You are on page 1of 10

Oncogene (2007) 26, 6386–6395

& 2007 Nature Publishing Group All rights reserved 0950-9232/07 $30.00
www.nature.com/onc

ORIGINAL ARTICLE

KIT oncoprotein interactions in gastrointestinal stromal tumors:


therapeutic relevance

M-J Zhu1,2, W-B Ou1,2, CDM Fletcher1,2, PS Cohen3, GD Demetri2,4 and JA Fletcher1,2
1
Department of Pathology, Brigham & Women’s Hospital, Boston, MA, USA; 2Departments of Pathology, Pediatrics and Medicine,
Harvard Medical School, Boston, MA, USA; 3Novartis Oncology, Florham Park, NJ, USA and 4Ludwig Center at Dana-Farber
Cancer Institute, Boston, MA, USA

Most gastrointestinal stromal tumors (GISTs) express Introduction


oncogenic and constitutively active forms of the KIT or
platelet-derived growth factor receptor alpha (PDGFRA) Gastrointestinal stromal tumors (GISTs) are the most
receptor tyrosine kinase proteins, and these kinase common mesenchymal tumors of the digestive tract.
oncoproteins serve as targets for effective therapies. Given GIST cells share morphological and immunophenotypic
that mutant KIT oncoproteins serve crucial transforming features – including expression of the KIT receptor
roles in GISTs, we evaluated interactions with the KIT tyrosine kinase (RTK) – with interstitial cells of Cajal
oncoproteins and determined signaling pathways that (ICC), which are the pacemaker cells responsible for
are dependent on KIT oncogenic activation in GISTs. coordinating gastrointestinal peristaltic activity (Hein-
Tyrosine-phosphorylated KIT oncoproteins interacted with rich et al., 2002). Most GISTs (85%) contain gain-of-
PDGFRA, PDGFRB, phosphatidylinositol 3-kinase (PI3-K) function KIT oncogenic mutations, which are associated
and PKCh in GIST cells, and these interactions were with constitutive KIT tyrosine phosphorylation, and
abolished by KIT inhibition with imatinib or PKC412 or activation of cell proliferation and survival signaling
KIT RNAi. Notably, tyrosine-phosphorylated PDGFRA pathways (Duensing et al., 2004b). Alternately, a small
was prominent in frozen GIST tumors expressing KIT subset of GISTs has oncogenic mutations in the gene
oncoproteins, suggesting that KIT-mediated PDGFRA encoding platelet-derived growth factor receptor alpha
phosphorylation is an efficient and biologically conse- (PDGFRA) that confer constitutive activation (Hein-
quential mechanism in GISTs. Activated signaling inter- rich et al., 2003; Hirota et al., 2003). Although KIT and
mediates were identified by immunoaffinity purification of PDGFRA oncogenic mutations are mutually exclusive
tyrosine-phosphorylated proteins in GIST cells before and in GISTs, approximately 30% of KIT-mutant GISTs
after treatment with KIT inhibitors, and these analyses express PDGFRA strongly (Heinrich et al., 2003).
show that GRB2, SHC, CBL and MAPK activation are Notably, the oncogenic KIT and PDGFRA mutations
largely KIT dependent in GISTs, whereas PI3-K, STAT1 can be targeted effectively with imatinib mesylate
and STAT3 activation are partially KIT dependent. In (Gleevec) in GIST cell culture models and in more than
addition, we found that phosphorylation of several 80% of GIST patients (Tuveson et al., 2001; Demetri
tyrosine kinase proteins – including JAK1 and EPHA4 et al., 2002; Verweij et al., 2004).
– did not depend on KIT activation. Likewise, paxillin KIT is a member of the PDGFR subfamily of RTK
activation was independent of the KIT oncogenic signal. proteins, which feature extracellular Ig domains and a
These studies identify signaling pathways that can provide split kinase domain (Heinrich et al., 2002). KIT under-
both KIT-dependent and KIT-independent therapeutic goes homodimerization and autophosphorylation when
synergies in GIST, and thereby highlight clinical strate- bound by the ligand, stem cell factor, or when activated
gies that might consolidate GIST therapeutic response to by oncogenic mutations (Hirota et al., 1998; Rubin
KIT/PDGFRA inhibition. et al., 2001). In addition, KIT can heterodimerize with
Oncogene (2007) 26, 6386–6395; doi:10.1038/sj.onc.1210464; the PDGFR family members FLT3 (Otto et al., 2001)
published online 23 April 2007 and PDGFRA (Hirota et al., 2003), and presumably
with other RTKs. Therefore, the clinical success of KIT
Keywords: KIT; PDGFRA; kinase; interaction; signal inhibitors in patients with GIST might result from
transduction inactivation of KIT downstream signaling pathways and
KIT heterodimerization partners.
Although most GIST patients have dramatic thera-
peutic responses to imatinib, some patients are resistant
Correspondence: Dr M-J Zhu or Dr JA Fletcher, Department of to imatinib, and very few patients exhibit a complete
Pathology, Brigham and Women’s Hospital, 75 Francis Street, Boston, response to this drug (Demetri et al., 2002). The aim of
MA 02115, USA.
E-mails: mzhu2@partners.org or jetcher@partners.org
the studies reported herein was to evaluate cell signaling
Received 1 June 2006; revised 23 October 2006; accepted 9 March 2007; pathways transmitting proliferation and survival signals
published online 23 April 2007 from KIT, and to determine which signaling intermedi-
KIT oncoprotein interactions in GIST
M-J Zhu et al
6387
ates are dependent on KIT activation in GIST. These structurally similar RTKs, the possibility of a spurious
studies, performed in human GIST cell lines before and coimmunoprecipitation, resulting from direct binding
after treatment with KIT inhibitors, interrogated of KIT by the PDGFRA antiserum, was evaluated. This
signaling proteins that play key roles in tyrosine possibility was excluded using a KIT-mutant GIST
kinase-mediated cell proliferation, survival and adhe- that expressed KIT strongly but lacked PDGFRA
sion. Mechanisms of oncogenic signaling pathway expression: KIT was not detected in the PDGFRA
activation were evaluated further by determining KIT immunoprecipitations from this GIST (data not shown).
interactions with various kinase and adaptor proteins. ABL was not demonstrably phosphorylated, and was
These studies show that certain signaling intermediates not complexed with KIT, in GIST3 or GIST4 (Figure 1).
are largely KIT dependent in GISTs, whereas others are In sum, these findings demonstrate interactions between
not. These insights will be helpful in designing rational KIT, PDGFRA and PI3-K p85, and suggest that
combination-targeted therapy strategies in GIST. ABL – despite the precedent for interaction with KIT
in human leukemias (Hallek et al., 1996) – neither
complexes substantially with KIT nor is demonstrably
Results activated in GIST.

