate transformation. Nature 405: 800-804, 2000. [PubMed: 10866202] [Full Text: https://doi.org/10.1038/35015585]

  44. Wu, W. J., Tu, S., Cerione, R. A. Activated Cdc42 sequesters c-Cbl and prevents EGF receptor degradation. Cell 114: 715-725, 2003. [PubMed: 14505571] [Full Text: https://doi.org/10.1016/s0092-8674(03)00688-3]

  45. Wu, X., Quondamatteo, F., Lefever, T., Czuchra, A., Meyer, H., Chrostek, A., Paus, R., Langbein, L., Brakebusch, C. Cdc42 controls progenitor cell differentiation and beta-catenin turnover in skin. Genes Dev. 20: 571-585, 2006. [PubMed: 16510873] [Full Text: https://doi.org/10.1101/gad.361406]

  46. Yang, J., Zhang, Z., Roe, S. M., Marshall, C. J., Barford, D. Activation of Rho GTPases by DOCK exchange factors is mediated by a nucleotide sensor. Science 325: 1398-1402, 2009. [PubMed: 19745154] [Full Text: https://doi.org/10.1126/science.1174468]

  47. Yasuda, S., Oceguera-Yanez, F., Kato, T., Okamoto, M., Yonemura, S., Terada, Y., Ishizaki, T., Narumiya, S. Cdc42 and mDia3 regulate microtubule attachment to kinetochores. Nature 428: 767-771, 2004. [PubMed: 15085137] [Full Text: https://doi.org/10.1038/nature02452]

  48. Zheng, Y., Fischer, D. J., Santos, M. F., Tigyi, G., Pasteris, N. G., Gorski, J. L., Xu, Y. The faciogenital dysplasia gene product FGD1 functions as a Cdc42Hs-specific guanine-nucleotide exchange factor. J. Biol. Chem. 271: 33169-33172, 1996. [PubMed: 8969170] [Full Text: https://doi.org/10.1074/jbc.271.52.33169]


Contributors:
Ada Hamosh - updated : 09/18/2019
Cassandra L. Kniffin - updated : 04/04/2018
Ada Hamosh - updated : 02/21/2018
Patricia A. Hartz - updated : 3/8/2016
Ada Hamosh - updated : 1/8/2016
Ada Hamosh - updated : 6/24/2015
Ada Hamosh - updated : 12/9/2013
Ada Hamosh - updated : 5/6/2013
Paul J. Converse - updated : 10/26/2012
Matthew B. Gross - updated : 5/10/2011
Ada Hamosh - updated : 5/9/2011
Patricia A. Hartz - updated : 1/6/2011
Matthew B. Gross - updated : 5/11/2010
Ada Hamosh - updated : 1/8/2010
Ada Hamosh - updated : 10/13/2009
Ada Hamosh - updated : 2/18/2009
Patricia A. Hartz - updated : 4/28/2008
Patricia A. Hartz - updated : 8/30/2007
Patricia A. Hartz - updated : 1/19/2007
Patricia A. Hartz - updated : 5/3/2006
Patricia A. Hartz - updated : 3/28/2006
Patricia A. Hartz - updated : 10/7/2004
Ada Hamosh - updated : 9/28/2004
Ada Hamosh - updated : 4/16/2004
Cassandra L. Kniffin - updated : 3/5/2003
Ada Hamosh - updated : 1/29/2003
Patricia A. Hartz - updated : 6/5/2002
Stylianos E. Antonarakis - updated : 9/7/2000
Ada Hamosh - updated : 7/20/2000
Ada Hamosh - updated : 3/10/2000
Victor A. McKusick - updated : 8/23/1999
Jennifer P. Macke - updated : 4/8/1998
Jennifer P. Macke - updated : 5/28/1997
Alan F. Scott - updated : 3/6/1996

Creation Date:
Victor A. McKusick : 1/17/1991

Edit History:
alopez : 03/21/2023
carol : 08/04/2020
alopez : 09/18/2019
carol : 04/09/2018
carol : 04/06/2018
ckniffin : 04/04/2018
alopez : 02/21/2018
carol : 02/22/2017
carol : 08/08/2016
carol : 03/18/2016
carol : 3/9/2016
alopez : 3/8/2016
alopez : 1/13/2016
alopez : 1/8/2016
alopez : 1/8/2016
alopez : 6/24/2015
alopez : 12/9/2013
alopez : 5/6/2013
mgross : 11/20/2012
terry : 10/26/2012
mgross : 5/10/2011
alopez : 5/10/2011
terry : 5/9/2011
mgross : 1/24/2011
terry : 1/6/2011
wwang : 5/17/2010
mgross : 5/11/2010
mgross : 5/11/2010
alopez : 1/11/2010
alopez : 1/11/2010
terry : 1/8/2010
alopez : 10/22/2009
terry : 10/13/2009
carol : 7/7/2009
alopez : 2/20/2009
terry : 2/18/2009
mgross : 4/28/2008
ckniffin : 2/5/2008
mgross : 10/4/2007
terry : 8/30/2007
mgross : 1/19/2007
mgross : 6/7/2006
terry : 5/3/2006
wwang : 4/3/2006
terry : 3/28/2006
mgross : 10/7/2004
alopez : 10/4/2004
tkritzer : 9/28/2004
alopez : 4/19/2004
terry : 4/16/2004
cwells : 11/10/2003
tkritzer : 3/14/2003
ckniffin : 3/5/2003
alopez : 3/3/2003
alopez : 1/29/2003
terry : 1/29/2003
alopez : 11/19/2002
terry : 11/18/2002
carol : 6/5/2002
terry : 11/15/2001
mgross : 9/7/2000
alopez : 7/20/2000
alopez : 3/10/2000
mcapotos : 12/7/1999
psherman : 11/3/1999
psherman : 10/18/1999
jlewis : 9/3/1999
terry : 8/23/1999
kayiaros : 7/13/1999
psherman : 3/18/1999
psherman : 4/21/1998
dholmes : 4/8/1998
dholmes : 4/8/1998
alopez : 8/1/1997
alopez : 7/23/1997
alopez : 7/23/1997
mark : 12/16/1996
terry : 12/10/1996
terry : 4/17/1996
mark : 3/6/1996
carol : 4/1/1994
supermim : 3/16/1992
carol : 1/2/1992
carol : 3/4/1991
carol : 1/17/1991