Вы находитесь на странице: 1из 14

Advances in Biological Regulation 55 (2014) 114

Contents lists available at ScienceDirect

Advances in Biological Regulation


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

An expanding role for RAS GTPase activating


proteins (RAS GAPs) in cancer
Ophlia Maertens a, b, Karen Cichowski a, b, c, *
a
Genetics Division, Department of Medicine, Brigham and Womens Hospital, Boston, MA 02115, USA
b
Harvard Medical School, Boston, MA 02115, USA
c
Ludwig Center at Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA

a b s t r a c t

Keywords: The RAS pathway is one of the most commonly deregulated


RAS pathways in human cancer. Mutations in RAS genes occur in nearly
GAP 30% of all human tumors. However in some tumor types RAS
GTPase activating protein
mutations are conspicuously absent or rare, despite the fact that
RAS GAP
RAS and downstream effector pathways are hyperactivated.
NF1
RASAL2 Recently, RAS GTPase Activating Proteins (RAS GAPs) have
DAB2IP emerged as an expanding class of tumor suppressors that, when
IQGAP inactivated, provide an alternative mechanism of activating RAS.
Therapeutic resistance RAS GAPs normally turn off RAS by catalyzing the hydrolysis of
RAS-GTP. As such, the loss of a RAS GAP would be expected to
promote excessive RAS activation. Indeed, this is the case for the
NF1 gene, which plays an established role in a familial tumor
predisposition syndrome and a variety of sporadic cancers. How-
ever, there are 13 additional RAS GAP family members in the
human genome. We are only now beginning to understand why
there are so many RAS GAPs, how they differentially function, and
what their potential role(s) in human cancer are. This review will
focus on our current understanding of RAS GAPs in human disease
and will highlight important outstanding questions.
2014 Elsevier Ltd. All rights reserved.

* Corresponding author. 458d NRB, 77 Avenue Louis Pasteur, Boston, MA 02115, USA. Tel.: 1 (617) 525 4722.
E-mail address: kcichowski@rics.bwh.harvard.edu (K. Cichowski).

http://dx.doi.org/10.1016/j.jbior.2014.04.002
2212-4926/ 2014 Elsevier Ltd. All rights reserved.
2 O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114

Introduction

RAS proteins are small molecular weight GTPases that couple extracellular signals to intracellular
effector pathways. The most extensively studied RAS isoforms (H-RAS, K-RAS and N-RAS) play a critical
role in fundamental cellular processes, including proliferation, survival, differentiation, motility and
transcription (Karnoub and Weinberg, 2008). RAS proteins together with their two key regulators,
guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs), constitute mo-
lecular switches that cycle between on and off conformations caused by the binding of GTP or GDP,
respectively (Fig. 1). The transition between the inactive GDP-bound and active GTP-bound state of RAS
is controlled by GEFs, which activate RAS by promoting the exchange of GDP for GTP, and GAPs, which
turn off RAS by catalyzing RAS-mediated GTP hydrolysis (Bos et al., 2007). The RAS pathway is one of
the most commonly deregulated pathways in human cancer (Karnoub and Weinberg, 2008). Activating
mutations in RAS genes occur in many different tumor types. These mutations typically render RAS
constitutively GTP-bound, resulting in the activation of downstream effector pathways in the absence
of extracellular stimuli (Karnoub and Weinberg, 2008). However, RAS GAPs have emerged as an
expanding new class of tumor suppressor genes. Accumulating evidence indicates that inactivation of
several RAS GAP family members represents an important and alternative mechanism to (hyper)
activate RAS and its downstream effectors in tumors (Fig. 1). This review will focus on the emerging
biological role of RAS GAPs in human cancer.
There are 14 predicted RAS GAP genes in the human genome (Bernards, 2003). All contain a RAS
GAP domain but share little similarity outside of this region (Fig. 2). Interestingly, members of the
IQGAP subfamily contain an alternative amino acid within a key region of the catalytic domain, and
therefore do not exhibit RAS GAP activity but still affect RAS signaling (Brill et al., 1996; Wang et al.,
2007; Weissbach et al., 1994). Regions anking the RAS GAP domains are thought to promote pro-
teinprotein and proteinlipid interactions, second messenger binding and phosphorylation by protein
kinases. These interactions may facilitate association with specic subcellular membranes or com-
partments, regulating spatially restricted GTPase activation (Bernards and Settleman, 2004). Accord-
ingly, individual RAS GAPs may target specic GTPases by associating with specic membranes and/or
signaling complexes. Furthermore, RAS GAP activity and its effects on downstream signaling pathways
appears to be regulated in response to specic growth factors which is likely tissue- and context-
specic (Bernards and Settleman, 2005; Cichowski et al., 2003; McGillicuddy et al., 2009). Finally, at
least some RAS GAPs also have distinct RAS-independent functions (Min et al., 2010; White et al., 2012).
Together, this illustrates that RAS GAPs play diverse and discrete, yet non-redundant, roles in cell
signaling. The existence of distinct sets of modular domains in different RAS GAP proteins further
indicates that RAS GAP activity is highly complex and very tightly regulated. Nevertheless, still

Fig. 1. The RAS GTPase cycle and its deregulation in cancer. (A) RAS proteins together with their two key regulators, guanine
nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs), constitute molecular switches that cycle between on and
off conformations caused by the binding of GTP or GDP, respectively. (B) Activating mutations in RAS genes occur in many different
tumor types. These mutations typically render RAS constitutively GTP-bound, resulting in the activation of downstream effector
pathways in the absence of extracellular stimuli. (C) Increasing evidence suggests that inactivation of several RAS GAP family
members represents an important and alternative mechanism to (hyper)activate RAS and its downstream effectors in cancer.
O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114 3

Fig. 2. The RAS GAP family. There are 14 predicted RAS GAP genes in the human genome. All contain a RAS GAP domain but share
little similarity outside of this region. Members of the IQGAP subfamily contain an alternative amino acid within a key region of the
catalytic domain, and therefore do not exhibit RAS GAP activity but still affect RAS signaling.

relatively little is known about the precise function and regulation of many of these RAS GAP proteins
in normal processes or cancer. Moreover, the fundamental question of why so many RAS GAPs exist is
still not well understood.
The most extensively studied RAS GAP is neurobromin, the protein product of the NF1 gene
(Cichowski and Jacks, 2001). Despite the fact that germline mutations in NF1 were known to underlie
the cancer predisposition syndrome neurobromatosis type 1 (NF1), until recently a role for NF1 in
sporadic cancers had not been appreciated. Through genomic, cellular, and mouse modeling studies it
has now become evident that the NF1 gene/protein plays a much broader role in cancer than previously
recognized. There is also accumulating evidence that other RAS GAPs, such as DAB2IP, RASAL2, and
possibly RASA1, IQGAP2 and RASAL1 may function as bona de tumor suppressors and a surprising pro-
tumorigenic role has been proposed for IQGAP1. In this review, we will summarize studies describing
aberrant RAS GAP function in cancer for which sufcient validation supports a causal role in tumor
growth and metastasis. We will also highlight the emerging role of RAS GAPs in mediating resistance to
targeted therapies. Finally, we will address the mechanisms that alter RAS GAP gene or protein
expression in malignant transformation.

NF1: A tumor suppressor in familial and sporadic cancers

The NF1 gene and protein

The NF1 gene was identied by positional cloning in 1990 and encodes the RAS GAP known as
neurobromin (Ballester et al., 1990; Martin et al., 1990; Xu et al., 1990). Neurobromin is ubiquitously
expressed, although it is most abundant in the nervous system. Beyond the RAS GAP domain and the
immediately adjacent lipid-binding SEC14-PH domain (DAngelo et al., 2006; Welti et al., 2007), no
obvious protein motifs have been recognized in neurobromin. As the RAS GAP domain constitutes
only a small portion of the total protein (w13%), it has been suggested that other non-RAS associated
functions might exist. To this end, studies in Drosophila have shown that neurobromin can regulate
the cAMP pathway by regulating adenylyl cyclase activity (Guo et al., 2000; Tong et al., 2002; Walker
et al., 2013), although the molecular mechanism by which this occurs remains elusive.