KIT interactions in primary GISTs Kinase expression varies in GIST cell lines
Kinase protein interactions with oncogenic KIT were Variability of key kinase expression was evaluated by
evaluated in two untreated GIST specimens with hetero- immunoblotting in the GIST882, GIST48 and GIST430
zygous KIT exon 11 (GIST3) or exon 9 (GIST4) mutations cell lines. KIT, PI3-K p85 and PKCy were expressed at
(Figure 1). GIST3 and GIST4 lacked PDGFRA gene similar levels in each of these lines, whereas PDGFRA,
mutations (data not shown), but expressed phosphorylated PDGFRB and ABL expression varied considerably,
PDGFRA in addition to constitutively phosphorylated being strongest in GIST882, GIST48 and GIST430,
KIT (Figure 1). Phosphatidylinositol 3-kinase (PI3-K) respectively (Figure 2).
p85 immunoprecipitates revealed a dominant 170/
160 kD tyrosine-phosphorylated doublet in both GISTs Kinase interactions in imatinib-sensitive and imatinib-
(Figure 1), consistent in size with the transmembrane resistant GIST cell lines
forms of PDGFRA (170 kD) and KIT (160 kD). PI3-K Kinase activation and interactions were evaluated in the
p85 interactions with KIT and PDGFRA were GIST882 and GIST48 cell lines. Kinase interactions in
confirmed by immunostaining (Figure 1). These findings these cell lines were evaluated before and after
suggest that KIT and PDGFRA were the predominant pharmacologic inhibition of KIT/PDGFR by imatinib
activated RTKs in GIST3 and GIST4. In addition, in GIST882 (Figure 3) and PKC412 in the imatinib-
interaction between PDGFRA and the mature form of resistant GIST48 (Figure 4). Wild-type PDGFRA and
KIT was evident in the PDGFRA immunoprecipita- PDGFRB were phosphorylated and coprecipitated with
tions (Figure 1). Because KIT and PDGFRA are oncogenic KIT (Figures 3a and 4a), suggesting that

Figure 1 KIT-dependent kinase interactions in primary, frozen, GISTs. Internal standards are frozen GISTs, GIST1 (KIT mutant)
and GIST2 (PDGFRA mutant), which express high levels of KIT and PDGFRA, respectively. GIST3 (a) and GIST4 (b) each contain
KIT mutations: the first three lanes in each panel are total cell lysates, whereas the remaining four lanes are immunoprecipitates of KIT
(Santa Cruz, sc-168), PDGFRA (Santa Cruz, sc-338), ABL (Santa Cruz, sc-131) and PI3-K p85 (Upstate, Lake Placid, NY, USA, 06-
497), evaluating kinase activation and interactions. The PY99 immunostain shows KIT and PDGFRA activation, and suggests that
activated KIT and PDGFRA are complexed with PI3-K p85. KIT complexing with PDGFRA and PI3-K p85 is confirmed by the KIT
immunostains of PDGFRA and PI3-K p85 immunoprecipitates, respectively. PI3-K p85 complexing with KIT and PDGFRA, but not
ABL, is also shown by the PI3-K immunostains of KIT, PDGFRA and ABL immunoprecipitates, respectively.

