metkinase-dependentlossofthee3ligasecblingastric cancer s · proline-rich region and a uba domain...

13
Met Kinase-dependent Loss of the E3 Ligase Cbl in Gastric Cancer * S Received for publication, January 5, 2012 Published, JBC Papers in Press, January 18, 2012, DOI 10.1074/jbc.M112.339820 Andrea Z. Lai ‡§ , Michael Durrant § , Dongmei Zuo , Colin D. H. Ratcliffe ‡§ , and Morag Park ‡§¶1 From the Goodman Cancer Research Centre and Departments of § Biochemistry, Medicine, and Oncology, McGill University, Montreal, Quebec H3A 1A3, Canada Background: Dysregulated Met signaling can promote tumorigenesis. Results: Active Met kinase promotes loss of Cbl protein. This is rescued upon inhibition of Met kinase. Conclusion: Met-dependent Cbl loss in gastric cancers releases other Cbl targets, such as the EGF receptor, from Cbl-mediated attenuation. Significance: Uncoupling of Met and other RTKs from Cbl negative regulation in gastric cancers provides a mechanism for enhanced RTK signaling in cancer. Strict regulation of signaling by receptor tyrosine kinases (RTKs) is essential for normal biological processes, and disrup- tion of this regulation can lead to tumor initiation and progres- sion. Signal duration by the Met RTK is mediated in part by the E3 ligase Cbl. Cbl is recruited to Met upon kinase activation and promotes ubiquitination, trafficking, and degradation of the receptor. The Met RTK has been demonstrated to play a role in various types of cancer. Here, we show that Met-dependent loss of Cbl protein in MET-amplified gastric cancer cell lines repre- sents another mechanism contributing to signal dysregulation. Loss of Cbl protein is dependent on Met kinase activity and is partially rescued with a proteasome inhibitor, lactacystin. Moreover, Cbl loss not only uncouples Met from Cbl-mediated negative regulation but also releases other Cbl targets, such as the EGF receptor, from Cbl-mediated signal attenuation. Thus, Met-dependent Cbl loss may also promote cross-talk through indirect enhancement of EGF receptor signaling. Receptor tyrosine kinases (RTKs) 2 are key components of signaling cascades that promote cellular proliferation and dif- ferentiation. These processes are essential for normal human embryogenesis and homeostasis and, when dysregulated, can drive the unconstrained cell growth and invasiveness charac- teristic of many human cancers. The Met RTK was initially identified as an oncogenic fusion protein (Tpr-Met) following treatment of the human osteo- genic sarcoma (HOS) cell line with the carcinogen N-methyl- N-nitro-nitrosoguanidine (1, 2). Met was subsequently identi- fied as the receptor for hepatocyte growth factor (HGF) or scatter factor (3, 4). HGF-Met signaling promotes scatter and an invasive morphogenic response in epithelial cells in cell cul- ture (5) and is required during embryogenesis for development of the placenta, liver, kidney, and neuronal and skeletal muscles (6). In the adult, the HGF-Met signaling axis is involved in wound healing and liver regeneration (7, 8). Tight regulation of Met signaling is required for many of these processes, and dys- regulation of the Met signaling axis has been implicated in var- ious human cancers. Several mechanisms leading to dysregulation of Met in can- cer have been identified. These include autocrine/paracrine activation, Met overexpression, genomic amplification, point mutation, and alternative splicing (9). MET amplification occurs frequently in gastric cancers (10) and to a lesser extent in non-small cell lung cancer and glioblastomas (11–14). The loss of negative regulation represents an additional mechanism through which oncogenic activation of Met can occur (15). Negative regulation of Met is primarily mediated through the Cbl E3 ligase. The Cbl family of E3 ligases consists of three mammalian homologues: c-Cbl, Cbl-b, and Cbl-3 (16 – 18). These cytoplasmic proteins are conserved in their N-ter- minal halves and consist of a tyrosine kinase binding (TKB) domain, a linker region, and a RING domain, the latter of which is required for functional E3 ligase activity (reviewed in Ref. 19). The C-terminal portions are less well conserved and include a proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). The UBA domains of both c-Cbl and Cbl-b facilitate dimeriza- tion, but only the Cbl-b UBA domain is able to bind ubiquitin (20 –22). The presence of key tyrosine residues as well as pro- line-rich regions allows Cbl proteins to function also as scaf- folds capable of recruiting a number of SH2 and SH3 domain- containing proteins (19). Both c-Cbl and Cbl-b act as E3 ligases and ubiquitinate their target substrates (reviewed in Ref. 23). The overlap of c-Cbl and Cbl-b function is evident, as CBL / or CBLB / mice are both viable, but mice deficient in both are embryonic lethal (23, 24). Similarly, in osteoclasts, the depletion of both proteins is required to disrupt the microtubule network and induce apo- * This work was supported by a Canada Graduate Scholarship (CGS) doctoral research award (to A. L.), a Cancer Consortium grant (to M. D.), and an oper- ating grant (to M. P.), all from the Canadian Institutes of Health Research (CIHR). S This article contains supplemental Figs. 1–7. 1 Holds the Diane and Sal Guerrera Chair in Cancer Genetics at McGill Univer- sity. To whom correspondence should be addressed: Goodman Cancer Ctr., 1160 Ave. Des Pins Ouest, Cancer Pavilion Room 511, Montreal, Que- bec H3A 1A3, Canada. Tel.: 514-398-5749; Fax: 514-398-6769; E-mail: [email protected]. 2 The abbreviations used are: RTK, receptor tyrosine kinase; HGF, hepatocyte growth factor; TKB, tyrosine kinase binding; SH2 and -3, Src homology 2 and 3; EGFR, EGF receptor; DMSO, dimethyl sulfoxide; UBA, ubiquitin-associated. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 287, NO. 11, pp. 8048 –8059, March 9, 2012 © 2012 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A. 8048 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 287 • NUMBER 11 • MARCH 9, 2012 by guest on June 20, 2019 http://www.jbc.org/ Downloaded from

Upload: lamtu

Post on 21-Jun-2019

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

Met Kinase-dependent Loss of the E3 Ligase Cbl in GastricCancer*□S

Received for publication, January 5, 2012 Published, JBC Papers in Press, January 18, 2012, DOI 10.1074/jbc.M112.339820

Andrea Z. Lai‡§, Michael Durrant§, Dongmei Zuo‡, Colin D. H. Ratcliffe‡§, and Morag Park‡§¶�1

From the ‡Goodman Cancer Research Centre and Departments of §Biochemistry, ¶Medicine, and �Oncology, McGill University,Montreal, Quebec H3A 1A3, Canada

Background: Dysregulated Met signaling can promote tumorigenesis.Results: Active Met kinase promotes loss of Cbl protein. This is rescued upon inhibition of Met kinase.Conclusion:Met-dependent Cbl loss in gastric cancers releases other Cbl targets, such as the EGF receptor, fromCbl-mediatedattenuation.Significance: Uncoupling of Met and other RTKs from Cbl negative regulation in gastric cancers provides a mechanism forenhanced RTK signaling in cancer.

Strict regulation of signaling by receptor tyrosine kinases(RTKs) is essential for normal biological processes, and disrup-tion of this regulation can lead to tumor initiation and progres-sion. Signal duration by the Met RTK is mediated in part by theE3 ligase Cbl. Cbl is recruited toMet upon kinase activation andpromotes ubiquitination, trafficking, and degradation of thereceptor. The Met RTK has been demonstrated to play a role invarious types of cancer. Here, we show thatMet-dependent lossof Cbl protein inMET-amplified gastric cancer cell lines repre-sents another mechanism contributing to signal dysregulation.Loss of Cbl protein is dependent on Met kinase activity andis partially rescued with a proteasome inhibitor, lactacystin.Moreover, Cbl loss not only uncouples Met from Cbl-mediatednegative regulation but also releases other Cbl targets, such asthe EGF receptor, from Cbl-mediated signal attenuation. Thus,Met-dependent Cbl loss may also promote cross-talk throughindirect enhancement of EGF receptor signaling.

Receptor tyrosine kinases (RTKs)2 are key components ofsignaling cascades that promote cellular proliferation and dif-ferentiation. These processes are essential for normal humanembryogenesis and homeostasis and, when dysregulated, candrive the unconstrained cell growth and invasiveness charac-teristic of many human cancers.The Met RTK was initially identified as an oncogenic fusion

protein (Tpr-Met) following treatment of the human osteo-genic sarcoma (HOS) cell line with the carcinogen N-methyl-

N�-nitro-nitrosoguanidine (1, 2). Met was subsequently identi-fied as the receptor for hepatocyte growth factor (HGF) orscatter factor (3, 4). HGF-Met signaling promotes scatter andan invasive morphogenic response in epithelial cells in cell cul-ture (5) and is required during embryogenesis for developmentof the placenta, liver, kidney, and neuronal and skeletal muscles(6). In the adult, the HGF-Met signaling axis is involved inwound healing and liver regeneration (7, 8). Tight regulation ofMet signaling is required for many of these processes, and dys-regulation of the Met signaling axis has been implicated in var-ious human cancers.Several mechanisms leading to dysregulation of Met in can-

cer have been identified. These include autocrine/paracrineactivation, Met overexpression, genomic amplification, pointmutation, and alternative splicing (9). MET amplificationoccurs frequently in gastric cancers (10) and to a lesser extent innon-small cell lung cancer and glioblastomas (11–14).The loss of negative regulation represents an additional

mechanism through which oncogenic activation of Met canoccur (15). Negative regulation of Met is primarily mediatedthrough the Cbl E3 ligase. The Cbl family of E3 ligases consistsof threemammalian homologues: c-Cbl, Cbl-b, and Cbl-3 (16–18). These cytoplasmic proteins are conserved in their N-ter-minal halves and consist of a tyrosine kinase binding (TKB)domain, a linker region, and a RINGdomain, the latter of whichis required for functional E3 ligase activity (reviewed in Ref. 19).The C-terminal portions are less well conserved and include aproline-rich region and a UBA domain (c-Cbl and Cbl-b) (19).The UBA domains of both c-Cbl and Cbl-b facilitate dimeriza-tion, but only the Cbl-b UBA domain is able to bind ubiquitin(20–22). The presence of key tyrosine residues as well as pro-line-rich regions allows Cbl proteins to function also as scaf-folds capable of recruiting a number of SH2 and SH3 domain-containing proteins (19).Both c-Cbl and Cbl-b act as E3 ligases and ubiquitinate their

target substrates (reviewed in Ref. 23). The overlap of c-Cbl andCbl-b function is evident, asCBL�/� orCBLB�/�mice are bothviable, but mice deficient in both are embryonic lethal (23, 24).Similarly, in osteoclasts, the depletion of both proteins isrequired to disrupt the microtubule network and induce apo-

* This work was supported by a Canada Graduate Scholarship (CGS) doctoralresearch award (to A. L.), a Cancer Consortium grant (to M. D.), and an oper-ating grant (to M. P.), all from the Canadian Institutes of Health Research(CIHR).