Neurobromatosis type 1

Germline mutations and deletions of the NF1 gene underlie the familial cancer syndrome neuro-
bromatosis type 1 (NF1), which affects 1 in 3500 individuals worldwide (Riccardi, 1992). The hallmark
4 O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114

clinical feature is the development of peripheral nervous system tumors known as neurobromas.
While these lesions are typically benign, patients can develop hundreds of tumors throughout the
peripheral nervous system, which can be painful, debilitating and in some instances lethal. Moreover,
approximately 10% of individuals with NF1 will develop a malignant peripheral nerve sheath tumor
(MPNST). NF1 patients are also predisposed to developing a variety of other tumors, including gliomas,
pheochromocytomas and myeloid leukemia (Riccardi, 1992). Non-cancerous symptoms of the disease
include intellectual decits, bone deformations and hyperpigmentation defects such as caf-au-lait
spots and Lisch nodules. This broad spectrum of clinical manifestations in NF1 patients highlights the
requisite function of neurobromin in a wide variety of tissues and cell types.
In most instances NF1 behaves as a classical tumor suppressor gene. Genomic studies and mouse
models have shown that loss of heterozygosity (LOH) or second hit NF1 mutations are required for the
development of neurobromas, MPNSTs, pheochromocytomas and myeloid malignancies (Cichowski
et al., 1999; Jacks et al., 1994; Legius et al., 1993; Rutkowski et al., 2000; Serra et al., 2000; Shannon
et al., 1994; Vogel et al., 1999; Xu et al., 1992; Zhu et al., 2002). More than two decades of work has
now revealed critical insight relating to tumor cells of origin, cooperating genetic events, non-cell
autonomous signals, and molecular pathways underlying the pathophysiology of these NF1-related
tumors (Upadhyaya and Cooper, 2012). For example, loss of the wild-type NF1 allele in Schwann cell
precursors is the rate-limiting step for neurobroma development; however in some settings tumor
development requires NF1 haploinsufcency of surrounding cells within the microenvironment (Le
et al., 2011; Maertens et al., 2007; Mayes et al., 2011; Serra et al., 2000; Wu et al., 2008; Zheng et al.,
2008; Zhu et al., 2002). We also know that additional cooperating genetic lesions, such as P53 or
INK4A/Arf loss, are required for the formation of malignant gliomas and MPNSTs (Cichowski et al.,
1999; Joseph et al., 2008; Kourea et al., 1999; Legius et al., 1994; Nielsen et al., 1999; Vogel et al.,
1999; Zhu et al., 2005). In all instances it is clear that RAS-GTP levels are elevated in NF1-decient
lesions and that the downstream MAPK and PI3K/AKT pathways are generally activated as a conse-
quence of NF1 loss (Basu et al., 1992; DeClue et al., 1992; Johannessen et al., 2005; Lau et al., 2000).
Preclinical studies have further demonstrated the requirement for these RAS effector pathways in NF1-
mutant tumors (Chang et al., 2013; Dasgupta et al., 2005; De Raedt et al., 2011; Jessen et al., 2013;
Johannessen et al., 2005). Interestingly however, it appears that different effector pathways may be
dominant in different tumor types. For example while PI3K/mTOR appears to be the dominant pathway
in MPNSTs and gliomas (Dasgupta et al., 2005; Johannessen et al., 2005), the MAPK pathway is more
critical in benign neurobromas and JMML (Chang et al., 2013; Jessen et al., 2013).

NF1 loss in sporadic cancers

Recently, it has become evident that the NF1 gene/protein plays a much broader role in cancer than
previously recognized. High throughput genomic studies suggest that NF1 is mutated and/or inacti-
vated in a wide range of sporadic tumors, such as glioblastoma, lung adenocarcinoma, breast cancer,
leukemia, ovarian cancer and neuroblastoma (Boudry-Labis et al., 2013; Ding et al., 2008; Hodis et al.,
2012; Holzel et al., 2010; Krauthammer et al., 2012; McGillicuddy et al., 2009; Parsons et al., 2008;
TCGA, 2008; TCGA, 2011; TCGA, 2012). Although additional functional studies are required to under-
stand the biological signicance of many of these observations, cellular and animal models have
provided mechanistic insight into how NF1 cooperates with other genetic events in glioblastoma and
melanoma (Maertens et al., 2013; McGillicuddy et al., 2009; Zhu et al., 2005). Importantly, NF1 alter-
ations are not always mutually exclusive with RAS mutations or aberrations in RAS downstream
effector pathways such as BRAF or PTEN. Moreover, the mechanism of NF1 inactivation and its resulting
expression levels seem to be dictated by the genetic context (McGillicuddy et al., 2009).
In sporadic human gliomas, for example, NF1 has been shown to be inactivated via two distinct
mechanisms: proteasomal degradation and genetic loss (McGillicuddy et al., 2009). Notably, complete
genetic inactivation can only be tolerated in the presence of P53 mutations (McGillicuddy et al., 2009).
When P53 is intact, NF1-decient tumor cells become senescent in vitro and in vivo, as observed in
benign neurobromas and astrocytomas from NF1 patients (Riccardi, 1992). Loss of NF1 thus triggers a
senescence response, which restricts further progression of benign lesions (Courtois-Cox et al., 2006).
In order to evade senescence, additional genetic defects are required. This explains why bi-allelic
O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114 5

inactivation of NF1 occurs in the context of P53 loss in NF1-related malignant gliomas and MPNSTs
(Cichowski et al., 1999; Vogel et al., 1999; Zhu et al., 2005), as well as in sporadic gliomas (McGillicuddy
et al., 2009). However, neurobromin can also be excessively degraded by the proteasome. Importantly,
neurobromin instability promotes tumorigenesis but is not sufcient to trigger a senescence response
(McGillicuddy et al., 2009). These ndings illustrate how different mechanisms of inactivation may be
required in genetically distinct tumors, in order to evade the senescence response.
Likewise, oncogene induced senescence is known to be important for restricting melanoma
development in response to activating BRAF mutations. Specically, oncogenic RAF can trigger a potent
negative feedback loop that suppresses RAS and downstream PI3K/AKT signaling, which plays a causal
role in the senescent response (Courtois-Cox et al., 2006; Maertens et al., 2013). Studies using genet-
ically engineered mouse models have demonstrated that in this setting Nf1 loss actually prevents Braf-
induced senescence of melanocytes, by relieving RAS and consequential PI3K repression (Maertens
et al., 2013). In mice, compound mutations in BRaf and Nf1 trigger excessive melanocyte hyper-
proliferation, which requires PI3K activation, and ultimately drive melanoma development. Impor-
tantly, NF1 mutations co-occur with BRAF mutations in human melanoma, and neurobromin
reconstitution potently suppresses the growth of human melanoma cells as xenografts, further sup-
porting a causal role for NF1 loss in melanomagenesis (Hodis et al., 2012; Krauthammer et al., 2012;
Maertens et al., 2013; Nissan et al., 2014). However, NF1 mutations also occur in the absence of acti-
vating BRAF mutations and can also co-occur with activating NRAS mutations (Hodis et al., 2012;
Krauthammer et al., 2012; Maertens et al., 2013; Nissan et al., 2014). This may be due to the nding
that, at least in melanoma, NF1 loss activates K- and H- but not N-RAS (Maertens et al., 2013). This
observation illustrates how NF1 mutations differ from activating mutations in a single RAS allele and
has important implications for mechanisms leading to drug resistance.

NF1 and resistance to targeted therapies

Targeted therapies directed against genetically well-dened vulnerabilities in human tumors have
been validated in the clinic. However, the relatively rapid acquisition of resistance to such treatments in
virtually all cases signicantly remains a substantial challenge to the clinical management of advanced
cancers (Lackner et al., 2012). As such, identifying mechanisms of resistance to inform the development
of second-line therapies or more durable rst-line treatments is of paramount importance.
Accumulating evidence suggests that NF1 plays an important role in mediating resistance to tar-
geted therapies. More specically, NF1 loss has been shown to drive resistance to kinase inhibitors in a
number of clinical settings, including BRAF-driven melanoma and EGFR-driven lung cancer (de Bruin
et al., 2014; Maertens et al., 2013; Nissan et al., 2014; Van Allen et al., 2014; Wang et al., 2014; Whittaker
et al., 2013). Genome-wide functional screens further identied loss of NF1 as a mediator of resistance
against tamoxifen in breast cancer cell lines and dasatinib in lung cancer cell lines (Beauchamp et al.,
2014; Mendes-Pereira et al., 2012). Elevated activation of RAS and its downstream pathways has been
shown to be the underlying mechanism of acquired resistance in all these cases. It is therefore tempting
to speculate that reduced expression levels of other functional RAS GAPs may also affect the sensitivity
to kinase inhibitors or different drugs in other cancers. Notably, while NF1 mutations have been
identied in resistant tumors and cell lines, loss of NF1 mRNA and protein expression appears to be
more common than bi-allelic alterations (de Bruin et al., 2014; Van Allen et al., 2014; Whittaker et al.,
2013).