Oncogene
KIT oncoprotein interactions in GIST
M-J Zhu et al
6388
patterns differed between GIST882 and GIST48, as can
be seen by comparing protein expression ratios in the
input (total protein) and phosphotyrosine eluate (acti-
vated protein) lanes (Figure 5). For example, GRB2,
FAK and PDGFRA were more strongly phosphory-
lated – on a per molecule basis – in GIST882 than in
GIST48 (Figure 5). However, KIT, PI3-K p85, PI3-K
p110a and SHC were strongly tyrosine phosphorylated
in both cell lines (Figure 5). In addition, there were
substantial similarities between GIST882 and GIST48
with respect to which of the protein activation events
were KIT/PDGFR dependent. PI3-K p85, PI3-K p110a,
GRB2 and SHC tyrosine phosphorylation were sub-
stantially KIT/PDGFR dependent in both cell lines,
whereas SRC-family kinases and FAK were minimally
KIT/PDGFR dependent in GIST882, and KIT/
PDGFR independent in GIST48 (Figure 5). Weak,
partially KIT dependent, tyrosine phosphorylation was
demonstrated for STAT1 (GIST48 only), STAT3,
JAK2, ABL (GIST48 only), MAPK (GIST882 only),
PKCy, CBL (GIST882 only) and HSP90. Weak, KIT
independent, tyrosine phosphorylation was demon-
strated for EPHA4, JAK1 and paxillin. However, the
SRC-family kinase data do not exclude the possibility
of functional alterations, in that shifts from SFK
Figure 2 Kinase expression in GIST cell lines with KIT oncogenic
mutations. KIT, PI3-K p85 and PKCy were expressed at similar c-terminal inhibitory tyrosine phosphorylation to inter-
levels in each of these lines, whereas PDGFRA, PDGFRB and nal activating tyrosine phosphorylation can occur with-
ABL expression varied considerably, being strongest in GIST882, out substantially changing the net phosphorylation
GIST48 and GIST430, respectively. levels. The weakly activated wild-type PDGFRB in
GIST48 was inhibited by PKC412, and the strongly
activated PDGFRA in GIST882 was inhibited by
PDGFRs heterodimerize with, and are cross-phos- imatinib (Figure 5). These findings are in keeping with
phorylated by, the constitutively activated KIT mutants the immunoprecipitation studies (Figure 3), and suggest
in GIST882 and GIST48. KIT inactivation inhibited the that PKC412 or imatinib-mediated PDGFR inhibition
complexing between KIT and PDGFR (Figures 3b and results both from direct effects and inactivation of
4b). PI3-K p85 and p110a, and PKCy also complexed heterodimerized KIT oncoproteins. It is particularly
with oncogenic KIT, as shown best in the KIT and notable that PKCy tyrosine phosphorylation was inhibi-
PKCy immunoprecipitates, respectively, and as for ted by imatinib (GIST882) and PKC412 (GIST48),
PDGFR, these interactions were partially inhibited by which is in keeping with KIT-dependent activation
imatinib (Figures 3 and 4). Likewise, PI3-K p85 of this characteristic GIST kinase (Duensing et al.,
interactions with PKCy were inhibited by imatinib 2004a).
(Figures 3b and 4b). The interaction between KIT and
PKCy appeared to be relatively weak, being most
convincing in the GIST882 cells. Interestingly, ABL Density-dependent effects on signaling protein activation
phosphorylation and interactions between KIT and The density (% confluence) of cancer cell lines, when
ABL were not detectable in either GIST882 or GIST48 grown as monolayers in vitro, has been shown to impact
(Figures 3 and 4). phosphorylation of STATs and other signaling proteins
(Vultur et al., 2004). In order to determine whether
GIST cell density impacts KIT-dependent signaling, we
KIT-dependent signaling in GIST cell lines compared phosphoprotein expression, with and without
Expression of activated signaling intermediates was KIT inhibitor treatment, in 100% confluent (‘confluent’)
evaluated by immunoblotting in phosphotyrosine vs 75% confluent (‘subconfluent’) GIST48 cells. These
immunopurifications from confluent GIST882 and studies were performed in phosphotyrosine immunoaf-
GIST48 cell lines. The rationale for evaluating signaling finity-purified fractions from the GIST cells, and showed
intermediates in confluent cells was to highlight activa- that PI3-K p85, FAK and paxillin phosphorylation were
tion events that might be strictly dependent on the KIT substantially increased in 75% confluent cells, compared
oncogenic signal, irrespective of whether the cells were to 100% confluent cells (Figure 6a and b). Indeed, FAK
actively proliferating. KIT dependence was determined phosphorylation was tenfold higher in the 75% con-
by comparing expression of each protein before and fluent cells than in 100% confluent cells (Figure 6b). The
after KIT/PDGFR inhibition by imatinib (in GIST882 FAK cell density effects were independent of KIT
cells) and PKC412 (in GIST48 cells). Phosphorylation activation, in that FAK remained strongly phosphorylated
Oncogene
KIT oncoprotein interactions in GIST
M-J Zhu et al
6389