□S This article contains supplemental Figs. 1–7.1 Holds the Diane and Sal Guerrera Chair in Cancer Genetics at McGill Univer-

sity. To whom correspondence should be addressed: Goodman CancerCtr., 1160 Ave. Des Pins Ouest, Cancer Pavilion Room 511, Montreal, Que-bec H3A 1A3, Canada. Tel.: 514-398-5749; Fax: 514-398-6769; E-mail:[email protected].

2 The abbreviations used are: RTK, receptor tyrosine kinase; HGF, hepatocytegrowth factor; TKB, tyrosine kinase binding; SH2 and -3, Src homology 2and 3; EGFR, EGF receptor; DMSO, dimethyl sulfoxide; UBA,ubiquitin-associated.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 287, NO. 11, pp. 8048 –8059, March 9, 2012© 2012 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A.

8048 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 287 • NUMBER 11 • MARCH 9, 2012

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from

Page 2: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

ptosis (25). However, differences in c-Cbl and Cbl-b functionexist. c-Cbl and Cbl-b are recruited to EGFR at temporally dis-tinct periods subsequent to EGF stimulation, where c-Cbl isrecruited early (5–15 min) and Cbl-b is recruited by 1 h post-stimulation (26).Moreover, whereas c-Cbl or Cbl-b ubiquitina-tion of its substrates, such as RTKs, primarily promotes degra-dation, Cbl-b-targeted ubiquitination of some substrates, suchas the p85 subunit of phosphatidylinositol 3-kinase, impingeson protein-protein interactions but leaves overall protein levelsunchanged (reviewed in Ref. 23), highlighting the potentiallydistinct roles of Cbl proteins.Upon Met kinase activation, intracellular tyrosine residues

are phosphorylated, creating docking sites for a number ofdownstream signalingmolecules. Phosphorylation of Tyr-1003in the Met juxtamembrane domain allows for direct recruit-ment of Cbl through theCbl TKBdomain (27). Cbl recruitmentto Met is also mediated indirectly through Grb2 (5, 27, 28).Upon Cbl recruitment, Met is ubiquitinated (27). This is re-quired for efficient recognition of Met during trafficking andsubsequent degradation in the lysosome (29, 30). The uncou-pling of Met from Cbl, through substitution of the Cbl bindingsite Tyr-1003 with a phenylalanine residue, results in a Metreceptor that is poorly ubiquitinated, exhibits enhanced stabil-ity and prolonged phosphorylation, and is transforming in vitroand in vivo (29).Moreover, Tpr-Met, a truncated, constitutivelyactive, cytoplasmic variant of Met, lacks the juxtamembraneregion containing Tyr-1003, does not recruit the Cbl TKBdomain, is not ubiquitinated, and fails to enter the endocyticdegradative pathway (31). This escape from entry into the deg-radative pathway may represent a common mechanism thatcontributes to the oncogenic activation of many RTKs follow-ing chromosomal reorganization (15).The importance of Cbl-mediated negative regulation of Met

as amechanism counteracting tumorigenesis is further empha-sized by the identification of naturally occurringMet variants incancers that lack the Cbl binding site. Alternatively splicedmutants ofMet that result in the excision of exon 14 containingthe Cbl TKB domain binding site (Tyr-1003) have been identi-fied in both non-small cell lung cancer cell lines and adeno-carcinoma lung tumors (32–34). These Met variants showenhanced stability and prolonged signaling and oncogeniccapacity (33). In addition, the gastric cancer cell line Hs746Tamplifies MET with a mutation that results in the loss of exon14 (35). Hence, the loss of negative regulation by Cbl may beselected for even whenMET is amplified.Here, we show that conditions in whichMET is amplified in

human gastric cancers leads to the loss of Cbl protein. Thisreflects another mechanism through which Met is able touncouple from Cbl-dependent negative regulation. Moreover,a loss of Cbl would not only enhance signaling by Met but hasthe capability of dysregulating the signaling by otherCbl targetssuch as EGFR. This represents a mechanism of RTK cross-talkin human tumors whereby Cbl loss is dependent onMet kinaseactivity.

EXPERIMENTAL PROCEDURES

Antibodies and Reagents—Antibody 148 was raised in rabbitagainst a C-terminal peptide of human Met (36). Met DL-21

antibody was purchased from Upstate Biotechnology (LakePlacid, NY). c-Cbl, Cbl-b, Src, and ubiquitin (P4D1) antibodieswere acquired from Santa Cruz Biotechnology (Santa Cruz,CA). Met (AF276) and c-Cbl antibodies used for immunofluo-rescence were obtained fromR&D Systems (Minneapolis, MN)and Epitomics Inc. (Burlingame, CA). Actin and tubulin anti-bodies were obtained from Sigma-Aldrich. Phospho-specificMet Tyr 1234/1235, EGFR Tyr 992, the general phosphoty-rosine pTyr-100, K4B-specific polyubiquitin (D9D5), and totalEGFR antibodies were purchased fromCell Signaling Technol-ogy (Mississauga, Ontario, Canada). HA.11 monoclonal andphospho-Src tyrosine 418 antibodieswere obtained fromCova-nce (Berkeley, CA) and Invitrogen, respectively. �-Cateninantibody was purchased from BD Biosciences.HGF was a generous gift from Genentech (San Francisco,

CA), and EGF was purchased from Roche Diagnostics. Conca-namycin, lactacystin, and PP2 were purchased from EMDChemicals (Gibbstown, NJ) and utilized at final concentrationsof 0.1, 10, and 10 �M, respectively. The Met inhibitor PHA-665752 was a kind gift from Pfizer (final concentration 0.1 �M).Cell Culture and Transient Transfections—HEK 293, Oka-

jima, and MKN45 cells were cultured in Dulbecco’s modifiedEagle’s medium with 10% fetal bovine serum (FBS). Snu-5 andKATO II cell lines (a kind gift from Dr. Daniel Haber) werecultured in RPMI supplemented with 10% FBS. Transienttransfections with HEK 293 cells were performed using Lipo-fectamine Plus reagent according to the manufacturer’s proto-col (Invitrogen).Immunoprecipitation and Western Blotting—HEK 293,

Okajima, MKN45, Snu-5, and KATO II cells were harvestedin TGH lysis buffer (50 mMHEPES, pH 7.5, 150 mMNaCl, 1.5mM MgCl2, 1 mM EGTA, 1% Triton X-100, 10% glycerol, 1mM phenylmethylsulfonyl fluoride, 1 mM sodium fluoride,1 mM sodium vanadate, 10 �g/ml aprotinin, and 10 �g/mlleupeptin). HEK 293 transfections were harvested at 48 hpost-transfection. Lysates were incubated with antibody for1 h at 4 °C with gentle rotation followed by a 1-h incubationwith protein A- or G-Sepharose beads. Captured proteinswere collected by washing three times in TGH lysis buffer,eluted by boiling in SDS sample buffer, resolved by SDS-PAGE, and transferred to a nitrocellulose membrane. Mem-branes were blocked in 3% bovine serum albumin in TBST(10 mM Tris, pH 8.0, 150 mM NaCl, 2.5 mM EDTA, and 0.1%Tween 20) for 1 h and incubated with primary antibodies inTBST overnight at 4 °C. Membranes were then incubatedwith secondary antibodies in TBST for 1 h. After threewashes with TBST, the bound proteins were visualized withan ECL detection kit (Amersham Biosciences).ForWestern blot analysis of c-Cbl and Cbl-b ubiquitination,

cells were harvested in 150�l of denaturingTSD lysis buffer (50mM Tris-HCl, pH 7.5, 1% SDS, 5 mM DTT, 50 �M MG132, 50mM N-ethylmaleimide, 1 mM phenylmethylsulfonyl fluoride,1 mM sodium fluoride, 1 mM sodium vanadate, 10 �g/ml apro-tinin, and 10 �g/ml leupeptin). Lysates were boiled at 99 °C for5 min and then pelleted in a microcentrifuge at 10,000 rpm for5 min. One hundred microliters of supernatant was thendiluted to a final concentration of 0.1% SDSwith TNESV buffer(50mMTris-HCl, pH7.5, 1%Nonidet P-40, 100mMNaCl, 2mM