Neurobromin inactivation by the proteasome

Despite its identication as a RAS GAP over 20 years ago, the regulation of neurobromin activity is
still not entirely understood. Neurobromin has been reported to be phosphorylated, but the func-
tional consequences of these phosphorylation events are unknown (Feng et al., 2004; Izawa et al., 1996;
Tokuo et al., 2001). Interestingly however, neurobromin is acutely degraded by the ubiquitin pro-
teasome pathway in response to a variety of growth factors through the activation of protein kinase C
(Cichowski et al., 2003; McGillicuddy et al., 2009). Importantly, these studies also demonstrated that
neurobromin degradation is essential for maximal RAS activation in response to many growth factors
6 O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114

and that its re-expression is required to appropriately attenuate RAS signaling (Cichowski et al., 2003).
More recently the Cullin 3 and KBTBD7 (kelch repeat and BTB domain-containing 7) ubiquitin ligase
complex has been shown to control both the regulated proteasomal degradation of neurobromin and
the pathogenic destabilization of neurobromin in glioblastomas (Hollstein and Cichowski, 2013).
Together these ndings may have important clinical implications, as proteasome inhibitors, Cullin
inhibitors, PKC inhibitors and neddylation inhibitors would be expected to stabilize neurobromin and
therefore could be evaluated in tumor types such as glioblastoma, where neurobromin is inactivated/
degraded by the proteasome.

Additional mechanisms of neurobromin regulation

The mechanism by which neurobromin is recruited to specic intracellular compartments to


regulate growth factor induced RAS signaling also remains unclear; however, neurobromin contains a
lipid-binding SEC14-PH domain, which likely contributes to membrane localization (DAngelo et al.,
2006; Welti et al., 2007). Interestingly, recent work suggests that SPRED1 recruits neurobromin to
active signaling complexes at the plasma membrane (Stowe et al., 2012). Members of the SPRED
(Sprouty related protein with an N-terminal EVH1 domain) family turn off RAS signaling following
receptor kinase activation (Kim and Bar-Sagi, 2004). Specically, SPRED1 has been reported to recruit
neurobromin to the membrane via its EVH1 domain. From this location neurobromin promotes the
hydrolysis of RAS-GTP (Stowe et al., 2012). Notably, SPRED proteins also bind to c-KIT, and perhaps to
other receptor tyrosine kinases, suggesting that neurobromin regulates RAS locally in response to
specic receptor signaling, rather than simply suppressing RAS throughout the plasma membrane.
Notably, mutations in the SPRED1 gene underlie a syndrome that is clinically related to neurobro-
matosis type 1 (Brems et al., 2007). As such, the overlapping features between both clinical entities may
be explained by this direct interaction (Stowe et al., 2012).
Taken together, it has become clear that the NF1 tumor suppressor plays a broad and important role in
familial as well as sporadic cancer. Further work on the regulatory control and structural domain analysis
of neurobromin will have a profound impact on our mechanistic understanding of the precise function
of neurobromin in different tissues and contexts, and hence its biological consequences when lost.

DAB2IP: A tumor and metastasis suppressor that functions by concurrently regulating different
oncogenic pathways

DAB2IP loss promotes its effects via both RAS-dependent and -independent pathways

DAB2IP is a second important RAS GAP tumor suppressor to be validated as a human tumor sup-
pressor. However, while genetic alterations in DAB2IP do occur in human cancers, the DAB2IP gene
appears to be more frequently inactivated by epigenetic silencing and is suppressed in lung, breast,
prostate, hepatocellular and gastrointestinal tumors (Chen et al., 2005; Dote et al., 2005, 2004; Yano
et al., 2005; Zhang et al., 2012). Functional studies have now specically revealed a direct causal role
for DAB2IP loss in driving metastatic prostate cancer (Min et al., 2010; Wu et al., 2014; Xie et al., 2010).
However, DAB2IP appears to mediate its effects via both RAS-dependent and -independent pathways,
at least in this tumor type.
Activating RAS mutations are rare in prostate cancer, despite the fact that RAS effector pathways are
frequently activated and their activation correlates with progression and metastasis (Gioeli et al., 1999;
Jeong et al., 2008; Malik et al., 2002; Shen and Abate-Shen, 2007). Studies using an orthotopic model of
prostate cancer further demonstrated that HRASV12 could drive prostate cancer in the presence of
cooperating immortalizing genetic alterations (Berger et al., 2004). Because DAB2IP appeared to be
epigenetically silenced in prostate cancer cell lines, a functional role for DAB2IP inactivation in prostate
cancer was formally investigated. Interestingly, while HRASV12 promoted the development of high-
grade prostate adenocarcinomas in the orthotopic model described above, DAB2IP suppression simi-
larly drove tumor development but also triggered widespread metastasis (Min et al., 2010). These
results suggested that DAB2IP loss was more potent than an activated RAS allele and/or that additional
pathways were activated in response to DAB2IP suppression.
O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114 7

As a member of the SynGAP family DAB2IP contains a PH domain and a single C2 domain both
N-terminal to the RAS GAP domain (Fig. 2). While both domains are thought to promote consti-
tutive plasma membrane association, a third more C-terminal period-like domain has been shown
to mediate an interaction with TRAF2, which suppresses NF-kB signaling (Zhang et al., 2004). As
such, loss of DAB2IP results in the activation of both the RAS and NF-kB pathways (Min et al., 2010).
Additional genetic and functional studies have now shown that in prostate cancer DAB2IPs effects
on RAS (through the RAS GAP domain) regulate primary tumor development, whereas its effects on
NF-kB (through the period-like domain) regulate metastasis (Min et al., 2010). Interestingly, DAB2IP
has also been reported to interact with GSK3b and PP2A via its C2 domain (Xie et al., 2010).
Therefore, while additional functional studies are required to fully elucidate DAB2IP function, this
RAS GAP appears to serve as a signaling scaffold that coordinately regulates two or three prominent
oncogenic pathways.

Mechanisms of DAB2IP inactivation in cancer

While DAB2IP mutations and deletions are found in different types of cancer (Barretina et al.,
2012; Forbes et al., 2011), epigenetic suppression seems to be the more prevalent mechanism of
inactivation. For example, DAB2IP is a target of the polycomb repressive complex 2 (PRC2)
(Schwartz and Pirrotta, 2013; Yu et al., 2007). Functional studies have further shown that EZH2,
which encodes the histone methyltransferase component of PRC2, is overexpressed in many cancer
types, including prostate cancer, and its expression correlates with tumor grade and metastasis
(Simon and Lange, 2008). Chromatin immunoprecipitation, functional studies and genomic analysis
have shown that EZH2 drives prostate tumorigenesis and metastasis, in part, via transcriptional
silencing of DAB2IP (Min et al., 2010). However, like many PRC2 targets, DAB2IP may ultimately
become more stably silenced by methylation of promoter sequences (Dote et al., 2005, 2004; Yano
et al., 2005). Thus the signicance of sequential silencing via histone and DNA modications should
be investigated further.