Figure 3 Kinase activation and interactions in untreated (a) and imatinib-treated (b) GIST882 cells. Lanes 1 and 2 contain GIST882
total cell lysates, with and without imatinib treatment, respectively. Lanes 3–10 contain GIST882 lysates immunoprecipitated with
normal rabbit serum (lane 3) or antibodies to KIT (Santa Cruz, sc-168; lane 4), PDGFRA (Santa Cruz, sc-338; lane 5), ABL (Santa
Cruz, sc-131; lane 6), PI3-K p85 (Upstate, 06-497; lane 7), PDGFRB (Santa Cruz, sc-432; lane 8), PKCy (Santa Cruz, sc-1875; lane 9)
and PI3-K p110a (Cell Signaling #4254; lane 10). The PY99 and KIT immunostains show activation-dependent complexing of KIT
with PDGFRA, PDGFRB, PKCy and PI3-K. Complexing between PI3-K p85 and p110a, as expected, is not KIT dependent.

in subconfluent GIST48 after PKC412 treatment. By only the smaller, immature, cytoplasmic, KIT form
contrast, the accentuated PI3-K p85 and paxillin (KIT N-terminal polyclonal rabbit antibody, Santa
phosphorylation in 75% confluent GIST48 was KIT Cruz, Santa Cruz, CA, USA, sc-5535) or both KIT
dependent, as shown after PKC412-mediated KIT forms (mouse monoclonal antibody, Santa Cruz, sc-
inactivation, which reduced the phosphoprotein expres- 13508). These studies show that PDGFRA, PI3-K and
sion to levels found in the 100% confluent GIST48 GRB2 complex preferentially with mature KIT (Figure 7).
(Figure 6a and b). These studies were corroborated, in Interestingly, both immature and mature KIT coimmu-
total cell lysates of GIST48 at four different cell noprecipitated with PI3-K and PKCy, whereas only the
densities, showing stronger FAK Y397 and paxillin mature forms of KIT and PDGFRA coimmunoprecipi-
Y118 phosphorylation at lower cell density (Figure 6c). tated (Figures 1, 3a and 7). These findings suggest that
CBL and PKCy were also more strongly phosphory- the cytoplasmic PI3-K and PKCy proteins might
lated – and in a KIT-dependent manner – at 75% modulate all forms of KIT signaling, whereas PDGFRA
confluence, whereas PI3-K p110a and JAK1 were more primarily modulates mature KIT signaling.
strongly phosphorylated in 100% confluent cells (Figure
6a and b). STAT, JAK2 and HSP90 tyrosine phosphory- KIT RNAi corroboration of small molecule KIT-inhibitor
lation were unaffected by GIST48 cell density (Figure 6a assays
and b). Because imatinib and PKC412 are multikinase inhibi-
tors, and therefore not specific for KIT, we used KIT
Preferential interactions with the mature form of KIT RNAi methods to confirm key findings from the studies
Interactions with the two major KIT forms (migrating, (described above) on KIT-dependent GIST cell signal-
in relationship to the molecular size markers in this ing mechanisms. KIT knockdown was accomplished by
study, at 160 and 145 kD, but also described tradition- infecting GIST882 cells with lentiviral shRNA vectors,
ally as migrating at 145 and 125 kD) were evaluated in and corroborated that PI3-K p85 and CBL activa-
immunoprecipitations with KIT antibody recognizing tion were KIT dependent (Figure 8a). KIT-mediated
Oncogene
KIT oncoprotein interactions in GIST
M-J Zhu et al
6390

Figure 4 Kinase activation and interactions in untreated (a) and PKC412-treated (b) GIST48 cells. Lanes 1 and 2 contain GIST48
total cell lysates, with and without PKC412 treatment, respectively. Lanes 3–10 contain GIST48 lysates immunoprecipitated with
normal rabbit serum (lane 3) or antibodies to KIT (Santa Cruz, sc-168; lane 4), PDGFRA (Santa Cruz, sc-338; lane 5), ABL (Santa
Cruz, sc-131; lane 6), PI3-K p85 (Upstate, 06-497; lane 7), PDGFRB (Santa Cruz, sc-432; lane 8), PKCy (Santa Cruz, sc-1875; lane 9)
and PI3-K p110a (Cell Signaling, Danvers, MA, USA, #4254; lane 10). The PY99 and KIT immunostains show activation-dependent
complexing of KIT with PKCy and PI3-K. Complexing between PI3-K p85 and p110a, as expected, is not KIT dependent.