Met Kinase-dependent Cbl Loss in Gastric Cancer

MARCH 9, 2012 • VOLUME 287 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 8049

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from

Page 3: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

EDTA, 10 mM N-ethylmaleimide, 1 mM phenylmethylsulfonylfluoride, 1 mM sodium fluoride, 1 mM sodium vanadate, 10�g/ml aprotinin, and 10 �g/ml leupeptin). Immunoprecipita-tions were performed as described above but with TNESVbuffer used in place of TGH lysis buffer.For blots that required quantitation, membranes were

blocked with Li-COR blocking buffer (Li-COR Biosciences)and incubated with primary antibodies as described above fol-lowed by incubation with infrared (IR)-conjugated secondaryantibodies prior to detection and analysis on the Odyssey IRimaging system (Li-COR Biosciences).Generation of Cbl-b Constructs—Site-directed mutagenesis

was performed using the QuikChange kit (Stratagene, La Jolla,CA) according to the manufacturer’s instructions to create theCbl-b constructs with the following primers (and their comple-mentary primers): Cbl-b-(1–483) (5�-CACCAGTCACATC-ACCAGGATAGTCTCCCCTTGCCCAGAGAAG-3�); -(1–541) (5�-GTGAGAAAACAAGATTAACCACTCCCAGCAC-CAC-3�); -(1–603) (5�-GTTTGGGACTAATCAGTAAGTG-GGATGTCGACTC-3�); -(1–664) (5�-GATGCCCTCCCTC-CATCTCTCTGACCTAGGCCACCTCCTGCAAGGCATA-GTC-3�); and -(1–821) (5�-CACCTTGGAAGTGAAGAATA-AGATCTTCCTCCCCGGCTTTCTCCTCCTCC-3�). Cbl-b9PA and Cbl-b-(1–664) 9PA constructs were made with theQuikChangeTM multi site-directed mutagenesis kit (Strat-agene) as per the manufacturer’s instructions with the follow-ing primers: 5�-GACCCACTCCAGATCCCACATCTAAGC-CTGGCACCCGTGGCTCCTGCCCTGGATCTAATTCAG-AAAGGCATAGTTAG-3�; 5�-GTGAGAAAACAAGATAA-ACCACTCCCAGCAGCACCTCCTGCCTTAGCAGATCC-TCCTCCACCGCCACCTGAAAGACCTCC-3�; and 5�-GCAGCACCTCCTGCCTTAGCAGATCCTCCTGCACCG-CCGGCTGAAGCACCTCCACCAATCCCACCAGACAAT-AGACTG-3�.siRNA—MKN45 or KATO II cells were plated at a confluency

of 3� 105 cells/well in a 6-well dish and transiently transfected insuspension with 20 nM humanMETON-TARGETplus SMART-pool siRNA, 50 nM humanCBL siGENOMESMARTpool siRNA,50 nMhumanCBLB siGENOMESMARTpool siRNA, or an equalconcentration of scrambled control siRNApurchased fromDhar-macon (Lafayette, CO) with polyethylenimine (PEI) (Sigma-Aldrich). PEI was utilized at a final concentration of 3.75 �g/ml.48 h after initial transfection, cells were retransfected as describedpreviously. Cells were then lysed 72 h after the initial transfection.Quantitative analysis of knockdown was assessed using theLi-COROdyssey infrared imaging system as described previously.Quantitative Real-time PCR—Total cellular RNA was ex-

tracted using TRIzol reagent (Invitrogen) following the manu-facturer’s protocol. 800 ng of RNAwas reverse-transcribed andamplified as described previously (29). The genes 60 S acidicribosomal protein P0 (RPLP0), TATA-box-binding protein(TBP), splicing factor, arginine/serine-rich 4 (SFRS4), and actin(ACTB) were used with geNorm software (37) to generate thenormalization factors for c-Cbl and Cbl-b mRNA levels. Theprimer sequences were as follows: RPLP0 (sense, 5�-CTCAAC-ATCTCCCCCTTCTC-3�; antisense, 5�-GACTCGTTTGTA-CCCGTTGA-3�); TBP (sense, 5�-TCAGGAAGACGACGTA-ATGG-3�; antisense, 5�-TTACAGAAGGGCATCACCTG-3�);

SFRS4 (sense, 5�-GTTTGGTAGCCGTAGCACAA-3�; anti-sense, 5�-TGTGGTCATTCCAGCCTTAG-3�); ACTB (sense,5�-ATCCCCCAAAGTTCACAATG-3�; antisense, 5�-GTGG-CTTTTAGGATGGCAAG-3�); CBL (sense, 5�-TCGGCTCC-AGAAATTCATTC-3�; antisense, 5�-CCCTGAAGCCATCA-ATCAGT-3�); and CBLB (sense, 5�-CCAAGAGATGAAGGC-TCCAG-3�; antisense, 5�-CTGGTGAGTTCTGCCTGTCA-3�). Real-time PCR was performed using LightCycler 480 andLightCycler 480 SYBRGreen IMaster (Roche Applied Science)as per the manufacturer’s instructions. Measurements weretaken in duplicate, three times for each set of samples, and threesets of samples (biological replicates) were measured. The levelof c-Cbl or Cbl-b mRNA is expressed as the mean -fold differ-ence normalized to the level of c-Cbl or Cbl-b mRNA in theDMSO control.Confocal Immunofluorescence Microscopy—MKN45 cells

were seeded at 1.6 � 105 on glass coverslips (Bellco Glass Inc.,Vineland, NJ) in 24-well plates (NalgeneNunc, Rochester, NY).Forty-eight hours later, cells were washed once with PBS andthen fixed with 2% paraformaldehyde (Fisher Scientific) in PBSfor 20 min followed by washing three times in PBS. Residualparaformaldehyde was removed with three 5-min washes with100 mM glycine in PBS. Cells were permeabilized with 0.2%Triton X-100/PBS and blocked for 30 min with blocking buffer(2% bovine serum albumin, 0.2% Triton X-100, 0.05% Tween20, and PBS). Coverslips were incubated with primary and sec-ondary antibodies diluted in blocking buffer for 1 h and 40min,respectively, at room temperature, and nuclei were counter-stained with 4�,6-diamidino-2-phenylindole. Coverslips weremounted with Immu-Mount (Thermo Shandon Inc., Pitts-burgh, PA). Confocal images were taken using a Zeiss 510Metalaser scanning confocal microscope (Carl Zeiss, Canada Ltd.,Toronto,Ontario, Canada)with an�100 objective. Image anal-ysis was carried out using the LSM 5 image browser (EmpixImaging, Mississauga, Ontario, Canada).Co-localization Studies—Quantification of co-localization

betweenMet and c-Cbl or Met and Cbl-b was performed usingMetaMorph software for object-based co-localization mea-surements. The results were logged into Excel for analysis.

RESULTS

Cbl Protein Levels Are Elevated following Inhibition of MetKinase—To establish whether amplification of MET in theOkajima, MKN45, Snu-5, and KATO II gastric cancer cell linesleads to constitutive activation of Met protein (10), proteinsfrom whole cell lysates were immunoblotted with Met andphospho-Met antibodies (Fig. 1, A and B). The phospho-Metantibody is specific for tyrosines 1234 and 1235 in the activationloop of the kinase domain.High levels ofMet and phospho-Metwere observed in all of the cell lines tested.To examine the status of Cbl proteins, lysates were immuno-

blotted with antibodies raised to the C-terminal regions ofc-Cbl and Cbl-b. Whereas the c-Cbl antibody is selective forimmunoprecipitation and immunoblotting of c-Cbl (supple-mental Fig. 1), someweak cross-reactivity of theCbl-b antibodywith c-Cblwas observedwhen c-Cblwas overexpressed in tran-sient transfections (supplemental Fig. 1). However, in the gas-tric cancer cell lines used, the Cbl-b antibody detected only

Met Kinase-dependent Cbl Loss in Gastric Cancer

8050 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 287 • NUMBER 11 • MARCH 9, 2012

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from

Page 4: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

Cbl-b, as established by the difference in molecular mass ofc-Cbl (110 kDa) when compared with Cbl-b (125 kDa).Immunoblotting with c-Cbl and Cbl-b antibodies demon-

strated low levels of c-Cbl protein in all gastric cancer cell linestested as well as variable levels of Cbl-b (Fig. 1, A and B). Inter-estingly, when Met kinase activity was inhibited with a smallmoleculeMet kinase inhibitor (0.1 �M PHA-665752) (38) for 4,8, or 16 h, a pronounced increase in c-Cbl protein levels wasobserved in all cell lines, and amoderate increase inCbl-b levelswas observed in two of the four cell lines (Snu-5 and KATO II)(Fig. 1,A and B). PHA-665752 is a small molecule inhibitor thatspecifically targets Met, and inhibition of unrelated kinasesoccurs only at an IC50 � 2.5 �M (38). Quantitation of c-Cbl andCbl-b protein levels from three experimental replicates re-vealed an inverse correlation between Met kinase activity andc-Cbl or Cbl-b levels in each cell line (supplemental Fig. 2).To determine whether the change in Cbl protein levels, upon

the addition of Met inhibitor, occurs through enhanced tran-

scription, RNA from cells treated or not withMet inhibitor wasisolated and reverse-transcribed, and the c-Cbl andCbl-b levelswere determined using quantitative real-time PCR. At 16 hpost-treatment with Met inhibitor, a time at which c-Cbl pro-tein levels were maximally elevated, no significant increases inc-Cbl mRNA were observed (Fig. 1, C and D), when comparedwith the DMSO control. Hence, differences in Cbl protein levelupon Met inhibitor treatment were not due to an increase inCBL transcription.To confirm that the increase in Cbl protein level was Met-

dependent, and not due to off-target effects of the inhibitor,knockdown of Met protein was executed with siRNA in KATOII cells (Fig. 1E). Upon depletion of Met protein, c-Cbl levelsincreased (Fig. 1E), confirming the dependence of Cbl proteinloss on Met.As c-Cbl and Cbl-b can heterodimerize (21, 22), we assessed

whether this might play a role in their stability. To addresswhetherCbl-b influences c-Cbl stability and vice versa, c-Cbl or