RASAL2: another tumor and metastasis suppressor

RAS mutations are also relatively uncommon in breast cancer, despite the fact that RAS effector
pathways are hyperactivated in the most advanced tumors (Forbes et al., 2011; Mueller et al., 2000;
Sivaraman et al., 1997; von Lintig et al., 2000). However, the RAS GAP gene RASAL2 has now been
shown to function as a tumor suppressor in breast cancer through its effects on RAS (McLaughlin et al.,
2013). Interestingly, Rasal2 loss also generally promotes the metastasis of tumors in p53 mutant mice,
suggesting that it mat play a broader role in tumor development and/or progression (McLaughlin et al.,
2013).
RASAL2 was initially identied as a potential RAS GAP tumor suppressor in a functional cell-based
screen (Min et al., 2010). Interestingly, mutations in the catalytic RAS GAP domain of RASAL2 were
observed in a small but signicant percentage of breast tumors, suggesting its putative role in this
setting (Sjoblom et al., 2006). Therefore a functional role for RASAL2 inactivation was formally inves-
tigated. By performing gain- and loss-of-function studies in human xenografts and genetically engi-
neered mouse models, McLaughlin et al. demonstrated that RASAL2 suppression promoted breast
tumor development as a consequence of activating K- and H-RAS. Moreover, in a luminal model of
breast cancer, RASAL2 mutations promoted metastasis (McLaughlin et al., 2013).
Like DAB2IP, RASAL2 appears to be more frequently silenced by epigenetic mechanisms in breast
cancer. Specically, its suppression (via promoter methylation) was shown to be enriched in luminal B
tumors (McLaughlin et al., 2013). Immunohistochemical and protein array analysis further demon-
strated that low RASAL2 protein levels are associated with metastasis (McLaughlin et al., 2013).
Moreover, low RASAL2 mRNA levels correlate with recurrence and poor overall survival of individuals
with luminal B cancers (McLaughlin et al., 2013). These observations may provide a mechanistic
explanation why luminal B tumors are more aggressive and refractory to therapies than luminal A
lesions. Additional studies evaluating the role of RASAL2 and other RAS GAPs in breast cancer will be an
important endeavor in the future.
8 O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114

IQGAP1/2: scaffolding proteins with opposing effects

The IQGAP protein subfamily consists of three members with considerable sequence homology but
variable tissue distribution (Brill et al., 1996; Wang et al., 2007; Weissbach et al., 1994). The IQGAP
proteins are scaffolding proteins that contain multiple protein-interacting domains (White et al., 2012).
These regions include an actin-binding calponin homology domain (CH), a polyproline-binding domain
(WW), four IQ calmodulin-binding motifs (IQ) and a RAS GAP-related domain (Fig. 2). The RAS GAP-
related domain lacks an arginine residue essential for GTP hydrolysis and therefore none of the IQGAP
proteins exhibit RAS GAP activity (Brill et al., 1996; Wang et al., 2007; Weissbach et al., 1994). Interest-
ingly, this RAS GAP subfamily appears to be comprised of both oncogenes and tumor suppressors.
The best characterized member of the IQGAP family is IQGAP1. Through its interaction with specic
proteins IQGAP1 is implicated in multiple fundamental cellular activities (White et al., 2012). For
example, IQGAP1 binds to the Rho GTPases Rac1 and Cdc42 to regulate the assembly of the cyto-
skeleton (Erickson et al., 1997) and combines with microtubule-associated proteins to regulate cell
polarization and determine the direction of cell movement (Fukata et al., 2002). Furthermore, IQGAP1
binds members of the MAPK and PI3K/AKT signaling pathways to mediate cell proliferation and dif-
ferentiation (Bourguignon et al., 2005; Ren et al., 2007; Roy et al., 2004, 2005) and works with
calmodulin, b-catenin and E-cadherin to regulate intercellular adhesion and migration (Fukata et al.,
1999; Ho et al., 1999; Kuroda et al., 1998). Many of the identied IQGAP1 binding partners have
well-dened roles in tumorigenesis (White et al., 2009). This observation, together with the ability of
IQGAP1 to modulate a wide variety of cellular processes, suggests that IQGAP1 may be involved in
various aspects of tumor biology.
Accumulating evidence indeed suggests a positive role for IQGAP1 in cancer. Specically, IQGAP1 is
overexpressed and/or amplied in colorectal cancer, pancreatic adenocarcinoma, glioblastoma and
ovarian cancer, and its expression is correlated with poor prognosis (Dong et al., 2006; McDonald et al.,
2007; Nabeshima et al., 2002; Wang et al., 2013). Similar observations have been reported in mouse
models. Overexpression of Iqgap1 is associated with progression in a genetically engineered mouse
model that recapitulates the different stages of human prostate cancer (Ouyang et al., 2008). More
recently, IQGAP1 has been shown to function as a scaffold that is required for RAS-driven tumori-
genesis and metastasis. Specically, Iqgap1/ mice were resistant to Hras-driven chemical carcino-
genesis and depletion of IQGAP1 reduced the invasive properties of RAS-driven cancer cells (Jameson
et al., 2013). Nevertheless, the precise mechanism by which IQGAPs may directly contribute to
tumorigenesis and the mechanisms triggering IQGAP1s abnormal expression in cancer remain to be
elucidated. Notably, IQGAP1 appears to be dispensable for homeostasis and disruption of IQGAP1
binding to ERK diminishes tumorigenesis in vivo (Jameson et al., 2013), suggesting a therapeutic
window could be achieved by targeting IQGAP1 in some cancers.
In contrast to IQGAP1s apparent pro-tumorigenic role, IQGAP2 was found to be inactivated by pro-
moter methylation in gastric cancer (Jin et al., 2008) and Iqgap2 deciency leads to the development of
hepatocellular carcinoma in mice (Schmidt et al., 2008). Remarkably, concurrent Iqgap1 loss partially
rescues the tumor phenotype of Iqgap2/ mice and a reciprocal relationship exists between IQGAP1 and
IQGAP2 expression in human liver cancer cell lines (Schmidt et al., 2008; White et al., 2010). Together,
these data suggest that IQGAP1 and IQGAP2 have opposing oncogenic and tumor suppressive roles.
While there is no evidence for a direct interaction between both proteins, their relative expression and/
or stoichiometry might determine various cellular and biological consequences. Further studies are
needed to elucidate the relative contribution, specic binding partners, subcellular localization, distinct
tissue expression and post-translational modications of IQGAP1 and IQGAP2. This will help provide
insight into the seemingly paradoxical roles of these two very similar IQGAP proteins in cancer. Similarly,
additional work is necessary to determine the possible role of IQGAP3 in neoplasia.

Other RAS GAPs in cell signaling and cancer

P120 RAS GAP was the rst protein to be characterized as a RAS GAP (Trahey et al., 1988; Vogel et al.,
1988). In addition to the RAS GAP domain P120 contains SH2, SH3, PH, and C2 domains (Fig. 2). All four
domains are thought to be involved in targeting P120 to membranes, and initial studies suggested that
O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114 9

P120 can associate with receptor tyrosine kinases. For example, SH2 domains enable binding of P120 to
phosphorylated epidermal growth factor receptor, platelet derived growth factor receptor, and insulin
receptors which promotes inactivation of RAS in the respective signaling pathways (Agazie and
Hayman, 2003; Cooper and Kashishian, 1993; Ekman et al., 1999; Fantl et al., 1992; Kazlauskas et al.,
1990; Margolis et al., 1990). There is also evidence that the SH3 domain of P120 interacts with DLC1
and as such interferes with the Rho GAP activity of this protein (Yang et al., 2009). Although mutations
in the RASA1 gene, which encodes P120, have been found in individuals affected with a capillary and
arteriovenous malformation syndrome (Eerola et al., 2003), its link with cancer has remained more
obscure. An early genetic study reported somatic RASA1 mutations in basal cell carcinoma (Friedman
et al., 1993) and integrative bioinformatic analysis identied RASA1 as a candidate tumor suppressor
gene associated with triple negative and HER2 positive subtypes of breast cancer (Hu et al., 2009). A
recent computational study analyzing the patterns of mutational signatures in a broad panel of tumors
also predicts RASA1 to be a putative tumor suppressor (Davoli et al., 2013).
Somatic inactivation of RASAL1 by epigenetic silencing and mutations has been reported in a
number of human malignancies, including colorectal, thyroid and gastric cancer and nasopharyngeal
and esophageal tumors (Jin et al., 2007; Liu et al., 2013; Ohta et al., 2009; Seto et al., 2011). Knockdown
of RASAL1 as well as expression of RASAL1 mutants in cell lines resulted in RAS activation, increased
signaling via downstream MAPK and PI3K/AKT effectors, and accelerated growth of xenografted tu-
mors. Conversely, ectopic expression of wild-type RASAL1 had the opposite effects (Liu et al., 2013;
Ohta et al., 2009; Seto et al., 2011). Moreover, a genetic screen identied the transcription factor
PITX1 as a suppressor of RAS activity and tumorigenicity by upregulating RASAL1 (Kolfschoten et al.,
2005). Together, these data indicate that RASAL1 might function as a bona de tumor suppressor by
regulating RAS activity and that PITX1 may be one of the factors mediating its aberrant expression in
cancer. Reduced RASAL1 expression is detected more frequently in advanced lesions, suggesting that
RASAL1 might be involved in both tumor initiation and progression (Liu et al., 2013; Ohta et al., 2009;
Seto et al., 2011). Additional studies are required to formally demonstrate a causative role for RASAL1
loss in cancer.