activation of PDGFRA was evaluated by PDGFRA levels, comparable to those in PDGFRA-mutant GISTs
immunostaining of anti-phosphotyrosine immunopreci- (Heinrich et al., 2003). Previous studies demonstrate
pitates (Figure 8a) and by phosphotyrosine immunos- that PDGFRA oncoproteins expressed by transfected
taining of PDGFRA immunoprecipitates (Figure 8b), PDGFRA mutants in vitro can bind with and cross-
and both of these studies showed decreased PDGFRA activate wild-type KIT (Hirota et al., 2003). Herein, we
activation after shRNA KIT knockdown. Thus, these demonstrate interactions and evidence for cross-activa-
findings are consistent with oncogenic KIT-mediated tion between oncogenic KIT and wild-type PDGFRA
PDGFRA cross-activation in GIST cells. proteins in human GIST tissues. Complexing between
KIT and PDGFRA was shown in frozen samples of
primary human GISTs as well as in the GIST882
cell line (Figures 1 and 3). The KIT and PDGFRA
Discussion complexes were dependent on activation of the KIT
oncoproteins, as evidenced by disruption of the com-
KIT and PDGFRA mutational activation events are plexes after GIST882 exposure to imatinib (Figure 3). In
mutually exclusive in GISTs, but these alternate addition, the studies in frozen primary GISTs shed light
oncogenic tyrosine kinase mechanisms have similar on the nature of KIT oncogenic signaling complexes in
biologic and transforming consequences (Heinrich untreated GISTs, and reveal that another known
et al., 2003). Although KIT expression is a diagnostic imatinib target – PDGFRA – complexes with and is
hallmark of the majority of GIST cells, the small subset likely coactivated by KIT oncoproteins in GIST. These
of GISTs with PDGFRA oncoproteins feature uncha- findings were confirmed by KIT RNAi knockdown
racteristically low – and even undetectable – KIT assays, in which KIT shRNA-mediated inhibition
expression (Heinrich et al., 2003; Medeiros et al., resulted in decreased PDGFRA activation (Figure 8),
2004). On the other hand, approximately 30% of KIT- underscoring that PDGFRA cross-activation can be
mutant GISTs express wild-type PDGFRA at high mediated by KIT oncoproteins in GISTs. These studies
Oncogene
KIT oncoprotein interactions in GIST
M-J Zhu et al
6391

Figure 5 (a–d) KIT/PDGFR-dependent activation of kinase signaling proteins in GIST882 (with and without KIT/PDGFR
inhibition by imatinib) and GIST48 (with and without KIT/PDGFR inhibition by PKC412). Total cell lysates (lanes 1 and 4 in each
blot) and tyrosine-phosphorylated proteins immunoaffinity purified from the cell lysates (lanes 2 and 3 in each blot) were
immunostained with antibodies (specified in Materials and methods) to evaluate KIT-dependent activation of various signaling
proteins. KIT activation (large arrows in PY99 stains) was substantially suppressed after inhibitor treatment in both cell lines, as was
PDGFR activation. Treatment-mediated KIT/PDGFR inhibition was accompanied by dephosphorylation of PI3-K p85, PI3-K
p110a, GRB2, SHC, PKCy, STATs, MAPK, CBL and HSP90.

show how the KIT oncogenic signal can be amplified to associations between KIT and PDGFRB in GIST
involve other RTKs in GIST, and suggest that imatinib (Figures 3 and 4). However, our studies do not show
therapeutic efficacy might be based, in part, on interactions between ABL and KIT, and nor was ABL
inhibition of both the KIT oncoprotein and hetero- demonstrably activated in the GIST frozen tumors and
dimerization (PDGFRA) partners. cell lines (Figures 1, 3 and 5). These findings suggest that
Interestingly, the interactions between KIT and ABL, although a classical imatinib target and
PDGFRA involved predominantly the mature, trans- a KIT-interacting protein in hematological models
membrane forms of these kinases (Figures 1 and 7). (Hallek et al., 1996), does not modulate or enhance
Therefore, KIT:PDGFRA interactions, in GIST, likely the KIT oncogenic signal in GISTs.
result from heterodimerization – or other complexing – PI3-K pathways play crucial roles in transmitting
at the cell surface, with resultant activation of canonical survival and proliferation signals from various RTK
RTK signaling cascades. The GIST882 kinase interaction oncoproteins (Herbst et al., 1995; Sordella et al., 2004).
studies, performed in confluent cultures, demonstrate In addition, multiprotein complexes involving KIT,
KIT:PDGFRA interactions under basal unstimulated PI3-K and PDGFRB have been demonstrated after
conditions in GIST. Similarly, our studies revealed weak transfection of KIT and PDGFRB constructs in 293
Oncogene
KIT oncoprotein interactions in GIST
M-J Zhu et al
6392

Figure 6 (a–c) Cell density effects on KIT/PDGFR-dependent activation of kinase signaling proteins in GIST48, with and without
KIT/PDGFR inhibition by PKC412. As in Figure 5, total cell lysates (lanes 1 and 4 in a and b) and immunoaffinity-purified tyrosine-
phosphorylated proteins (lanes 2 and 3 in a and b) were immunostained to evaluate KIT-dependent activation of various signaling
proteins. Tyrosine phosphorylation was evaluated in cells harvested from 100% confluent (images from Figure 5) vs 75% confluent
cultures (a and b). PI3-K p85, FAK and Paxillin were more strongly phosphorylated in 75% confluent cultures, PI3-K p110a and
JAK1 were more strongly phosphorylated in 100% confluent cultures. In the 75% confluent cultures, the accentuated phosphorylation
of PI3-K p85 and paxillin, but not FAK, is KIT dependent. STAT, JAK2 and HSP90 phosphorylation were unaffected by culture
confluence. Increased FAK and paxillin phosphorylation, in subconfluent cultures, was confirmed in GIST48 total cell lysates
immunostained with antibodies to phospho-FAK and phospho-paxillin (c). These studies showed that FAK and paxillin tyrosine
phosphorylation were reduced in 100% confluent cultures, compared to the subconfluent cultures.