FIGURE 1. c-Cbl protein levels are dependent on Met kinase activity. A, Okajima and MKN45 gastric cancer cell lines were treated with the Met inhibitor (0.1�M PHA-665752) for 4, 8, and 16 h. An equal volume of DMSO was used as a vehicle control. Whole cell lysates were then immunoblotted with antibodies forthe specified proteins. B, Snu-5 and KATO II gastric cancer cell lines were treated as described for A. C and D, total RNA from the 16-h time point was isolated fromeach of the cell lines, and quantitative real-time PCR was performed to ascertain levels of c-Cbl and Cbl-b mRNA. E, Met protein levels were depleted throughtransient transfection of Met siRNA for 72 h. Lysates were immunoblotted with antibodies for the specified proteins. F, the level of Met protein, normalized toactin levels, was quantified from three independent experiments. Plotted here is the mean � S.E. *, statistical significance of p � 0.03 was determined usingStudent’s t test.

Met Kinase-dependent Cbl Loss in Gastric Cancer

MARCH 9, 2012 • VOLUME 287 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 8051

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from

Page 5: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

Cbl-b protein levels were depleted with targeted siRNA. Nei-ther Cbl-b nor c-Cbl knockdown significantly altered steady-state stability levels of the other protein (supplemental Fig. 3).Cbl Levels are Independent of Src Kinase Activity—Overex-

pression of activated Src kinase has been shown to lead to adecrease inCbl protein levels (39). Src phosphorylation has alsobeen demonstrated downstream of activated Met kinase (40).To test whether Met-dependent Cbl loss requires Src, cellswere treated with the Src inhibitor PP2 (10�M), aMet inhibitor(0.1 �M PHA-665752), or both. In gastric cancer cells withamplifiedMET, Src kinase is constitutively active, as evident bythe basal tyrosine phosphorylation of Src Tyr-418 (Fig. 2).Interestingly, treatment with the Met inhibitor did not detect-ably decrease Src tyrosine phosphorylation, and inhibition ofSrcwith PP2, as evident by the loss of Tyr-418 phosphorylation,did not significantly alterMet tyrosine phosphorylation (Fig. 2).Thus, the basal Src activation observed in these gastric cancercell lines appears independent of Met kinase activity. Impor-tantly, the level of c-Cbl or Cbl-b protein did not increase uponinhibition of Src kinase and was elevated only following inhibi-tion of Met kinase (Fig. 2). Moreover, inhibition of both Metand Src kinases increased the amount of Cbl proteins to a levelcomparable, but not exceeding, that with Met inhibitor alone.Hence, together these data support the conclusion that in thesegastric cancer cell lines, Cbl protein levels are dependent on thekinase activation status of Met and not Src.Transient Overexpression of Met Promotes Loss of Cbl-b—To

elucidate the requirements through which Met promotes lossof Cbl in gastric cancer cell lines, a transient transfection andstructure-function approach was undertaken. Met was initiallyexpressed at increasing concentrations with either HA-taggedc-Cbl or Cbl-b (Fig. 3A). In HEK 293 cells, overexpression ofMet protein alone is sufficient to activate Met kinase (Fig. 3A)(36) and constitutive phosphorylation of Met. Increasing levelsof Met expression resulted in a significant decrease in Cbl-bprotein levels, whereas c-Cbl protein levels were largely unaf-fected under these conditions (Fig. 3A). Tpr-Met, the trun-

cated, constitutively active, cytoplasmic variant of Met, alsoinduced a dramatic loss in Cbl-b protein upon transient co-ex-pression (Fig. 3B). As expected, the overexpression of eitherc-Cbl or Cbl-b promoted an efficient loss of total Met proteinand a corresponding decrease in phospho-Met (pTyr-1234/1235) levels (Fig. 3A) (27). However, neither c-Cbl nor Cbl-boverexpression promotes loss of Tpr-Met, as Tpr-Met lacksTyr-1003, the binding site for the Cbl TKB domain (27) (Fig.3B).Quantitation of c-Cbl and Cbl-bmRNA levels in the absence

and presence of Met show that Cbl-b mRNA levels are notdecreased upon transient co-expression withMet (supplemen-tal Fig. 4A). Thus, this finding supports our data in gastric can-cer cell lines, demonstrating that a decrease in Cbl protein lev-els in the presence of activeMet protein is not due to alterationsin the level mRNA. Co-expression of either c-Cbl or Cbl-b withincreasing levels of EGFR or activated Src (Src Y527F) resultedin a decrease in both c-Cbl and Cbl-b protein levels (Fig. 4, Cand D), corroborating previously published data (39, 41).Because transient Met expression promoted a significant andconsistent loss of Cbl-b protein levels, structure-function stud-ies were carried out with Cbl-b.Met-dependent Cbl-b Loss Requires Met Kinase Activity and

Intact Cbl-b TKB and RING Domains—As a means to validatethe requirement for Met kinase activity in promoting Cbl loss,wild-type Met and a mutant unable to promote transfer of the�-ATP (kinase-dead K1110A) constructs were expressed aloneor with HA-tagged Cbl-b. Co-expression of wild-type Met andCbl-b resulted in a loss of bothMet and Cbl-b protein (Fig. 4A).However, this loss was not observed upon co-expression ofMetK1110Awith Cbl-b (Fig. 4A), demonstrating the dependence ofCbl protein loss on Met kinase activity.To determine the regions or domains of Cbl-b required for

Cbl-b loss, a panel of HA-tagged Cbl-b mutants (Fig. 4B) con-sisting of wild-type Cbl-b, a Cbl-b unable to bind Met throughits TKB domain (G298E), an E3 ligase-dead mutant (Cbl-bC373A), and multiple truncation mutants that excise the UBA

FIGURE 2. Met-dependent Cbl loss is independent of Src kinase activity. Each of the gastric cell lines (A, Okajima and MKN45; B, Snu-5 and KATO II) wastreated with nothing, DMSO (vehicle control), 0.1 �M Met inhibitor PHA-665752 (PHA), 10 �M Src inhibitor (PP2), or both PHA-665752 and PP2 for 16 h. Wholecell lysates were then immunoblotted with antibodies for the specified proteins.

Met Kinase-dependent Cbl Loss in Gastric Cancer

8052 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 287 • NUMBER 11 • MARCH 9, 2012

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from

Page 6: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

domain (Cbl-b �UBA), proline-rich regions, and/or key tyro-sine residues (Tyr-665/Tyr-709) (Cbl-b-(1–483), -(1–541),-(1–603), -(1–664), and -(1–821)) were examined for their sta-bility when expressed in HEK 293 cells alone or with wild-typeMet. With the exception of the G298E and C373A mutants, inthe presence of Met, the protein levels of each Cbl-b constructwere decreased as compared with their expression levels in theabsence of Met (Fig. 4C). Cbl-b G298E, which abrogates TKBfunction, uncoupling Met-Cbl-b direct interaction, and Cbl-bC373A, which lacks functional E3 ligase activity in the RINGdomain, did not exhibit protein loss upon co-expression withMet, indicating a necessity for TKB and RING function in pro-moting Cbl-b loss downstream from Met (Fig. 4C). In supportof this observation, when c-Cbl or Cbl-b were transientlyexpressed in HEK 293 cells withMet and FLAG-tagged ubiqui-tin, increased ubiquitination of Cbl-b, as compared with c-Cbl,was observed, an effect that was enhanced upon treatment withthe proteasome inhibitor lactacystin prior to cell lysis (supple-mental Fig. 5). The low level of c-Cbl ubiquitination followingtransient transfection substantiates the lack of c-Cbl proteinloss, when compared with Cbl-b, upon overexpression withMet.Cbl-b is in part recruited toMet indirectly through the Grb2

adaptor protein (27). The requirement for indirect associationof Met and Cbl-b (via Grb2) for Met-dependent Cbl-b loss wasalso assessed. TheC-terminal half of Cbl-b contains six putativeGrb2 binding sites. Three of these sites were absent in Cbl-b-(1–664), and the remaining three sites were mutated throughsite-directed mutagenesis (Cbl-b-(1–664) 9PA). This con-struct, as demonstrated through GST-Grb2 pulldown (Fig. 4F),is dramatically impaired in its ability to bind Grb2 as comparedwith wild-type Cbl-b. The protein levels of Cbl-b-(1–664) 9PAdecrease in the presence of Met (Fig. 4E), demonstrating thatrecruitment of Grb2 is dispensable for Cbl-b protein loss. Toour knowledge, this is the first demonstration of these threeGrb2 binding sites onCbl-b. Altogether, this demonstrates thatthe C-terminal UBA domain on Cbl-b, as well as the proline-

rich regions and tyrosines 665 and 709, is dispensable for Met-dependent Cbl-b protein loss after transient transfection. Fur-thermore, as Cbl-b ubiquitination was observed and Cbl-b E3ligase activity is required, these data support the conclusionthat that Cbl-b loss may occur through self-ubiquitination.Met-dependentCbl Loss Requires ProteasomeActivity—Basal