Inactivation mechanisms of RAS GAPs in cancer: caveats and opportunities

Unlike the frequent direct mutational activation of RAS, a preponderance of evidence suggests that
RAS GAPs are commonly deregulated by non-genetic mechanisms. These non-mutational inactivation
mechanisms include epigenetic silencing, transcriptional repression and proteasome-mediated
degradation. Additional mechanisms are likely to be discovered. Regulation of RAS GAP expression
by non-coding RNAs, for example, remains an unexplored research area. Furthermore, post-
translational modications, proteinprotein interactions and aberrant localization can be expected
to positively or negatively regulate RAS GAP function. It is very likely that, as has been shown for PTEN,
the regulation of RAS GAPs by various molecular mechanisms allows to generate a continuum of
functional RAS GAP levels with differential consequences in inherited syndromes and sporadic cancers
(Song et al., 2012).
As such, protein analysis, rather than mutational or copy number status, might represent a more
accurate way to assess the true frequency of RAS GAP inactivation in cancer. The development of
reliable antibodies against specic members of the RAS GAP family, and guidelines on how to use these
in a clinical setting, is therefore of high priority. It will allow to evaluate the RAS GAP status in clinical
samples. Given the association of particular RAS GAPs with metastasis, poor prognosis and resistance
to targeted therapies, this assay could be used as a useful biomarker during clinical trials and for
routine pathological staining.
In contrast to the pro-tumorigenic IQGAP1 gene, the remaining RAS GAP tumor suppressor genes are
not considered classically druggable targets because it is traditionally easier to develop small molecule
antagonists rather than agonists. However, the observation that RAS GAPs are predominantly inacti-
vated by non-mutational mechanisms represents a major therapeutic opportunity. In principle,
restoring the expression of the wild-type (and functional) RAS GAP could be clinically exploited as a
strategy to inhibit the activation of wild-type RAS in particular cancers. For example, the insight that
neurobromin is degraded by the ubiquitin-proteasome pathway in glioblastoma suggests that small
10 O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114

molecule inhibitors aimed at blocking its destruction, would be expected to work in these tumors
(Hollstein and Cichowski, 2013; McGillicuddy et al., 2009). Similar approaches targeting the protea-
somal degradation of P53 by small molecules such as nutlin-3, which inhibits p53-MDM2 binding, have
been an active area of therapeutic development in hematologic malignancies and neuroblastoma
(Cheok et al., 2011).
Gaining further critical insight into the different mechanisms that alter RAS GAP expression in
cancer is therefore of particular importance. It should not only reveal prognostic biomarkers but can
also point towards the most druggable targets for therapeutic intervention. In the meantime the
challenge will be to develop a reliable means of quantifying RAS GAP suppression, and to better un-
derstand the threshold and consequences of reduced RAS GAP levels, so that these insights may ul-
timately be exploited in the clinic.

Concluding remarks

A growing body of evidence suggests that several RAS GAP proteins play an important and previ-
ously unrecognized role in cancer. In addition to genetic defects, it is clear that pathological mecha-
nisms that modulate RAS GAP gene or protein expression are associated with malignant
transformation. While ablation of RAS GAP activity represents an alternative mechanism to activate
RAS, the implications of RAS GAP loss appear to be more distinct and enigmatic and in some instances
promote the activation of additional oncogenic pathways. Further understanding of how the levels of
RAS GAP expression and activity are regulated, and what the biological consequences of RAS GAP
misregulation are, is therefore urgently needed. It can be expected that insights from these studies will
help shed light on the complex cellular signaling circuitry in normal biology and cancer.

Conicts of interest

The authors declare that there are no conicts of interest.

References

Agazie YM, Hayman MJ. Molecular mechanism for a role of SHP2 in epidermal growth factor receptor signaling. Mol Cell Biol
2003;23:787586.
Ballester R, Marchuk D, Boguski M, Saulino A, Letcher R, Wigler M, et al. The NF1 locus encodes a protein functionally related to
mammalian GAP and yeast IRA proteins. Cell 1990;63:8519.
Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, et al. The cancer cell line encyclopedia enables pre-
dictive modelling of anticancer drug sensitivity. Nature 2012;483:6037.
Basu TN, Gutmann DH, Fletcher JA, Glover TW, Collins FS, Downward J. Aberrant regulation of ras proteins in malignant tumour
cells from type 1 neurobromatosis patients. Nature 1992;356:7135.
Beauchamp EM, Woods BA, Dulak AM, Tan L, Xu C, Gray NS, et al. Acquired resistance to dasatinib in lung Cancer cell lines
conferred by DDR2 gatekeeper mutation and NF1 loss. Mol Cancer Ther 2014;13:47582.
Berger R, Febbo PG, Majumder PK, Zhao JJ, Mukherjee S, Signoretti S, et al. Androgen-induced differentiation and tumorigenicity
of human prostate epithelial cells. Cancer Res 2004;64:886775.
Bernards A. GAPs galore! A survey of putative Ras superfamily GTPase activating proteins in man and Drosophila. Biochim
Biophys Acta 2003;1603:4782.
Bernards A, Settleman J. GAP control: regulating the regulators of small GTPases. Trends Cell Biol 2004;14:37785.
Bernards A, Settleman J. GAPs in growth factor signalling. Growth Factors 2005;23:1439.
Bos JL, Rehmann H, Wittinghofer A. GEFs and GAPs: critical elements in the control of small G proteins. Cell 2007;129:86577.
Boudry-Labis E, Roche-Lestienne C, Nibourel O, Boissel N, Terre C, Perot C, et al. Neurobromatosis-1 gene deletions and
mutations in de novo adult acute myeloid leukemia. Am J Hematol 2013;88:30611.
Bourguignon LY, Gilad E, Rothman K, Peyrollier K. Hyaluronan-CD44 interaction with IQGAP1 promotes Cdc42 and ERK
signaling, leading to actin binding, Elk-1/estrogen receptor transcriptional activation, and ovarian cancer progression. J Biol
Chem 2005;280:1196172.
Brems H, Chmara M, Sahbatou M, Denayer E, Taniguchi K, Kato R, et al. Germline loss-of-function mutations in SPRED1 cause a
neurobromatosis 1-like phenotype. Nat Genet 2007;39:11206.
Brill S, Li S, Lyman CW, Church DM, Wasmuth JJ, Weissbach L, et al. The Ras GTPase-activating-protein-related human protein
IQGAP2 harbors a potential actin binding domain and interacts with calmodulin and Rho family GTPases. Mol Cell Biol
1996;16:486978.
Chang T, Krisman K, Theobald EH, Xu J, Akutagawa J, Lauchle JO, et al. Sustained MEK inhibition abrogates myeloproliferative
disease in Nf1 mutant mice. J Clin Invest 2013;123:3359.
Chen H, Tu SW, Hsieh JT. Down-regulation of human DAB2IP gene expression mediated by polycomb Ezh2 complex and histone
deacetylase in prostate cancer. J Biol Chem 2005;280:2243744.
O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114 11