cells (Herbst et al., 1995). In the present studies, the p85 protein kinase C (PKC) and subsequently shown to
regulatory and p110 catalytic PI3-K subunits were inhibit other kinases including VEGFR2, PDGFR and
constitutively associated with activated KIT and PDG- KIT (Fabbro et al., 2000), with activity in some
FRA in GIST frozen tumors and cell lines (Figures 1, 3 imatinib-resistant GIST mutations (Debiec-Rychter
and 7). These associations were largely abrogated after et al., 2005). The phosphotyrosine eluate studies
KIT inhibition by imatinib or PKC412 (Figures 3b and confirmed the immunoprecipitation assays (Figures 1
4b). PI3-K activation was partially KIT dependent and 3), showing imatinib/PKC412 inhibition of KIT,
(Figures 5 and 6) and was regulated by cell density PDGFRA and PI3-K (Figure 5). Comparisons between
(Figure 6). total cell lysates and phosphotyrosine purifications
To characterize KIT/PDGFR-dependent signaling (Figure 5) showed that KIT, PI3-K (regulatory and
pathways in GISTs, we used an efficient phosphoty- catalytic subunits), SHC and GRB2 were strongly
rosine eluate approach combining phosphotyrosine tyrosine phosphorylated in a KIT-dependent manner
affinity purification with KIT kinase inhibition in cell in both GIST882 and GIST48 cell lines. These findings
lines GIST882 and GIST48. This approach enabled are consistent with KIT oncogenic activation of the
efficient immunoblot evaluations of KIT/PDGFR- RAS/RAF/MAPK and PI3-K/AKT pathways, which
dependent tyrosine phosphorylation in a panel of are regulated by activation of GRB2/SHC and PI3-K,
signaling intermediates, in a single immunoblotting respectively (Tauchi et al., 1994; Blume-Jensen et al.,
experiment. KIT was inhibited by imatinib treatment 1998). Recruitment of GRB2 and SHC adaptor proteins
in GIST882, and by PKC412 treatment in the imatinib- to RTK-derived oncogenes has been shown to be
resistant GIST48 cell line. The staurosporine derivative sufficient for morphological cell transformation and
PKC412 was originally identified as an inhibitor of experimental metastases (Saucier et al., 2002), whereas
Oncogene
KIT oncoprotein interactions in GIST
M-J Zhu et al
6393
PI3-K appears to play a particularly crucial role in GRB2 and SHC might serve as alternate therapeutic
tyrosine kinase oncoprotein signaling (Sordella et al., targets, whose inhibition could be beneficial in GIST
2004). Therefore, our findings suggest that PI3-K, patients resistant to imatinib.
Our present studies corroborate previous reports that
STAT activation, in GISTs, is relatively weak and only
partially dependent on the activated KIT oncoprotein
(Duensing et al., 2004b). This situation differs from
the KIT kinase-domain mutants in mast cell disease,
where STAT is activated strongly and crucial to KIT
transforming activity (Ning et al., 2001). In addition,
whereas STAT activation is regulated by cell density in a
carcinoma model (Vultur et al., 2004), being more
strongly activated in subconfluent cultures than in
confluent cultures, we did not observe marked increase
in STAT activation in subconfluent GIST cultures.
The involvement of cell adhesion proteins has not
been evaluated previously in GISTs, and our studies
show that the FAK and paxillin adhesion-related
proteins are activated in a cell density-dependent
manner. That is, activation of both FAK and paxillin
activation was strongest in subconfluent, actively pro-
liferating, cultures. FAK activation was KIT indepen-
dent in both confluent and subconfluent cultures
(Figures 5 and 6), whereas paxillin activation was KIT
independent in confluent cultures and partially KIT
dependent in subconfluent cultures. These findings
suggest that imatinib impact on adhesion proteins, in
clinical GISTs, might vary in different parts of the same
tumor, depending on the local architecture and cell
Figure 7 PDGFRA, PI3-K p85 and GRB2 interactions with
density. Notably, the KIT-independent nature of FAK
mature vs immature KIT oncoproteins. GIST882 lysates were activation, in these GIST models, suggests that FAK
immunoprecipitated with antibodies recognizing immature KIT therapeutic inhibition might provide clinical synergies
only (Santa Cruz, sc-5535) vs both immature and mature KIT for KIT inhibition by imatinib.
(Santa Cruz, sc-13508). The immunostains show that PDGFRA,
PI3-K p85 and GRB2 interact predominantly with the mature KIT
The novel PKC family member, PKCy, is an
oncoprotein. intriguing biomarker in GIST, given its narrow range
of expression in normal cells, and preliminary evidence

Figure 8 Kinase activation in GIST882 cells after infection with lentiviral empty vector (lanes 1 and 3) vs KIT shRNA (lanes 2 and 4).
In each panel, lanes 1 and 2 are total cell lysates, whereas lanes 3 and 4 are immunoprecipitates evaluating kinase activation.
Immunostains of the PY99 (Santa Cruz, sc-7020) immunoprecipitates (a) showed that KIT shRNA infections reduced the expression
of total and phospho-KIT to 50% of control levels, resulting in 35, 51 and 52% reductions in tyrosine-phosphorylated PI3-K p85, CBL
and PDGFRA, respectively. Protein expression quantitation was performed using a FUJI LAS1000 þ digital capture system.
Similarly, phosphotyrosine immunostaining of PDGFRA immunoprecipitates (b) demonstrated 60% reduction in PDGFRA
phosphorylation, after KIT shRNA knockdown.