activation of Met kinase in the gastric cancer cell lines corre-lates with low levels of Cbl proteins. This suggests that Metsignaling may target Cbl protein for degradation. To testwhether Cbl loss was induced in response to Met activity,HEK 293 cells, which stably express a doxycycline-inducibleMet, were transiently transfected with HA-Cbl-b and stimu-lated with HGF (supplemental Fig. 6A). By 2 h of HGF stimula-tion, Cbl-b levels decreased when compared with the unstimu-lated control (supplemental Fig. 6A). To determine whetherCbl-b protein was being degraded through the lysosome or theproteasome, inhibitors (concanamycin or lactacystin, respec-tively) for both were used. Cells were pretreated with concana-mycin or lactacystin for 4 h prior toHGF stimulation. The addi-tion of concanamycin did not affect Cbl-b loss with HGFstimulation, whereas the presence of lactacystin partially res-cuedCbl-b levels (supplemental Fig. 6A). In Snu-5 andKATOIIcells, lactacystin treatment resulted in an increase in c-Cbl lev-els, as shown in Fig. 5, A and B. Quantitation of three indepen-dent experiments demonstrates that upon Met inhibition,c-Cbl protein levels increased 2.5-fold (Fig. 5C). The same trendwas observed in Okajima and MKN45 cells (supplemental Fig.6B). Altogether, these data implicate a role for the proteasomein c-Cbl degradation downstream from kinase-active Met.Considering that c-Cbl protein levels are increased upon

inhibition of the proteasome, we assessed whether c-Cbl isubiquitinated under basal conditions in gastric cancer cells. Tothis end, KATO II cells were treated overnight (for 16 h) withMet inhibitor followed by treatment with lactacystin for 8 h.Subsequent to cell lysis, c-Cbl proteins were immunoprecipi-tated under denaturing conditions to uncouple the associatedproteins, and ubiquitination was detected using anti-ubiquitin

FIGURE 3. Loss of Cbl protein upon co-expression with tyrosine kinases. c-Cbl and Cbl-b constructs were expressed in HEK 293 cells alone or with increasingamounts of Met (A), Tpr-Met (B), EGFR (C), or activated Src (Y527F) (D). Lysates were immunoblotted with antibodies specific to HA (c-Cbl or Cbl-b), Met, pMet,EGFR, pEGFR, Src, or tubulin as indicated.

Met Kinase-dependent Cbl Loss in Gastric Cancer

MARCH 9, 2012 • VOLUME 287 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 8053

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from

Page 7: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

antibodies (P4D1 and antibodies raised to Lys-48-linked poly-ubiquitin (D9D5)). c-Cbl is ubiquitinated in the presence ofactiveMet, which is diminished following the inhibition ofMet,even though c-Cbl protein levels are elevated (Fig. 5, F and G).The comparable patterns of total ubiquitin and Lys-48-linkedpolyubiquitin indicate that c-Cbl is polyubiquitinated and con-tains Lys-48-linked chains (Fig. 5F). As Lys-48-linked chains arethought to promote proteasomal degradation, and c-Cbl ubiq-uitination is most apparent where Met is active and following

treatment with lactacystin (Fig. 5F), we concluded that Metactivation promotes c-Cbl polyubiquitination and subsequentdegradation by the proteasome.In gastric cancer cells, c-Cbl co-immunoprecipitates with

Met and is tyrosine-phosphorylated, both of which events aredecreased in the presence of Met inhibitor and are hencedependent on Met kinase activity (Fig. 5E). In comparison,although Cbl-b can co-immunoprecipitate with Met in gastriccancer cell lines, Cbl-b tyrosine phosphorylation was not

FIGURE 4. Met-dependent Cbl-b loss requires Met kinase activity and Cbl-b TKB and RING function. A, wild-type and kinase-dead Met (K1110A) weretransiently transfected alone or with wild-type Cbl-b in HEK 293 cells. Cells were lysed 48 h after transfection and lysates probed for the specified proteins. B, aschematic of the structure of the different HA-tagged Cbl-b constructs used. HA-tagged c-Cbl is also presented for comparison. Both proteins are highlyhomologous in their TKB (for Cbl specificity), linker (L), and RING domains. The TKB domain is composed of three subdomains: a 4-helix bundle, a calcium-binding EF hand, and a variant SH2 domain, all of which contribute to phosphotyrosine binding. The linker region contains tyrosines that may be phosphor-ylated to enhance E3 ligase activity of the RING domain. The C-terminal portion is more variable between c-Cbl and Cbl-b and allows both proteins to act asscaffold proteins, as it includes tyrosine residues (Tyr-700, Tyr-731, and Tyr-770 for c-Cbl; Tyr-665 and Tyr-709 for Cbl-b) and proline-rich regions that recruit SH2and SH3 domain-containing proteins. The UBA domain is able to facilitate homo- and heterodimerization of c-Cbl and Cbl-b but is also functionally distinctbetween the two proteins. The Cbl-b UBA domain is able to bind ubiquitin moieties, whereas the c-Cbl UBA domain is not. C, this panel of Cbl-b constructs wasexpressed alone or with Met. Whole cell lysates were immunoblotted with antibodies specific to HA (Cbl-b), Met, or actin as designated. The molecular weightladder is indicated to the left of the HA panel. D, a schematic depicting the domains and mutations (where applicable) of the different HA-tagged Cbl-bconstructs. The Cbl-b 9PA construct has nine prolines, in putative Grb2 binding sites, mutated to alanines. E, wild-type, truncated, or mutated Cbl-b constructswere expressed alone or with Met. Membranes were probed with the specified antibodies, and molecular weight markers are shown to the left of the HA panel.F, lysates from HEK 293 cells where various Cbl-b constructs or Met were overexpressed were incubated with GST or GST-Grb2 proteins conjugated toglutathione-Sepharose beads. Met was used as a positive control, and all of the Cbl constructs shown, with the exception of Cbl-b-(1– 664) 9PA, are associatedwith the GST-Grb2 protein. WCL, whole cell lysate.

Met Kinase-dependent Cbl Loss in Gastric Cancer

8054 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 287 • NUMBER 11 • MARCH 9, 2012

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from

Page 8: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

observed (supplemental Fig. 6C). In support of the enhancedassociation of c-Cbl withMet, c-Cbl co-localizes withMet bothat the membrane and in punctate structures in MKN45 gastriccancer cells (supplemental Fig. 7B). Moreover, we observed64% co-localization of c-Cbl protein with Met by immunofluo-rescence as compared with only 34% of the Cbl-b protein (sup-plemental Fig. 7A). Hence, the enhanced association ofMet andc-Cbl compared with Cbl-b in gastric cancer cells, as observedby immunofluorescence, likely contributes to the enhancedubiquitination and decreased stability of c-Cbl over Cbl-b pro-tein levels.EGFR Degradation and Cbl Recruitment Are Impaired in

MET-amplified Gastric Cancer Cell Lines—Cbl proteins nega-tively regulate multiple receptor tyrosine kinases includingEGFR (reviewed in Ref. 42). Hence, to investigate whetherMet-dependent Cbl loss in gastric cancer cell lines augments thestability and/or signaling of other kinases, the stability of EGFRwas assessed inMKN45 cells upon EGF stimulation in the pres-ence or absence of Met inhibitor (Fig. 6A). Although stimula-tion with EGF promotes EGFR protein loss, EGFR loss is statis-

tically significant only in the presence of the Met inhibitor,when Cbl levels are elevated, and not in control cells (Fig. 6B).To establish whether the enhanced degradation of EGFR in

MKN45 cells in the presence of EGF and the Met inhibitorcorrespondswith increased association betweenCbl andEGFR,the ability of Cbl and EGFR to co-immunoprecipitate wasinvestigated. Under basal conditions in which Met is active,c-Cbl co-immunoprecipitates with a phospho-protein corre-sponding to the molecular weight of Met, which disappearsupon treatment with the Met inhibitor (Fig. 6C). Cbl-b alsoco-immunoprecipitateswith activeMet (Fig. 6D). Interestingly,upon stimulationwith EGF, phospho-EGFR predominantly co-immunoprecipitates with Cbl-b (Fig. 6, C and D). Notably, thisEGF-dependent association of Cbl-b and EGFR is significantlyenhanced whenMet kinase activity is inhibited (Fig. 6D). c-Cblalso co-immunoprecipitates with EGFR following stimulationwith EGF, and although this is enhanced following inhibition ofMet, this co-immunoprecipitation is consistently less that withCbl-b (Fig. 6C). Hence, under conditionswhereMet is inhibitedand Cbl protein levels are increased, Cbl is able to more effi-

FIGURE 5. c-Cbl protein is polyubiquitinated and its loss is dependent on proteasome activity. A, Snu-5 cells were treated with inhibitors for the lysosome(0.1 �M concanamycin (Con)) or proteasome (10 �M lactacystin (Lac)) for 8 h. Lysates were immunoblotted as specified. B, KATO II cells were treated withlysosome (0.1 �M concanamycin), proteasome (10 �M lactacystin), or Met inhibitor (0.1 �M PHA-665752 (PHA)) or both the proteasome and Met inhibitors for8 h. Lysates were probed with the designated antibodies. C, quantitation of Cbl protein levels (mean � S.E.) under the different conditions plotted here.D, whole cell lysates of KATO II cells treated with DMSO or 0.1 �M PHA-665752 overnight (16 h) followed by the addition of DMSO or 10 �M lactacystin (8 h) priorto cell lysis. Membranes were probed with the designated antibodies. E, lysates from D were immunoprecipitated (IP) with c-Cbl antibody and blotted asspecified. The molecular weight marker is shown at the left of each panel. F, KATO II lysates were immunoprecipitated with c-Cbl antibody and probed with thespecified total (P4D1) and polyubiquitin (K48 poly Ub) antibodies. G, ubiquitination of c-Cbl (normalized to amounts of c-Cbl immunoprecipitated) wasquantified from five independent experiments, and the mean � S.E. is plotted here.

