As S targets RING-type E3 ligase c-CBL to induce ...BCR-ABL was observed (Fig. 2Bii). These data...

6
As 4 S 4 targets RING-type E3 ligase c-CBL to induce degradation of BCR-ABL in chronic myelogenous leukemia Jian-Hua Mao a,1 , Xiao-Yan Sun b,1 , Jian-Xiang Liu a,1 , Qun-Ye Zhang a , Ping Liu a , Qiu-Hua Huang a , Keqin Kathy Li a , Quan Chen b , Zhu Chen a,2 , and Sai-Juan Chen a,2 a State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital Afliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; and b State Key Laboratory of Biomembrane and Membrane Biotechnology, Laboratory of Apoptosis and Cancer Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China Contributed by Zhu Chen, November 8, 2010 (sent for review September 10, 2010) Arsenic, a curative agent for acute promyelocytic leukemia, induces cell apoptosis and degradation of BCR-ABL in chronic myelogenous leukemia (CML). We demonstrated that ubiquitination and degra- dation of BCR-ABL was mediated by c-CBL, a RING-type E3 ligase that was also shown to be involved in ubiquitination for many other receptor/protein tyrosine kinases. Our data showed that c-CBL protein was considerably up-regulated by arsenic sulde (As 4 S 4 ). Interestingly, arsenic directly bound the RING nger domain of c-CBL to inhibit its self-ubiquitination/degradation without interfer- ing with the enhancement of ubiquitination and subsequent pro- teolysis of its substrate BCR-ABL. Degradation of BCR-ABL due to c-CBL induction as a result of arsenic treatment was also observed in vivo in CML mice. These ndings provide insight into the molec- ular mechanisms of arsenic and further support its therapeutic appli- cations in CML in combination with tyrosine kinase inhibitors and potentially also in other malignancies involving aberrant receptor/ protein tyrosine kinase signaling. C hronic myelogenous leukemia (CML) is a clonal malignant disease with a characteristic BCR-ABL fusion protein pos- sessing sustained tyrosine kinase activity (13). The tyrosine ki- nase inhibitor (TKI) imatinib has been used with remarkable effects in CML therapy, but resistance and relapse tend to develop after long-term administration (46). Arsenic compounds, in- cluding arsenic trioxide (As 2 O 3 ) and arsenic sulde (As 4 S 4 ), were used to treat leukemia in the mid-19th century (7). Over the past 3 decades these compounds have been used with great success in the treatment of acute promyelocytic leukemia (APL) (8, 9). In search of new alternatives to overcome resistance to TKIs, we tested the combinatorial effect of As 4 S 4 and imatinib in a CML setting. The two agents were found to work synergistically in inhibiting BCR-ABL kinase activity, inducing apoptosis of K562 cells and prolonging survival of CML mice (10, 11). BCR-ABL degradation through the ubiquitinproteasome pathway was ob- served after arsenic treatment, but the molecular events leading to BCR-ABL ubiquitination and degradation remained unclear. Conjugation of ubiquitin to substrate proteins is mediated by a series of enzymes (12, 13). E3 ligases play a key role in the de- termination of the specicity and destination of ubiquitinated substrate proteins. Identication of a specic E3 ligase for BCR- ABL should be crucial to understanding the molecular mecha- nisms of BCR-ABL degradation. Previous studies reported that c-CBL (Casitas B-lineage lymphoma) functioned as E3 ligase for a series of receptor/protein tyrosine kinases (1420). In addition, direct interaction between c-CBL and BCR-ABL was suggested (21). c-CBL has a highly conserved N terminus consisting of a ty- rosine kinase binding (TKB) domain and a RING nger (RF) domain, both reportedly being required for its E3 ligase activity (2224). The TKB domain mediates binding to substrate proteins, whereas the RF domain associates with ubiquitin conjugating enzyme E2 and catalyzes transfer of ubiquitin molecules to sub- strates. The less-conserved C terminus of c-CBL harbors proline- rich regions and a ubiquitin-associated domain. In this context, we explored the molecular mechanisms by which arsenic induces proteolysis of BCR-ABL. Results c-CBL Could Associate with BCR-ABL in a Multiprotein Complex. Previous studies suggested that arsenic could activate some mol- ecules in the ubiquitinproteasome pathway (11, 25). In an at- tempt to reveal the E3 ligase mediating BCR-ABL ubiquitination, we used immunoprecipitation (IP)-2D nano-HPLC-MS/MS on CML-derived K562 cells and 293T cells transfected with BCR- ABL-GFP construct for cataloging proteins potentially associated with the oncoprotein. A number of ubiquitinproteasome-related proteins were identied in puried precipitates, isolated by virtue of specic antibodies, which contained BCR-ABL (Table S1). Of these molecules, c-CBL attracted our attention because it belongs to the CBL family of E3 ligase and is up-regulated by arsenic (Fig. 1A). The interaction between c-CBL and BCR-ABL was con- rmed in transfected cells, K562 cells, and imatinib-resistant K562-R cells (Fig. 1B). This observation prompted us to speculate that c-CBL might serve as E3 ligase for BCR-ABL. As 4 S 4 Up-regulated c-CBL and Induced BCR-ABL Degradation. When K562 cells were treated with As 4 S 4 (2 μM), transcriptional ex- pression changes of neither BCR-ABL nor c-CBL were detected. In contrast, at the protein level, BCR-ABL decreased and c-CBL increased upon effect of arsenic. The modulation of BCR-ABL and c-CBL both started at approximately 8 h after treatment with As 4 S 4 and proceeded in a time-dependent fashion (Fig. 1A, Top and Middle). Similar changes of BCR-ABL and c-CBL, as well as apoptosis could also be demonstrated in K562-resistant (R) cells treated with As 4 S 4 (2 μM) alone or with imatinib (2 μM) (Fig. 1A, Bottom, and Fig. S1). The apparently simultaneous but opposite changes provided a rst clue for a possible causal relationship between the fates of the two proteins. To validate the presumption that c-CBL might mediate arsenic-induced proteolysis of BCR- ABL, we overexpressed c-CBL in K562 cells. Indeed, a down- regulation of BCR-ABL was observed (Fig. 1C). We then designed ve different siRNA sequences to target the expression of c-CBL (Fig. S2) and chose the siRNA with the best silencing effect to establish c-CBL knockdown stable K562 cell lines. Interestingly, down-regulation of c-CBL in K562 cells resulted in increased BCR-ABL (Fig. 1D), suggesting the involvement of c-CBL in regulation of BCR-ABL turnover. In addition, when we treated the Author contributions: J.-H.M., Z.C., and S.-J.C. designed research; J.-H.M., X.-Y.S., Q.-Y.Z., P.L., and K.-Q.K.L. performed research; J.-H.M., J.-X.L., Q.-Y.Z., Q.-H.H., Q.C., Z.C., and S.-J.C. analyzed data; and J.-H.M., J.-X.L., Z.C., and S.-J.C. wrote the paper. The authors declare no conict of interest. 1 J.-H.M., X.-Y.S., and J.-X.L. contributed equally to this work. 2 To whom correspondence may be addressed. E-mail: [email protected] or [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1016311108/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1016311108 PNAS | December 14, 2010 | vol. 107 | no. 50 | 2168321688 MEDICAL SCIENCES Downloaded by guest on August 31, 2020