Oncogene
KIT oncoprotein interactions in GIST
M-J Zhu et al
6394
for a positive regulatory role in KIT oncogenic signaling pCMVDR8.91 and pMD.G helper virus packaging plasmids
in GIST (Duensing et al., 2004a). We show, for the first (at a 10:10:1 ratio) into 293T cells. These transfections were
time, that PKCy associates with KIT oncoproteins in performed using lipofectamine and PLUS reagent, and
GIST cell lines (Figure 3), and is tyrosine phosphory- lentivirus supernatants were harvested at 24, 36, 48 and 60 h.
Viral titers were determined in GIST882 cells, according to a
lated in a KIT-dependent manner, particularly in
protocol from Invitrogen (Carlsbad, CA, USA). GIST882 cells
subconfluent cells (Figures 5 and 6). Strong PKCy were infected, in the presence of 8 mg/ml of polybrene, and
expression has been reported – in adult tissues – only in were then lysed for immunoprecipitation and western blot
T cells and ICC, and in the leukemias and GISTs that analysis after 20 days of selection with 2.5 mg/ml puromycin.
derive, respectively, from these cell lineages. Therefore,
it is possible that PKCy inhibition might be a minimally
toxic and highly effective therapy for GIST. Our Immunoprecipitation and western blot analysis
preliminary studies with PKCy inactivation or RNAi Immunoprecipitation and immunoblotting methods were
essentially as described, and are detailed in the Supplementary
knockdown (Hubert, Ou and Fletcher, unpublished) do
methods. The chemiluminescence signals were captured and
indeed suggest that PKCy expression and activation are quantified using a FUJI LAS1000plus system with Science Lab
crucial to GIST cell survival and proliferation. 2001 ImageGauge 4.0 software (Fujifilm Medical Systems,
Stamford, CT, USA).

Materials and methods


Phosphotyrosine affinity purification
Reagents The phosphotyrosine fraction was isolated from GIST cells
Antibodies for immunoprecipitation and immunoblotting are using a minicolumn loaded with 1 ml of anti-phosphotyrosine-
described in the Supplementary methods. sepharose beads (Zymed, South San Francisco, CA, USA).
The anti-phosphotyrosine affinity columns were first washed
ten times with phosphate-buffered saline containing 0.02%
Tissue specimens and cell lines
sodium azide followed by ten washes with cell lysis buffer. The
GIST tissue specimens and cell lines are described in the
columns were then incubated at 41C overnight with 10 mg of
Supplementary methods.
GIST protein lysate, washed seven times with lysis buffer, and
the bound tyrosine-phosphorylated proteins were then eluted
Functional studies by loading the column ten times with 1 ml of 100 mM
Biochemical correlates for KIT inhibition were evaluated in phenylphosphate. The phosphotyrosine protein eluates were
GIST882 and GIST48 cells after incubation for 4 h in serum- dialysed to remove the phenylphosphate, then evaluated by
free media with 2 mM imatinib and 1 mM PKC412 (provided by immunoblotting, using 10% of the eluates per gel lane.
Novartis Pharma, Basel, Switzerland), respectively.

KIT shRNA studies Acknowledgements


A KIT lentiviral shRNA (short hairpin RNA) was obtained
from Dr William Hahn (Dana-Farber Cancer Institute and We thank Yongchun Zhou for critical review of the article and
Broad Institute RNAi consortium). This shRNA was as- valuable discussions, Chang-Jie Chen, Anette Duensing and
sembled by ligating KIT forward [50 -CCGGCCATAAGGTTT Maureen Thyne for useful discussions and technical assistance.
CGTTTCTGTACTCGAGTACAGAAACGAAACCTTATGG This work was supported by grants from an anonymous
TTTTTG-30 ] and reverse [50 -AATTCAAAAACCATAAGGTTT donor, the Life Raft Group, Cesarini Team for the Pan-
CGTTTCTGTACTCGAGTACAGAAACGAAACCTTATGG Massachusetts Challenge, the Virginia and Daniel K Ludwig
-30 ] oligomers into the AgeI and EcoRI sites of a pLKO.1puro Trust for Cancer Research, the Ronald O Perelman Fund for
lentiviral vector. Lentivirus was produced by cotransfecting Cancer Research, the Stutman GIST Cancer Research Fund,
pLKO.1puro empty vector or pLKO.1puro-KIT (shRNA), the Rubenstein Foundation and Leslie’s Links.