Met Kinase-dependent Cbl Loss in Gastric Cancer

MARCH 9, 2012 • VOLUME 287 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 8055

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from

Page 9: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

ciently target EGFR for degradation due to enhanced associa-tion with EGFR and decreased competition for Cbl bindingfromMet.

DISCUSSION

Protein tyrosine kinases act as molecular switches to controla variety of cellular signals, and their dysregulation contributesto many human malignancies. Notably, members of the Cblfamily can serve as negative regulators for many tyrosinekinases, and loss of Cbl-dependent negative regulation is rec-ognized as amechanism that contributes to tumorigenesis (43).Here we have demonstratedMet-dependent loss of Cbl proteinas a mechanism to decrease Cbl-mediated negative regulationof RTKs in human cancer. Although other reports have detailedthe loss of Cbl protein downstream from EGFR and Src kinases(39, 41, 44), this is the first report to demonstrate that Cbl pro-tein loss is dependent on an activated Met kinase in humantumors.The mechanism of Cbl loss downstream fromMet is depen-

dent on an intact Cbl TKB domain and RING domain. This isdistinct from Src-dependent Cbl loss, which occurs indepen-dently of the Cbl RING domain (Table 1) (39). Furthermore,Src-dependent Cbl loss requires tyrosines Tyr-700, Tyr-731,and Tyr-774 in the C-terminal half of Cbl (39, 45), of which theequivalent tyrosines (Tyr-665 and Tyr-709) are dispensable forMet-dependent Cbl-b loss (Fig. 4). In support of this, we havedemonstrated that Met-dependent c-Cbl or Cbl-b loss in mul-tiple gastric tumor cell lines is independent of Src kinase activ-ity. In amanner similar toMet, Cbl-b loss downstream from the

EGFR also requires both intact Cbl TKB and RING domainsand not C-terminal proline-rich or UBA regions (41).Inhibition of the proteasome rescues Cbl levels downstream

from Met kinase. This supports the requirement for an intactCbl E3 ligase function whereby Cbl is auto-ubiquitinated fol-lowing Met activation. Consistent with this understanding,c-Cbl ubiquitination is elevated in gastric cancer cell lineswhereMet is activated. Similarly ubiquitination of Cbl has been

FIGURE 6. Met-dependent Cbl loss releases EGFR from Cbl-targeted degradation. A, MKN45 cells were treated with Met inhibitor (36 h) and then stimulatedor not with EGF (100 ng/ml) for 1 h. Proteins were immunoblotted with specific antibodies. B, EGFR protein levels � Met inhibitor, EGF, or both EGF and inhibitorwere quantified from five representative experiments. EGFR levels were normalized to actin levels, and the means � S.E. were plotted. *, statistical significanceof p � 0.04 was determined using Student’s t test. C and D, lysates from A were immunoprecipitated with antibodies specific to c-Cbl (C) or Cbl-b (D) and theco-immunoprecipitated proteins separated by SDS-PAGE, and immunoblotted with the designated antibodies. Sizes of the molecular weight markers aredepicted on the left, and the arrows designate the migration of phospho-proteins corresponding to Met and EGFR.

TABLE 1Structural requirements for Cbl loss downstream from various tyro-sine kinases

Met Kinase-dependent Cbl Loss in Gastric Cancer

8056 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 287 • NUMBER 11 • MARCH 9, 2012

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from

Page 10: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

observed downstream from several activated kinases includingEGFR, Src, and CSF-1 receptor (39, 41, 46). Met kinase activityis required for Cbl loss (Figs. 1 and 4A). Tyrosine phosphoryla-tion of Cbl is required for activation of its E3 ligase (47, 48), andmutants that either remove the tyrosine phosphorylation site inthe linker region of Cbl or promote loss of ligase activity areincreasingly found in tumors (49–53). Hence, the ubiquitina-tion of Cbl, subsequent to constitutive Met kinase activation,may be a negative feedback mechanism regulating c-Cbl stabil-ity in human tumors. Other E3 proteins have been shown toself-ubiquitinate and target themselves for degradation, andregulation of Cbl activity, by mediating its stability, has beendocumented previously to occur (reviewed in Ref. 44).We show a preferential loss of c-Cbl protein over Cbl-b

downstream from Met in multiple gastric cancer cells lines.This difference in specificity is reflected by enhanced associa-tion ofMetwith c-Cbl in gastric cancer cell lines over Cbl-b andelevated ubiquitination of c-Cbl under these conditions. Alter-natively, following transient co-transfections, we observed apreferential Met-dependent decrease in Cbl-b over c-Cbl pro-tein levels, which correlated with enhanced ubiquitination ofCbl-b under these conditions. Hence, Met-dependent Cbl losscorrelates with preferential association of Met with c-Cbl ver-susCbl-b under different conditions. Although the basis for thisis unclear, Cbl-b is preferentially targeted for ubiquitinationand degradation in T cells in response to CD28 stimulation ofTCR (reviewed in Ref. 23), whereas upon treatment of chronicmyeloid leukemia patients with the Bcr/Abl inhibitor imatinib,c-Cbl expression and protein levels increase, and Cbl-b levelsdecrease (23, 54). Moreover, c-Cbl and Cbl-b show distincttemporal association with the EGFR following stimulation withEGF (26), supporting the idea that c-Cbl versus Cbl-b may beenriched in distinct subcellular compartments.Constitutive Met activation following amplification in gas-

tric cancers is not the result of an autocrine loop, as HGFmRNA is undetectable in these cell lines (10). Moreover,sequencing of Met has not yet revealed the presence of knownactivating mutations in MKN45, Snu-5, and KATO II gastriccancer cell lines whereMET is amplified (10). Thus, the loss ofCbl protein and negative regulation of Met may be a contribut-ing factor enhancingMet prolonged activation in the absence ofligand. Tpr-Met, an oncogenic Met variant, is the result of agenomic rearrangement, where a leucine zipper dimerizationdomain promotes enforced dimerization of the Met kinase inthe absence of ligand, driving constitutive activation and phos-phorylation (2, 55). However, anothermechanism contributingto the transforming ability of Tpr-Met is the uncoupling fromCbl-mediated negative regulation (31). Tpr-Met is cytoplasmi-cally located, lacks the juxtamembrane Tyr-1003, and is thusunable to associate with Cbl, enter the endocytic pathway, andbe degraded efficiently (31). Restoration of the Cbl binding site,membrane localization, and Cbl expression are required for thedown-regulation of Tpr-Met and suppression of transforma-tion (31). In murine models, wild-type or mutated Met(M1250T) under the murine mammary tumor virus (MMTV)promoter are weakly transforming (56). However, miceexpressing a Met receptor with both M1250T and Y1003Fmutations, thereby diminishing the ability of Cbl to bind and

ubiquitinate Met, exhibit a greater penetrance and a shorterlatency (100 days less) (56), demonstrating that the loss ofCbl-mediated negative regulation enhances Met oncogeniccapabilities. Interestingly, the human gastric cancer cell lineHs746T expresses a mutatedMet receptor (Met �exon14) thatlacks the direct Cbl binding site (Tyr-1003) in addition toamplification of MET (35). In these cells, inhibition of consti-tutively active Met does not impact c-Cbl or Cbl-b protein lev-els (data not shown), supporting the idea that Met-dependent

FIGURE 7. Model of Met-dependent Cbl loss in gastric cancer cells withamplified MET. A, under basal conditions, signaling by active Met promotescell proliferation, survival, migration, and invasion. Constitutively active Metalso promotes phosphorylation, ubiquitination, and eventual proteasomaldegradation of Cbl protein in a Src-independent manner. This uncouplesEGFR from Cbl-mediated negative regulation, allowing it to also promotedownstream signaling. B, in the presence of the Met inhibitor, where Cbl is nolonger phosphorylated and ubiquitinated, Cbl protein accumulates allowingfor more efficient targeting of EGFR for lysosomal degradation in the pres-ence of EGF stimulation.

Met Kinase-dependent Cbl Loss in Gastric Cancer

MARCH 9, 2012 • VOLUME 287 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 8057

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from

Page 11: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

Cbl loss requires association between the two proteins andhighlighting that selection can occur for alternative mecha-nisms that uncouple Met from Cbl. Moreover, in lung cancerswhere tested, more than 80% of the tumors with c-CBLgenomic changes also exhibitmutations in eitherMET orEGFR(52). Exogenous expression of c-Cbl in lung cancer cells can alsoinhibit tumor growth and metastasis in a xenograft mousemodel (57). Thus, taken together, these findings support theidea that the Met-dependent loss of Cbl proteins exhibited inthe four gastric cancer cell lines examined here (Okajima,MKN45, Snu-5, and KATO II) would lead to enhanced dys-regulation of MET and, potentially, other Cbl targets.Notably, in the four gastric cancer cell lines tested, Met-de-

pendent suppression of Cbl levels augments the stability ofother Cbl target proteins such as EGFR (Fig. 6). This has impor-tant implications for RTK cross-talk, whereby activation ofMetcan lead to tyrosine phosphorylation and activation of EGFRand vice versa (43). Hence, Met-dependent Cbl loss in gastriccancers thereby provides amechanism throughwhichMet acti-vation can indirectly enhance EGFR signaling (Fig. 7). Met mayalso promote EGFR stability by virtue of its overexpression,thereby sequestering Cbl away from EGFR. Thus, in these can-cer cells, Met recruitment of Cbl and targeted down-regulationmay take advantage of a normal feedback mechanism to regu-late Cbl, creating a platform of activated RTKs that togethercontribute to the potentiation of oncogenic signaling.