Transcript of As S targets RING-type E3 ligase c-CBL to induce ...BCR-ABL was observed (Fig. 2Bii). These data...

Page 1: As S targets RING-type E3 ligase c-CBL to induce ...BCR-ABL was observed (Fig. 2Bii). These data indicated that c-CBL should play a major role in BCR-ABL ubiquitination. We also determined

As4S4 targets RING-type E3 ligase c-CBL toinduce degradation of BCR-ABL in chronicmyelogenous leukemiaJian-Hua Maoa,1, Xiao-Yan Sunb,1, Jian-Xiang Liua,1, Qun-Ye Zhanga, Ping Liua, Qiu-Hua Huanga, Keqin Kathy Lia,Quan Chenb, Zhu Chena,2, and Sai-Juan Chena,2

aState Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School ofMedicine, Shanghai 200025, China; and bState Key Laboratory of Biomembrane and Membrane Biotechnology, Laboratory of Apoptosis and Cancer Biology,Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China

Contributed by Zhu Chen, November 8, 2010 (sent for review September 10, 2010)

Arsenic, a curative agent for acute promyelocytic leukemia, inducescell apoptosis and degradation of BCR-ABL in chronic myelogenousleukemia (CML). We demonstrated that ubiquitination and degra-dationof BCR-ABLwasmediated by c-CBL, a RING-type E3 ligase thatwas also shown to be involved in ubiquitination for many otherreceptor/protein tyrosine kinases. Our data showed that c-CBLprotein was considerably up-regulated by arsenic sulfide (As4S4).Interestingly, arsenic directly bound the RING finger domain ofc-CBL to inhibit its self-ubiquitination/degradation without interfer-ing with the enhancement of ubiquitination and subsequent pro-teolysis of its substrate BCR-ABL. Degradation of BCR-ABL due toc-CBL induction as a result of arsenic treatment was also observedin vivo in CML mice. These findings provide insight into the molec-ularmechanisms of arsenic and further support its therapeutic appli-cations in CML in combination with tyrosine kinase inhibitors andpotentially also in other malignancies involving aberrant receptor/protein tyrosine kinase signaling.