Cheok CF, Verma CS, Baselga J, Lane DP. Translating p53 into the clinic. Nat Rev Clin Oncol 2011;8:2537.
Cichowski K, Jacks T. NF1 tumor suppressor gene function: narrowing the GAP. Cell 2001;104:593604.
Cichowski K, Santiago S, Jardim M, Johnson BW, Jacks T. Dynamic regulation of the Ras pathway via proteolysis of the NF1 tumor
suppressor. Genes Dev 2003;17:44954.
Cichowski K, Shih TS, Schmitt E, Santiago S, Reilly K, McLaughlin ME, et al. Mouse models of tumor development in neuro-
bromatosis type 1. Science 1999;286:21726.
Cooper JA, Kashishian A. In vivo binding properties of SH2 domains from GTPase-activating protein and phosphatidylinositol 3-
kinase. Mol Cell Biol 1993;13:173745.
Courtois-Cox S, Genther Williams SM, Reczek EE, Johnson BW, McGillicuddy LT, Johannessen CM, et al. A negative feedback
signaling network underlies oncogene-induced senescence. Cancer Cell 2006;10:45972.
DAngelo I, Welti S, Bonneau F, Scheffzek K. A novel bipartite phospholipid-binding module in the neurobromatosis type 1
protein. EMBO Rep 2006;7:1749.
Dasgupta B, Yi Y, Chen DY, Weber JD, Gutmann DH. Proteomic analysis reveals hyperactivation of the mammalian target of
rapamycin pathway in neurobromatosis 1-associated human and mouse brain tumors. Cancer Res 2005;65:275560.
Davoli T, Xu AW, Mengwasser KE, Sack LM, Yoon JC, Park PJ, et al. Cumulative haploinsufciency and triplosensitivity drive
aneuploidy patterns and shape the cancer genome. Cell 2013;155:94862.
de Bruin EC, Cowell CF, Warne PH, Jiang M, Saunders RE, Melnick MA, et al. Reduced NF1 expression confers resistance to EGFR
inhibition in lung cancer. Cancer Discov 2014;4:60619.
De Raedt T, Walton Z, Yecies JL, Li D, Chen Y, Malone CF, et al. Exploiting cancer cell vulnerabilities to develop a combination
therapy for ras-driven tumors. Cancer Cell 2011;20:40013.
DeClue JE, Papageorge AG, Fletcher JA, Diehl SR, Ratner N, Vass WC, et al. Abnormal regulation of mammalian p21ras con-
tributes to malignant tumor growth in von Recklinghausen (type 1) neurobromatosis. Cell 1992;69:26573.
Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, et al. Somatic mutations affect key pathways in lung
adenocarcinoma. Nature 2008;455:106975.
Dong P, Nabeshima K, Nishimura N, Kawakami T, Hachisuga T, Kawarabayashi T, et al. Overexpression and diffuse expression
pattern of IQGAP1 at invasion fronts are independent prognostic parameters in ovarian carcinomas. Cancer Lett 2006;243:
1207.
Dote H, Toyooka S, Tsukuda K, Yano M, Ota T, Murakami M, et al. Aberrant promoter methylation in human DAB2 interactive
protein (hDAB2IP) gene in gastrointestinal tumour. Br J Cancer 2005;92:111725.
Dote H, Toyooka S, Tsukuda K, Yano M, Ouchida M, Doihara H, et al. Aberrant promoter methylation in human DAB2 interactive
protein (hDAB2IP) gene in breast cancer. Clin Cancer Res 2004;10:20829.
Eerola I, Boon LM, Mulliken JB, Burrows PE, Dompmartin A, Watanabe S, et al. Capillary malformation-arteriovenous malfor-
mation, a new clinical and genetic disorder caused by RASA1 mutations. Am J Hum Genet 2003;73:12409.
Ekman S, Thuresson ER, Heldin CH, Ronnstrand L. Increased mitogenicity of an alphabeta heterodimeric PDGF receptor complex
correlates with lack of RasGAP binding. Oncogene 1999;18:24818.
Erickson JW, Cerione RA, Hart MJ. Identication of an actin cytoskeletal complex that includes IQGAP and the Cdc42 GTPase.
J Biol Chem 1997;272:244437.
Fantl WJ, Escobedo JA, Martin GA, Turck CW, del Rosario M, McCormick F, et al. Distinct phosphotyrosines on a growth factor
receptor bind to specic molecules that mediate different signaling pathways. Cell 1992;69:41323.
Feng L, Yunoue S, Tokuo H, Ozawa T, Zhang D, Patrakitkomjorn S, et al. PKA phosphorylation and 14-3-3 interaction regulate the
function of neurobromatosis type I tumor suppressor, neurobromin. FEBS Lett 2004;557:27582.
Forbes SA, Bindal N, Bamford S, Cole C, Kok CY, Beare D, et al. COSMIC: mining complete cancer genomes in the Catalogue of
Somatic Mutations in Cancer. Nucleic Acids Res 2011;39:D94550.
Friedman E, Gejman PV, Martin GA, McCormick F. Nonsense mutations in the C-terminal SH2 region of the GTPase activating
protein (GAP) gene in human tumours. Nat Genet 1993;5:2427.
Fukata M, Kuroda S, Nakagawa M, Kawajiri A, Itoh N, Shoji I, et al. Cdc42 and Rac1 regulate the interaction of IQGAP1 with beta-
catenin. J Biol Chem 1999;274:2604450.
Fukata M, Watanabe T, Noritake J, Nakagawa M, Yamaga M, Kuroda S, et al. Rac1 and Cdc42 capture microtubules through
IQGAP1 and CLIP-170. Cell 2002;109:87385.
Gioeli D, Mandell JW, Petroni GR, Frierson Jr HF, Weber MJ. Activation of mitogen-activated protein kinase associated with
prostate cancer progression. Cancer Res 1999;59:27984.
Guo HF, Tong J, Hannan F, Luo L, Zhong Y. A neurobromatosis-1-regulated pathway is required for learning in Drosophila.
Nature 2000;403:8958.
Ho YD, Joyal JL, Li Z, Sacks DB. IQGAP1 integrates Ca2/calmodulin and Cdc42 signaling. J Biol Chem 1999;274:46470.
Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M, Theurillat JP, et al. A landscape of driver mutations in melanoma. Cell
2012;150:25163.
Hollstein PE, Cichowski K. Identifying the Ubiquitin Ligase complex that regulates the NF1 tumor suppressor and Ras. Cancer
Discov 2013;3:88093.
Holzel M, Huang S, Koster J, Ora I, Lakeman A, Caron H, et al. NF1 is a tumor suppressor in neuroblastoma that determines
retinoic acid response and disease outcome. Cell 2010;142:21829.
Hu X, Stern HM, Ge L, OBrien C, Haydu L, Honchell CD, et al. Genetic alterations and oncogenic pathways associated with breast
cancer subtypes. Mol Cancer Res 2009;7:51122.
Izawa I, Tamaki N, Saya H. Phosphorylation of neurobromatosis type 1 gene product (neurobromin) by cAMP-dependent
protein kinase. FEBS Lett 1996;382:539.
Jacks T, Shih TS, Schmitt EM, Bronson RT, Bernards A, Weinberg RA. Tumour predisposition in mice heterozygous for a targeted
mutation in Nf1. Nat Genet 1994;7:35361.
Jameson KL, Mazur PK, Zehnder AM, Zhang J, Zarnegar B, Sage J, et al. IQGAP1 scaffold-kinase interaction blockade selectively
targets RAS-MAP kinase-driven tumors. Nat Med 2013;19:62630.
Jeong JH, Wang Z, Guimaraes AS, Ouyang X, Figueiredo JL, Ding Z, et al. BRAF activation initiates but does not maintain invasive
prostate adenocarcinoma. PLoS One 2008;3:e3949.
12 O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114