References

Blume-Jensen P, Janknecht R, Hunter T. (1998). The kit gastrointestinal stromal tumors (GISTs). Cancer Res 64:
receptor promotes cell survival via activation of PI 3-kinase 5127–5131.
and subsequent Akt-mediated phosphorylation of Bad on Duensing A, Medeiros F, McConarty B, Joseph NE,
Ser136. Curr Biol 8: 779–782. Panigrahy D, Singer S et al. (2004b). Mechanisms of
Debiec-Rychter M, Cools J, Dumez H, Sciot R, Stul M, oncogenic KIT signal transduction in primary gastrointes-
Mentens N et al. (2005). Mechanisms of resistance to tinal stromal tumors (GISTs). Oncogene 23: 3999–4006.
imatinib mesylate in gastrointestinal stromal tumors and Fabbro D, Ruetz S, Bodis S, Pruschy M, Csermak K, Man A
activity of the PKC412 inhibitor against imatinib-resistant et al. (2000). PKC412 – a protein kinase inhibitor with a
mutants. Gastroenterology 128: 270–279. broad therapeutic potential. Anticancer Drug Des 15: 17–28.
Demetri GD, von Mehren M, Blanke CD, Van den Abbeele Hallek M, nhauser-Riedl S, Herbst R, Warmuth M, Winkler
AD, Eisenberg B, Roberts PJ et al. (2002). Efficacy and A, Kolb HJ et al. (1996). Interaction of the receptor tyrosine
safety of imatinib mesylate in advanced gastrointestinal kinase p145c-kit with the p210bcr/abl kinase in myeloid
stromal tumors. N Engl J Med 347: 472–480. cells. Br J Haematol 94: 5–16.
Duensing A, Joseph NE, Medeiros F, Smith F, Hornick JL, Heinrich MC, Corless CL, Duensing A, McGreevey L, Chen CJ,
Heinrich MC et al. (2004a). Protein Kinase C theta Joseph N et al. (2003). PDGFRA activating mutations
(PKCtheta) expression and constitutive activation in in gastrointestinal stromal tumors. Science 299: 708–710.
Oncogene
KIT oncoprotein interactions in GIST
M-J Zhu et al
6395
Heinrich MC, Rubin BP, Longley BJ, Fletcher JA. (2002). Rubin BP, Singer S, Tsao C, Duensing A, Lux ML, Ruiz R
Biology and genetic aspects of gastrointestinal stromal et al. (2001). KIT activation is a ubiquitous feature of
tumors: KIT activation and cytogenetic alterations. Hum gastrointestinal stromal tumors. Cancer Res 61: 8118–8121.
Pathol 33: 484–495. Saucier C, Papavasiliou V, Palazzo A, Naujokas MA, Kremer
Herbst R, Shearman MS, Jallal B, Schlessinger J, Ullrich A. R, Park M. (2002). Use of signal specific receptor tyrosine
(1995). Formation of signal transfer complexes between stem kinase oncoproteins reveals that pathways downstream from
cell and platelet-derived growth factor receptors and SH2 Grb2 or Shc are sufficient for cell transformation and
domain proteins in vitro. Biochemistry 34: 5971–5979. metastasis. Oncogene 21: 1800–1811.
Hirota S, Isozaki K, Moriyama Y, Hashimoto K, Nishida T, Sordella R, Bell DW, Haber DA, Settleman J. (2004).
Ishiguro S et al. (1998). Gain-of-function mutations of c-kit in Gefitinib-sensitizing EGFR mutations in lung cancer acti-
human gastrointestinal stromal tumors. Science 279: 577–580. vate anti-apoptotic pathways. Science 305: 1163–1167.
Hirota S, Ohashi A, Nishida T, Isozaki K, Kinoshita K, Tauchi T, Feng GS, Marshall MS, Shen R, Mantel C, Pawson
Shinomura Y et al. (2003). Gain-of-function mutations of T et al. (1994). The ubiquitously expressed Syp phosphatase
platelet-derived growth factor receptor alpha gene in gastro- interacts with c-kit and Grb2 in hematopoietic cells. J Biol
intestinal stromal tumors. Gastroenterology 125: 660–667. Chem 269: 25206–25211.
Medeiros F, Corless CL, Duensing A, Hornick JL, Oliveira Tuveson DA, Willis NA, Jacks T, Griffin JD, Singer S,
AM, Heinrich MC et al. (2004). KIT-negative gastrointest- Fletcher CD et al. (2001). STI571 inactivation of the
inal stromal tumors: proof of concept and therapeutic gastrointestinal stromal tumor c-KIT oncoprotein: biologi-
implications. Am J Surg Pathol 28: 889–894. cal and clinical implications. Oncogene 20: 5054–5058.
Ning ZQ, Li J, Arceci RJ. (2001). Signal transducer and Verweij J, Casali PG, Zalcberg J, LeCesne A, Reichardt P,
activator of transcription 3 activation is required for Blay JY et al. (2004). Progression-free survival in gastro-
Asp(816) mutant c-Kit-mediated cytokine-independent sur- intestinal stromal tumours with high-dose imatinib: rando-
vival and proliferation in human leukemia cells. Blood 97: mised trial. Lancet 364: 1127–1134.
3559–3567. Vultur A, Cao J, Arulanandam R, Turkson J, Jove R,
Otto KG, Jin L, Spencer DM, Blau CA. (2001). Cell Greer P et al. (2004). Cell-to-cell adhesion modulates Stat3
proliferation through forced engagement of c-Kit and activity in normal and breast carcinoma cells. Oncogene 23:
Flt-3. Blood 97: 3662–3664. 2600–2616.

Supplementary Information accompanies the paper on the Oncogene website (http://www.nature.com/onc).

Oncogene

You might also like