Chronic myelogenous leukemia (CML) is a clonal malignantdisease with a characteristic BCR-ABL fusion protein pos-

sessing sustained tyrosine kinase activity (1–3). The tyrosine ki-nase inhibitor (TKI) imatinib has been used with remarkableeffects in CML therapy, but resistance and relapse tend to developafter long-term administration (4–6). Arsenic compounds, in-cluding arsenic trioxide (As2O3) and arsenic sulfide (As4S4), wereused to treat leukemia in the mid-19th century (7). Over the past3 decades these compounds have been used with great success inthe treatment of acute promyelocytic leukemia (APL) (8, 9). Insearch of new alternatives to overcome resistance to TKIs, wetested the combinatorial effect of As4S4 and imatinib in a CMLsetting. The two agents were found to work synergistically ininhibiting BCR-ABL kinase activity, inducing apoptosis of K562cells and prolonging survival of CML mice (10, 11). BCR-ABLdegradation through the ubiquitin–proteasome pathway was ob-served after arsenic treatment, but themolecular events leading toBCR-ABL ubiquitination and degradation remained unclear.Conjugation of ubiquitin to substrate proteins is mediated by

a series of enzymes (12, 13). E3 ligases play a key role in the de-termination of the specificity and destination of ubiquitinatedsubstrate proteins. Identification of a specific E3 ligase for BCR-ABL should be crucial to understanding the molecular mecha-nisms of BCR-ABL degradation. Previous studies reported thatc-CBL (Casitas B-lineage lymphoma) functioned as E3 ligase fora series of receptor/protein tyrosine kinases (14–20). In addition,direct interaction between c-CBL and BCR-ABL was suggested(21). c-CBL has a highly conserved N terminus consisting of a ty-rosine kinase binding (TKB) domain and a RING finger (RF)domain, both reportedly being required for its E3 ligase activity(22–24). The TKB domain mediates binding to substrate proteins,whereas the RF domain associates with ubiquitin conjugatingenzyme E2 and catalyzes transfer of ubiquitin molecules to sub-strates. The less-conserved C terminus of c-CBL harbors proline-rich regions and a ubiquitin-associated domain. In this context, we

explored the molecular mechanisms by which arsenic inducesproteolysis of BCR-ABL.

Resultsc-CBL Could Associate with BCR-ABL in a Multiprotein Complex.Previous studies suggested that arsenic could activate some mol-ecules in the ubiquitin–proteasome pathway (11, 25). In an at-tempt to reveal the E3 ligase mediating BCR-ABL ubiquitination,we used immunoprecipitation (IP)-2D nano-HPLC-MS/MS onCML-derived K562 cells and 293T cells transfected with BCR-ABL-GFP construct for cataloging proteins potentially associatedwith the oncoprotein. A number of ubiquitin–proteasome-relatedproteins were identified in purified precipitates, isolated by virtueof specific antibodies, which contained BCR-ABL (Table S1). Ofthese molecules, c-CBL attracted our attention because it belongsto the CBL family of E3 ligase and is up-regulated by arsenic (Fig.1A). The interaction between c-CBL and BCR-ABL was con-firmed in transfected cells, K562 cells, and imatinib-resistantK562-R cells (Fig. 1B). This observation prompted us to speculatethat c-CBL might serve as E3 ligase for BCR-ABL.

As4S4 Up-regulated c-CBL and Induced BCR-ABL Degradation. WhenK562 cells were treated with As4S4 (2 μM), transcriptional ex-pression changes of neither BCR-ABL nor c-CBL were detected.In contrast, at the protein level, BCR-ABL decreased and c-CBLincreased upon effect of arsenic. The modulation of BCR-ABLand c-CBL both started at approximately 8 h after treatment withAs4S4 and proceeded in a time-dependent fashion (Fig. 1A, Topand Middle). Similar changes of BCR-ABL and c-CBL, as well asapoptosis could also be demonstrated in K562-resistant (R) cellstreated with As4S4 (2 μM) alone or with imatinib (2 μM) (Fig. 1A,Bottom, and Fig. S1). The apparently simultaneous but oppositechanges provided a first clue for a possible causal relationshipbetween the fates of the two proteins. To validate the presumptionthat c-CBL might mediate arsenic-induced proteolysis of BCR-ABL, we overexpressed c-CBL in K562 cells. Indeed, a down-regulation of BCR-ABLwas observed (Fig. 1C).We then designedfive different siRNA sequences to target the expression of c-CBL(Fig. S2) and chose the siRNA with the best silencing effect toestablish c-CBL knockdown stable K562 cell lines. Interestingly,down-regulation of c-CBL in K562 cells resulted in increasedBCR-ABL (Fig. 1D), suggesting the involvement of c-CBL inregulation of BCR-ABL turnover. In addition, whenwe treated the