Jessen WJ, Miller SJ, Jousma E, Wu J, Rizvi TA, Brundage ME, et al. MEK inhibition exhibits efcacy in human and mouse
neurobromatosis tumors. J Clin Invest 2013;123:3407.
Jin H, Wang X, Ying J, Wong AH, Cui Y, Srivastava G, et al. Epigenetic silencing of a Ca(2)-regulated Ras GTPase-activating
protein RASAL denes a new mechanism of Ras activation in human cancers. Proc Natl Acad Sci U S A 2007;104:123538.
Jin SH, Akiyama Y, Fukamachi H, Yanagihara K, Akashi T, Yuasa Y. IQGAP2 inactivation through aberrant promoter methylation
and promotion of invasion in gastric cancer cells. Int J Cancer 2008;122:10406.
Johannessen CM, Reczek EE, James MF, Brems H, Legius E, Cichowski K. The NF1 tumor suppressor critically regulates TSC2 and
mTOR. Proc Natl Acad Sci U S A 2005;102:85738.
Joseph NM, Mosher JT, Buchstaller J, Snider P, McKeever PE, Lim M, et al. The loss of Nf1 transiently promotes self-renewal but
not tumorigenesis by neural crest stem cells. Cancer Cell 2008;13:12940.
Karnoub AE, Weinberg RA. Ras oncogenes: split personalities. Nat Rev Mol Cell Biol 2008;9:51731.
Kazlauskas A, Ellis C, Pawson T, Cooper JA. Binding of GAP to activated PDGF receptors. Science 1990;247:157881.
Kim HJ, Bar-Sagi D. Modulation of signalling by Sprouty: a developing story. Nat Rev Mol Cell Biol 2004;5:44150.
Kolfschoten IG, van Leeuwen B, Berns K, Mullenders J, Beijersbergen RL, Bernards R, et al. A genetic screen identies PITX1 as a
suppressor of RAS activity and tumorigenicity. Cell 2005;121:84958.
Kourea HP, Orlow I, Scheithauer BW, Cordon-Cardo C, Woodruff JM. Deletions of the INK4A gene occur in malignant peripheral
nerve sheath tumors but not in neurobromas. Am J Pathol 1999;155:185560.
Krauthammer M, Kong Y, Ha BH, Evans P, Bacchiocchi A, McCusker JP, et al. Exome sequencing identies recurrent somatic RAC1
mutations in melanoma. Nat Genet 2012;44:100614.
Kuroda S, Fukata M, Nakagawa M, Fujii K, Nakamura T, Ookubo T, et al. Role of IQGAP1, a target of the small GTPases Cdc42 and
Rac1, in regulation of E-cadherin- mediated cell-cell adhesion. Science 1998;281:8325.
Lackner MR, Wilson TR, Settleman J. Mechanisms of acquired resistance to targeted cancer therapies. Future Oncol 2012;8:9991014.
Lau N, Feldkamp MM, Roncari L, Loehr AH, Shannon P, Gutmann DH, et al. Loss of neurobromin is associated with activation of
RAS/MAPK and PI3-K/AKT signaling in a neurobromatosis 1 astrocytoma. J Neuropathol Exp Neurol 2000;59:75967.
Le LQ, Liu C, Shipman T, Chen Z, Suter U, Parada LF. Susceptible stages in Schwann cells for NF1-associated plexiform neuro-
broma development. Cancer Res 2011;71:468695.
Legius E, Dierick H, Wu R, Hall BK, Marynen P, Cassiman JJ, et al. TP53 mutations are frequent in malignant NF1 tumors. Genes
Chromosom Cancer 1994;10:2505.
Legius E, Marchuk DA, Collins FS, Glover TW. Somatic deletion of the neurobromatosis type 1 gene in a neurobrosarcoma
supports a tumour suppressor gene hypothesis. Nat Genet 1993;3:1226.
von Lintig FC, Dreilinger AD, Varki NM, Wallace AM, Casteel DE, Boss GR. Ras activation in human breast cancer. Breast Cancer
Res Treat 2000;62:5162.
Liu D, Yang C, Bojdani E, Murugan AK, Xing M. Identication of RASAL1 as a major tumor suppressor gene in thyroid cancer.
J Natl Cancer Inst 2013;105:161727.
Maertens O, De Schepper S, Vandesompele J, Brems H, Heyns I, Janssens S, et al. Molecular dissection of isolated disease features
in mosaic neurobromatosis type 1. Am J Hum Genet 2007;81:24351.
Maertens O, Johnson B, Hollstein P, Frederick DT, Cooper ZA, Messiaen L, et al. Elucidating distinct roles for NF1 in melano-
magenesis. Cancer Discov 2013;3:33849.
Malik SN, Brattain M, Ghosh PM, Troyer DA, Prihoda T, Bedolla R, et al. Immunohistochemical demonstration of phospho-Akt in
high Gleason grade prostate cancer. Clin Cancer Res 2002;8:116871.
Margolis B, Li N, Koch A, Mohammadi M, Hurwitz DR, Zilberstein A, et al. The tyrosine phosphorylated carboxyterminus of the
EGF receptor is a binding site for GAP and PLC-gamma. EMBO J 1990;9:437580.
Martin GA, Viskochil D, Bollag G, McCabe PC, Crosier WJ, Haubruck H, et al. The GAP-related domain of the neurobromatosis
type 1 gene product interacts with ras p21. Cell 1990;63:8439.
Mayes DA, Rizvi TA, Cancelas JA, Kolasinski NT, Ciraolo GM, Stemmer-Rachamimov AO, et al. Perinatal or adult Nf1 inactivation
using tamoxifen-inducible PlpCre each cause neurobroma formation. Cancer Res 2011;71:467585.
McDonald KL, OSullivan MG, Parkinson JF, Shaw JM, Payne CA, Brewer JM, et al. IQGAP1 and IGFBP2: valuable biomarkers for
determining prognosis in glioma patients. J Neuropathol Exp Neurol 2007;66:40517.
McGillicuddy LT, Fromm JA, Hollstein PE, Kubek S, Beroukhim R, De Raedt T, et al. Proteasomal and genetic inactivation of the
NF1 tumor suppressor in gliomagenesis. Cancer Cell 2009;16:4454.
McLaughlin SK, Olsen SN, Dake B, De Raedt T, Lim E, Bronson RT, et al. The RasGAP gene, RASAL2, is a tumor and metastasis
suppressor. Cancer Cell 2013;24:36578.
Mendes-Pereira AM, Sims D, Dexter T, Fenwick K, Assiotis I, Kozarewa I, et al. Genome-wide functional screen identies a
compendium of genes affecting sensitivity to tamoxifen. Proc Natl Acad Sci U S A 2012;109:27305.
Min J, Zaslavsky A, Fedele G, McLaughlin SK, Reczek EE, De Raedt T, et al. An oncogene-tumor suppressor cascade drives
metastatic prostate cancer by coordinately activating Ras and nuclear factor-kappaB. Nat Med 2010;16:28694.
Mueller H, Flury N, Eppenberger-Castori S, Kueng W, David F, Eppenberger U. Potential prognostic value of mitogen-activated
protein kinase activity for disease-free survival of primary breast cancer patients. Int J Cancer 2000;89:3848.
Nabeshima K, Shimao Y, Inoue T, Koono M. Immunohistochemical analysis of IQGAP1 expression in human colorectal carci-
nomas: its overexpression in carcinomas and association with invasion fronts. Cancer Lett 2002;176:1019.
Nielsen GP, Stemmer-Rachamimov AO, Ino Y, Moller MB, Rosenberg AE, Louis DN. Malignant transformation of neurobromas
in neurobromatosis 1 is associated with CDKN2A/p16 inactivation. Am J Pathol 1999;155:187984.
Nissan MH, Pratilas CA, Jones AM, Ramirez R, Won H, Liu C, et al. Loss of NF1 in cutaneous melanoma is associated with RAS
activation and MEK dependence. Cancer Res 2014;74:234050.
Ohta M, Seto M, Ijichi H, Miyabayashi K, Kudo Y, Mohri D, et al. Decreased expression of the RAS-GTPase activating protein
RASAL1 is associated with colorectal tumor progression. Gastroenterology 2009;136:20616.
Ouyang X, Jessen WJ, Al-Ahmadie H, Serio AM, Lin Y, Shih WJ, et al. Activator protein-1 transcription factors are associated with
progression and recurrence of prostate cancer. Cancer Res 2008;68:213244.
Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, et al. An integrated genomic analysis of human glioblastoma
multiforme. Science 2008;321:180712.
O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114 13