Acknowledgments—We thank members of the Park laboratory forhelpful comments on the manuscript. We also thank Genentech Inc.for providing HGF, Pfizer for PHA-665752, and Dr. D. Haber for theSnu-5 and KATO II cell lines.

REFERENCES1. Cooper, C. S., Park, M., Blair, D. G., Tainsky, M. A., Huebner, K., Croce,

C. M., and Vande Woude, G. F. (1984) Molecular cloning of a new trans-forming gene from a chemically transformed human cell line.Nature 311,29–33

2. Park, M., Dean, M., Cooper, C. S., Schmidt, M., O’Brien, S. J., Blair, D. G.,and Vande Woude, G. F. (1986) Mechanism ofMet oncogene activation.Cell 45, 895–904

3. Bottaro, D. P., Rubin, J. S., Faletto, D. L., Chan,A.M., Kmiecik, T. E., VandeWoude, G. F., and Aaronson, S. A. (1991) Identification of the hepatocytegrowth factor receptor as the c-met proto-oncogene product. Science 251,802–804

4. Naldini, L., Weidner, K. M., Vigna, E., Gaudino, G., Bardelli, A., Ponzetto,C., Narsimhan, R. P., Hartmann, G., Zarnegar, R., Michalopoulos, G. K., etal. (1991) Scatter factor and hepatocyte growth factor are indistinguish-able ligands for the MET receptor. EMBO J. 10, 2867–2878

5. Fournier, T. M., Kamikura, D., Teng, K., and Park, M. (1996) Branchingtubulogenesis but not scatter ofMadin-Darby canine kidney cells requiresa functional Grb2 binding site in the Met receptor tyrosine kinase. J. Biol.Chem. 271, 22211–22217

6. Vasyutina, E., Stebler, J., Brand-Saberi, B., Schulz, S., Raz, E., and Birch-meier, C. (2005) CXCR4 and Gab1 cooperate to control the developmentof migrating muscle progenitor cells. Genes Dev. 19, 2187–2198

7. Huh, C. G., Factor, V. M., Sanchez, A., Uchida, K., Conner, E. A., andThorgeirsson, S. S. (2004) Hepatocyte growth factor/c-Met signalingpathway is required for efficient liver regeneration and repair. Proc. Natl.Acad. Sci. U.S.A. 101, 4477–4482

8. Borowiak, M., Garratt, A. N., Wustefeld, T., Strehle, M., Trautwein, C.,and Birchmeier, C. (2004)Met provides essential signals for liver regener-ation. Proc. Natl. Acad. Sci. U.S.A. 101, 10608–10613

9. Peschard, P., and Park, M. (2007) From Tpr-Met to Met, tumorigenesisand tubes. Oncogene 26, 1276–1285

10. Smolen, G. A., Sordella, R., Muir, B., Mohapatra, G., Barmettler, A., Ar-chibald, H., Kim, W. J., Okimoto, R. A., Bell, D. W., Sgroi, D. C., Chris-tensen, J. G., Settleman, J., and Haber, D. A. (2006) Amplification of METmay identify a subset of cancers with extreme sensitivity to the selectivetyrosine kinase inhibitor PHA-665752. Proc. Natl. Acad. Sci. U.S.A. 103,2316–2321

11. Engelman, J. A., Zejnullahu, K., Mitsudomi, T., Song, Y., Hyland, C., Park,J. O., Lindeman, N., Gale, C. M., Zhao, X., Christensen, J., Kosaka, T.,Holmes, A. J., Rogers, A.M., Cappuzzo, F., Mok, T., Lee, C., Johnson, B. E.,Cantley, L. C., and Janne, P. A. (2007)MET amplification leads to gefitinibresistance in lung cancer by activating ERBB3 signaling. Science 316,1039–1043

12. Okuda, K., Sasaki, H., Yukiue, H., Yano, M., and Fujii, Y. (2008) Met genecopy number predicts the prognosis for completely resected non-smallcell lung cancer. Cancer Sci. 99, 2280–2285

13. Cappuzzo, F., Janne, P. A., Skokan, M., Finocchiaro, G., Rossi, E., Ligorio,C., Zucali, P. A., Terracciano, L., Toschi, L., Roncalli, M., Destro, A., In-carbone,M., Alloisio,M., Santoro, A., and Varella-Garcia,M. (2009)METincreased gene copy number and primary resistance to gefitinib therapy innon-small-cell lung cancer patients. Ann. Oncol. 20, 298–304

14. (2008) Nature 455, 1061–106815. Peschard, P., and Park, M. (2003) Escape from Cbl-mediated down-regu-

lation: a recurrent theme for oncogenic deregulation of receptor tyrosinekinases. Cancer Cell 3, 519–523

16. Langdon, W. Y., Hartley, J. W., Klinken, S. P., Ruscetti, S. K., and Morse,H. C., 3rd (1989) v-cbl, an oncogene from a dual recombinant murineretrovirus that induces early B-lineage lymphomas. Proc. Natl. Acad. Sci.U.S.A. 86, 1168–1172

17. Keane, M. M., Rivero-Lezcano, O. M., Mitchell, J. A., Robbins, K. C., andLipkowitz, S. (1995) Cloning and characterization of cbl-b: a SH3-bindingprotein with homology to the c-cbl proto-oncogene. Oncogene 10,2367–2377

18. Keane, M. M., Ettenberg, S. A., Nau, M. M., Banerjee, P., Cuello, M.,Penninger, J., and Lipkowitz, S. (1999) Cbl-3: a newmammalianCbl familyprotein. Oncogene 18, 3365–3375

19. Thien, C. B., and Langdon,W. Y. (2001) Cbl: many adaptations to regulateprotein tyrosine kinases. Nat. Rev. Mol. Cell Biol. 2, 294–307

20. Bartkiewicz, M., Houghton, A., and Baron, R. (1999) Leucine zipper-me-diated homodimerization of the adaptor protein c-Cbl. A role in c-Cbl’styrosine phosphorylation and its associationwith epidermal growth factorreceptor. J. Biol. Chem. 274, 30887–30895

21. Kozlov, G., Peschard, P., Zimmerman, B., Lin, T., Moldoveanu, T., Man-sur-Azzam, N., Gehring, K., and Park,M. (2007) Structural basis for UBA-mediated dimerization of c-Cbl ubiquitin ligase. J. Biol. Chem. 282,27547–27555

22. Peschard, P., Kozlov, G., Lin, T.,Mirza, I. A., Berghuis, A.M., Lipkowitz, S.,Park, M., and Gehring, K. (2007) Structural basis for ubiquitin-mediateddimerization and activation of the ubiquitin protein ligase Cbl-b.Mol. Cell27, 474–485

23. Thien, C. B., and Langdon,W. Y. (2005) c-Cbl and Cbl-b ubiquitin ligases:substrate diversity and the negative regulation of signalling responses.Biochem. J. 391, 153–166

24. Naramura, M., Jang, I. K., Kole, H., Huang, F., Haines, D., and Gu, H.(2002) c-Cbl and Cbl-b regulate T cell responsiveness by promoting li-gand-induced TCR down-modulation. Nat. Immunol. 3, 1192–1199

25. Purev, E., Neff, L., Horne, W. C., and Baron, R. (2009) c-Cbl and Cbl-b actredundantly to protect osteoclasts from apoptosis and to displaceHDAC6from beta-tubulin, stabilizing microtubules, and podosomes. Mol. Biol.Cell 20, 4021–4030

26. Pennock, S., andWang, Z. (2008) A tale of twoCbls: interplay of c-Cbl andCbl-b in epidermal growth factor receptor down-regulation. Mol. Cell.Biol. 28, 3020–3037

27. Peschard, P., Fournier, T. M., Lamorte, L., Naujokas, M. A., Band, H.,Langdon, W. Y., and Park, M. (2001) Mutation of the c-Cbl TKB domainbinding site on the Met receptor tyrosine kinase converts it into a trans-forming protein.Mol. Cell 8, 995–1004

Met Kinase-dependent Cbl Loss in Gastric Cancer

8058 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 287 • NUMBER 11 • MARCH 9, 2012

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from

Page 12: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

28. Tsygankov, A. Y., Teckchandani, A. M., Feshchenko, E. A., and Swamina-than, G. (2001) Beyond the RING: CBL proteins as multivalent adapters.Oncogene 20, 6382–6402

29. Abella, J. V., Peschard, P., Naujokas, M. A., Lin, T., Saucier, C., Urbe, S.,and Park, M. (2005) Met/Hepatocyte growth factor receptor ubiquitina-tion suppresses transformation and is required for Hrs phosphorylation.Mol. Cell. Biol. 25, 9632–9645

30. Hammond, D. E., Urbe, S., Vande Woude, G. F., and Clague, M. J. (2001)Down-regulation of MET, the receptor for hepatocyte growth factor.On-cogene 20, 2761–2770

31. Mak, H. H., Peschard, P., Lin, T., Naujokas, M. A., Zuo, D., and Park, M.(2007) Oncogenic activation of the Met receptor tyrosine kinase fusionprotein, Tpr-Met, involves exclusion from the endocytic degradativepathway. Oncogene 26, 7213–7221

32. Onozato, R., Kosaka, T., Kuwano, H., Sekido, Y., Yatabe, Y., and Mitsu-domi, T. (2009) Activation of MET by gene amplification or by splicemutations deleting the juxtamembrane domain in primary resected lungcancers. J. Thorac. Oncol. 4, 5–11