Ren JG, Li Z, Sacks DB. IQGAP1 modulates activation of B-Raf. Proc Natl Acad Sci U S A 2007;104:104659.
Riccardi VM. Neurobromatosis: phenotype, Natural History, and Pathogenesis; 1992.
Roy M, Li Z, Sacks DB. IQGAP1 binds ERK2 and modulates its activity. J Biol Chem 2004;279:1732937.
Roy M, Li Z, Sacks DB. IQGAP1 is a scaffold for mitogen-activated protein kinase signaling. Mol Cell Biol 2005;25:794052.
Rutkowski JL, Wu K, Gutmann DH, Boyer PJ, Legius E. Genetic and cellular defects contributing to benign tumor formation in
neurobromatosis type 1. Hum Mol Genet 2000;9:105966.
Schmidt VA, Chiariello CS, Capilla E, Miller F, Bahou WF. Development of hepatocellular carcinoma in Iqgap2-decient mice is
IQGAP1 dependent. Mol Cell Biol 2008;28:1489502.
Schwartz YB, Pirrotta V. A new world of Polycombs: unexpected partnerships and emerging functions. Nat Rev Genet 2013;14:
85364.
Serra E, Rosenbaum T, Winner U, Aledo R, Ars E, Estivill X, et al. Schwann cells harbor the somatic NF1 mutation in neuro-
bromas: evidence of two different Schwann cell subpopulations. Hum Mol Genet 2000;9:305564.
Seto M, Ohta M, Ikenoue T, Sugimoto T, Asaoka Y, Tada M, et al. Reduced expression of RAS protein activator like-1 in gastric
cancer. Int J Cancer 2011;128:1293302.
Shannon KM, OConnell P, Martin GA, Paderanga D, Olson K, Dinndorf P, et al. Loss of the normal NF1 allele from the bone
marrow of children with type 1 neurobromatosis and malignant myeloid disorders. N Engl J Med 1994;330:597601.
Shen MM, Abate-Shen C. Pten inactivation and the emergence of androgen-independent prostate cancer. Cancer Res 2007;67:
65358.
Simon JA, Lange CA. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat Res 2008;647:219.
Sivaraman VS, Wang H, Nuovo GJ, Malbon CC. Hyperexpression of mitogen-activated protein kinase in human breast cancer.
J Clin Invest 1997;99:147883.
Sjoblom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, et al. The consensus coding sequences of human breast and
colorectal cancers. Science 2006;314:26874.
Song MS, Salmena L, Pandol PP. The functions and regulation of the PTEN tumour suppressor. Nat Rev Mol Cell Biol 2012;13:
28396.
Stowe IB, Mercado EL, Stowe TR, Bell EL, Oses-Prieto JA, Hernandez H, et al. A shared molecular mechanism underlies the
human rasopathies Legius syndrome and Neurobromatosis-1. Genes Dev 2012;26:14216.
TCGA. Comprehensive genomic characterization denes human glioblastoma genes and core pathways. Nature 2008;455:1061
8.
TCGA. Integrated genomic analyses of ovarian carcinoma. Nature 2011;474:60915.
TCGA. Comprehensive molecular portraits of human breast tumours. Nature 2012;490:6170.
Tokuo H, Yunoue S, Feng L, Kimoto M, Tsuji H, Ono T, et al. Phosphorylation of neurobromin by cAMP-dependent protein
kinase is regulated via a cellular association of N(G),N(G)-dimethylarginine dimethylaminohydrolase. FEBS Lett 2001;494:
4853.
Tong J, Hannan F, Zhu Y, Bernards A, Zhong Y. Neurobromin regulates G protein-stimulated adenylyl cyclase activity. Nat
Neurosci 2002;5:956.
Trahey M, Wong G, Halenbeck R, Rubinfeld B, Martin GA, Ladner M, et al. Molecular cloning of two types of GAP complementary
DNA from human placenta. Science 1988;242:1697700.
Upadhyaya, M., and Cooper, D. N. (2012). Neurobromatosis Type 1, (Berlin Heidelberg: Springer-Verlag)
Van Allen EM, Wagle N, Sucker A, Treacy DJ, Johannessen CM, Goetz EM, et al. The genetic landscape of clinical resistance to RAF
inhibition in metastatic melanoma. Cancer Discov 2014;4:94109.
Vogel KS, Klesse LJ, Velasco-Miguel S, Meyers K, Rushing EJ, Parada LF. Mouse tumor model for neurobromatosis type 1.
Science 1999;286:21769.
Vogel US, Dixon RA, Schaber MD, Diehl RE, Marshall MS, Scolnick EM, et al. Cloning of bovine GAP and its interaction with
oncogenic ras p21. Nature 1988;335:903.
Walker JA, Gouzi JY, Long JB, Huang S, Maher RC, Xia H, et al. Genetic and functional studies implicate synaptic overgrowth and
ring gland cAMP/PKA signaling defects in the Drosophila melanogaster neurobromatosis-1 growth deciency. PLoS Genet
2013;9:e1003958.
Wang S, Watanabe T, Noritake J, Fukata M, Yoshimura T, Itoh N, et al. IQGAP3, a novel effector of Rac1 and Cdc42, regulates
neurite outgrowth. J Cell Sci 2007;120:56777.
Wang T, Wei JJ, Sabatini DM, Lander ES. Genetic screens in human cells using the CRISPR-Cas9 system. Science 2014;343:804.
Wang XX, Li XZ, Zhai LQ, Liu ZR, Chen XJ, Pei Y. Overexpression of IQGAP1 in human pancreatic cancer. Hepatobiliary Pancreat
Dis Int 2013;12:5405.
Weissbach L, Settleman J, Kalady MF, Snijders AJ, Murthy AE, Yan YX, et al. Identication of a human rasGAP-related protein
containing calmodulin-binding motifs. J Biol Chem 1994;269:2051721.
Welti S, Fraterman S, DAngelo I, Wilm M, Scheffzek K. The sec14 homology module of neurobromin binds cellular glycer-
ophospholipids: mass spectrometry and structure of a lipid complex. J Mol Biol 2007;366:55162.
White CD, Brown MD, Sacks DB. IQGAPs in cancer: a family of scaffold proteins underlying tumorigenesis. FEBS Lett 2009;583:
181724.
White CD, Erdemir HH, Sacks DB. IQGAP1 and its binding proteins control diverse biological functions. Cell Signal 2012;24:82634.
White CD, Khurana H, Gnatenko DV, Li Z, Odze RD, Sacks DB, et al. IQGAP1 and IQGAP2 are reciprocally altered in hepatocellular
carcinoma. BMC Gastroenterol 2010;10:125.
Whittaker SR, Theurillat JP, Van Allen E, Wagle N, Hsiao J, Cowley GS, et al. A genome-scale RNA interference screen implicates
NF1 loss in resistance to RAF inhibition. Cancer Discov 2013;3:35062.
Wu J, Williams JP, Rizvi TA, Kordich JJ, Witte D, Meijer D, et al. Plexiform and dermal neurobromas and pigmentation are
caused by Nf1 loss in desert hedgehog-expressing cells. Cancer Cell 2008;13:10516.
Wu K, Liu J, Tseng SF, Gore C, Ning Z, Shari N, et al. The role of DAB2IP in androgen receptor activation during prostate cancer
progression. Oncogene 2014;33:195463.
Xie D, Gore C, Liu J, Pong RC, Mason R, Hao G, et al. Role of DAB2IP in modulating epithelial-to-mesenchymal transition and
prostate cancer metastasis. Proc Natl Acad Sci U S A 2010;107:248590.
14 O. Maertens, K. Cichowski / Advances in Biological Regulation 55 (2014) 114

Xu GF, Lin B, Tanaka K, Dunn D, Wood D, Gesteland R, et al. The catalytic domain of the neurobromatosis type 1 gene product
stimulates ras GTPase and complements ira mutants of S. cerevisiae. Cell 1990;63:83541.
Xu W, Mulligan LM, Ponder MA, Liu L, Smith BA, Mathew CG, et al. Loss of NF1 alleles in phaeochromocytomas from patients
with type I neurobromatosis. Genes Chromosom Cancer 1992;4:33742.
Yang XY, Guan M, Vigil D, Der CJ, Lowy DR, Popescu NC. p120Ras-GAP binds the DLC1 Rho-GAP tumor suppressor protein and
inhibits its RhoA GTPase and growth-suppressing activities. Oncogene 2009;28:14019.
Yano M, Toyooka S, Tsukuda K, Dote H, Ouchida M, Hanabata T, et al. Aberrant promoter methylation of human DAB2 interactive
protein (hDAB2IP) gene in lung cancers. Int J Cancer 2005;113:5966.
Yu J, Yu J, Rhodes DR, Tomlins SA, Cao X, Chen G, et al. A polycomb repression signature in metastatic prostate cancer predicts
cancer outcome. Cancer Res 2007;67:1065763.
Zhang H, Zhang R, Luo Y, DAlessio A, Pober JS, Min W. AIP1/DAB2IP, a novel member of the Ras-GAP family, transduces TRAF2-
induced ASK1-JNK activation. J Biol Chem 2004;279:4495565.
Zhang X, Li N, Li X, Zhao W, Qiao Y, Liang L, et al. Low expression of DAB2IP contributes to malignant development and poor
prognosis in hepatocellular carcinoma. J Gastroenterol Hepatol 2012;27:111725.
Zheng H, Chang L, Patel N, Yang J, Lowe L, Burns DK, et al. Induction of abnormal proliferation by nonmyelinating schwann cells
triggers neurobroma formation. Cancer Cell 2008;13:11728.
Zhu Y, Ghosh P, Charnay P, Burns DK, Parada LF. Neurobromas in NF1: schwann cell origin and role of tumor environment.
Science 2002;296:9202.
Zhu Y, Guignard F, Zhao D, Liu L, Burns DK, Mason RP, et al. Early inactivation of p53 tumor suppressor gene cooperating with
NF1 loss induces malignant astrocytoma. Cancer Cell 2005;8:11930.

Вам также может понравиться