33. Kong-Beltran, M., Seshagiri, S., Zha, J., Zhu, W., Bhawe, K., Mendoza, N.,Holcomb, T., Pujara, K., Stinson, J., Fu, L., Severin, C., Rangell, L., Schwall,R., Amler, L., Wickramasinghe, D., and Yauch, R. (2006) Somatic muta-tions lead to an oncogenic deletion of Met in lung cancer. Cancer Res. 66,283–289

34. Ma, P. C., Jagadeeswaran, R., Jagadeesh, S., Tretiakova,M. S., Nallasura, V.,Fox, E. A., Hansen, M., Schaefer, E., Naoki, K., Lader, A., Richards, W.,Sugarbaker, D., Husain, A. N., Christensen, J. G., and Salgia, R. (2005)Functional expression and mutations of c-Met and its therapeutic inhibi-tion with SU11274 and small interfering RNA in non-small cell lung can-cer. Cancer Res. 65, 1479–1488

35. Asaoka, Y., Tada, M., Ikenoue, T., Seto, M., Imai, M., Miyabayashi, K.,Yamamoto, K., Yamamoto, S., Kudo, Y., Mohri, D., Isomura, Y., Ijichi, H.,Tateishi, K., Kanai, F., Ogawa, S., Omata, M., and Koike, K. (2010) Gastriccancer cell line Hs746T harbors a splice site mutation of c-Met causingjuxtamembrane domain deletion. Biochem. Biophys. Res. Commun. 394,1042–1046

36. Rodrigues, G. A., Naujokas,M.A., and Park,M. (1991)Alternative splicinggenerates isoforms of the Met receptor tyrosine kinase which undergodifferential processing.Mol. Cell. Biol. 11, 2962–2970

37. Vandesompele, J., De Preter, K., Pattyn, F., Poppe, B., Van Roy, N., DePaepe, A., and Speleman, F. (2002) Accurate normalization of real-timequantitative RT-PCR data by geometric averaging of multiple internalcontrol genes. Genome Biol. 3, RESEARCH0034

38. Christensen, J. G., Schreck, R., Burrows, J., Kuruganti, P., Chan, E., Le, P.,Chen, J., Wang, X., Ruslim, L., Blake, R., Lipson, K. E., Ramphal, J., Do, S.,Cui, J. J., Cherrington, J. M., and Mendel, D. B. (2003) A selective smallmolecule inhibitor of c-Met kinase inhibits c-Met-dependent phenotypesin vitro and exhibits cytoreductive antitumor activity in vivo. Cancer Res.63, 7345–7355

39. Bao, J., Gur, G., and Yarden, Y. (2003) Src promotes destruction of c-Cbl:implications for oncogenic synergy between Src and growth factor recep-tors. Proc. Natl. Acad. Sci. U.S.A. 100, 2438–2443

40. Ponzetto, C., Bardelli, A., Zhen, Z.,Maina, F., dalla Zonca, P., Giordano, S.,Graziani, A., Panayotou, G., andComoglio, P.M. (1994)Amultifunctionaldocking site mediates signaling and transformation by the hepatocytegrowth factor/scatter factor receptor family. Cell 77, 261–271

41. Ettenberg, S. A., Magnifico, A., Cuello, M., Nau, M. M., Rubinstein, Y. R.,Yarden, Y., Weissman, A. M., and Lipkowitz, S. (2001) Cbl-b-dependentcoordinated degradation of the epidermal growth factor receptor signal-ing complex. J. Biol. Chem. 276, 27677–27684

42. Swaminathan, G., and Tsygankov, A. Y. (2006) The Cbl family proteins:

ring leaders in regulation of cell signaling. J. Cell. Physiol. 209, 21–4343. Lai, A. Z., Abella, J. V., and Park, M. (2009) Cross-talk in Met receptor

oncogenesis. Trends Cell Biol. 19, 542–55144. Ryan, P. E., Davies, G. C., Nau, M.M., and Lipkowitz, S. (2006) Regulating

the regulator: negative regulation ofCbl ubiquitin ligases.Trends Biochem.Sci. 31, 79–88

45. Feshchenko, E. A., Langdon, W. Y., and Tsygankov, A. Y. (1998) Fyn, Yes,and Syk phosphorylation sites in c-Cbl map to the same tyrosine residuesthat become phosphorylated in activated T cells. J. Biol. Chem. 273,8323–8331

46. Wang, Y., Yeung, Y. G., and Stanley, E. R. (1999) CSF-1 stimulated mul-tiubiquitination of the CSF-1 receptor and of Cbl follows their tyrosinephosphorylation and association with other signaling proteins. J. Cell.Biochem. 72, 119–134

47. Levkowitz, G.,Waterman, H., Ettenberg, S. A., Katz,M., Tsygankov, A. Y.,Alroy, I., Lavi, S., Iwai, K., Reiss, Y., Ciechanover, A., Lipkowitz, S., andYarden, Y. (1999) Ubiquitin ligase activity and tyrosine phosphorylationunderlie suppression of growth factor signaling by c-Cbl/Sli-1.Mol. Cell 4,1029–1040

48. Kassenbrock, C. K., and Anderson, S. M. (2004) Regulation of ubiquitinprotein ligase activity in c-Cbl by phosphorylation-induced conforma-tional change and constitutive activation by tyrosine to glutamate pointmutations. J. Biol. Chem. 279, 28017–28027

49. Bisson, S. A., Ujack, E. E., and Robbins, S. M. (2002) Isolation and charac-terization of a novel, transforming allele of the c-Cbl proto-oncogene froma murine macrophage cell line. Oncogene 21, 3677–3687

50. Kales, S. C., Ryan, P. E., Nau, M. M., and Lipkowitz, S. (2010) Cbl andhuman myeloid neoplasms: the Cbl oncogene comes of age. Cancer Res.70, 4789–4794

51. Makishima, H., Cazzolli, H., Szpurka, H., Dunbar, A., Tiu, R., Huh, J.,Muramatsu, H., O’Keefe, C., Hsi, E., Paquette, R. L., Kojima, S., List, A. F.,Sekeres, M. A., McDevitt, M. A., and Maciejewski, J. P. (2009) Mutationsof e3 ubiquitin ligase Cbl family members constitute a novel commonpathogenic lesion in myeloid malignancies. J. Clin. Oncol. 27, 6109–6116

52. Tan, Y. H., Krishnaswamy, S., Nandi, S., Kanteti, R., Vora, S., Onel, K.,Hasina, R., Lo, F. Y., El-Hashani, E., Cervantes, G., Robinson, M., Hsu,H. S., Kales, S. C., Lipkowitz, S., Karrison, T., Sattler, M., Vokes, E. E.,Wang, Y. C., and Salgia, R. (2010)CBL is frequently altered in lung cancers:its relationship to mutations in MET and EGFR tyrosine kinases. PLoSOne 5, e8972

53. Naramura,M., Nadeau, S.,Mohapatra, B., Ahmad, G.,Mukhopadhyay, C.,Sattler, M., Raja, S. M., Natarajan, A., Band, V., and Band, H. (2011) Mu-tant Cbl proteins as oncogenic drivers in myeloproliferative disorders.Oncotarget 2, 245–250

54. McLean, L. A., Gathmann, I., Capdeville, R., Polymeropoulos, M. H., andDressman, M. (2004) Pharmacogenomic analysis of cytogenetic responsein chronic myeloid leukemia patients treated with imatinib. Clin. CancerRes. 10, 155–165

55. Rodrigues, G. A., and Park, M. (1993) Dimerization mediated through aleucine zipper activates the oncogenic potential of the Met receptor tyro-sine kinase.Mol. Cell. Biol. 13, 6711–6722

56. Ponzo, M. G., Lesurf, R., Petkiewicz, S., O’Malley, F. P., Pinnaduwage, D.,Andrulis, I. L., Bull, S. B., Chughtai, N., Zuo, D., Souleimanova, M., Ger-main,D.,Omeroglu, A., Cardiff, R. D., Hallett,M., and Park,M. (2009)Metinduces mammary tumors with diverse histologies and is associated withpoor outcome and human basal breast cancer. Proc. Natl. Acad. Sci. U.S.A.106, 12903–12908

57. Lo, F. Y., Tan, Y. H., Cheng, H. C., Salgia, R., andWang, Y. C. (2011) An E3ubiquitin ligase, c-Cbl: a new therapeutic target of lung cancer. Cancer117, 5344–5350

Met Kinase-dependent Cbl Loss in Gastric Cancer

MARCH 9, 2012 • VOLUME 287 • NUMBER 11 JOURNAL OF BIOLOGICAL CHEMISTRY 8059

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from

Page 13: MetKinase-dependentLossoftheE3LigaseCblinGastric Cancer S · proline-rich region and a UBA domain (c-Cbl and Cbl-b) (19). TheUBAdomainsofbothc-CblandCbl-bfacilitatedimeriza- tion,

Andrea Z. Lai, Michael Durrant, Dongmei Zuo, Colin D. H. Ratcliffe and Morag ParkMet Kinase-dependent Loss of the E3 Ligase Cbl in Gastric Cancer

doi: 10.1074/jbc.M112.339820 originally published online January 18, 20122012, 287:8048-8059.J. Biol. Chem. 

  10.1074/jbc.M112.339820Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

Supplemental material:

  http://www.jbc.org/content/suppl/2012/01/18/M112.339820.DC1

  http://www.jbc.org/content/287/11/8048.full.html#ref-list-1

This article cites 56 references, 26 of which can be accessed free at

by guest on June 20, 2019http://w

ww

.jbc.org/D

ownloaded from