Author contributions: J.-H.M., Z.C., and S.-J.C. designed research; J.-H.M., X.-Y.S., Q.-Y.Z.,P.L., and K.-Q.K.L. performed research; J.-H.M., J.-X.L., Q.-Y.Z., Q.-H.H., Q.C., Z.C., andS.-J.C. analyzed data; and J.-H.M., J.-X.L., Z.C., and S.-J.C. wrote the paper.

The authors declare no conflict of interest.1J.-H.M., X.-Y.S., and J.-X.L. contributed equally to this work.2To whom correspondence may be addressed. E-mail: [email protected] or [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1016311108/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1016311108 PNAS | December 14, 2010 | vol. 107 | no. 50 | 21683–21688

MED

ICALSC

IENCE

S

Dow

nloa

ded

by g

uest

on

Aug

ust 3

1, 2

020

Page 2: As S targets RING-type E3 ligase c-CBL to induce ...BCR-ABL was observed (Fig. 2Bii). These data indicated that c-CBL should play a major role in BCR-ABL ubiquitination. We also determined

c-CBL knockdownK562 cells withAs4S4, the degradation of BCR-ABL was significantly compromised (Fig. 1E).

c-CBL Mediates Ubiquitination of BCR-ABL at K1517. To determinewhether c-CBL represented a bona fide E3 ligase for BCR-ABL,we conducted an in vitro ubiquitination assay with relevant com-ponents. Migr1-BCR-ABL-GFP and pFlag-CMV4-c-CBL weretransfected into 293T cells. IP was performed with GFP and Flagantibodies. The purified precipitates were used as substrate and E3ligase, respectively. Moreover, because previous studies demon-strated that theG306ofTKBdomainand theC381ofRFdomainof

c-CBLwereessential forE3 ligase function (23), twosinglemutants,G306E(M1) and C381A(M2), as well as a double mutant, G306E/C381A(M1+2) (Fig. 2A),werealsoprepared to testwhether loss ofc-CBL enzymatic activity would affect BCR-ABL ubiquitination.As expected, whenwild-type c-CBLwas added (Fig. 2Bi, right-mostlane), ubiquitination of BCR-ABL increased significantly com-pared with that without c-CBL (middle lane). Nevertheless, whenc-CBL mutants were used, markedly reduced ubiquitination ofBCR-ABL was observed (Fig. 2Bii). These data indicated thatc-CBL should play a major role in BCR-ABL ubiquitination. Wealso determined the topology of BCR-ABL ubiquitination using

Fig. 1. c-CBL expression and BCR-ABL modulation. (A) RT-PCR and Western blot of BCR-ABL (Top) and c-CBL (Middle)in K562 cells, and Western blot of BCR-ABL and c-CBL(Bottom) in K562-R cells. (B) Wild-type BCR-ABL and c-CBLcotransfected into 293T cells. c-CBL was immunoprecipi-tated by anti-Flag (M2) antibody, and BCR-ABL was blottedby GFP antibody (Upper Left), or BCR-ABL immunopreci-pitated by GFP antibody and c-CBL blotted by anti-Flag(M2) antibody (Upper Right). Lower: Interaction of en-dogenous BCR-ABL and c-CBL in K562 and K562-R cellstreated with or without (Con) 2 μM of arsenic for 4 h. Co-IPwas performed using anti–c-CBL antibody and Westernblot with anti–c-CBL and anti-BCR antibodies in K562 cells.(C) Western blot of c-CBL and BCR-ABL in K562 cells over-expressing c-CBL. (D) RT-PCR (Upper) and Western blot(Lower) of c-CBL and BCR-ABL in K562 cells with stable c-CBL siRNA. (E) Western blot (Upper) and gray scan (Lower)of BCR-ABL in K562 cells. Gray value (y-axis) representsdensity of BCR-ABL bands in Western blot. R, ratio of grayvalues before and after arsenic treatment. GAPDH wasused as internal control. K562, cells without transfection; AS,As4S4; Vec, K562 cells transfected with empty vector; c-CBL,K562 cells transfected with c-CBL; NC, negative control withscramble siRNA; Cont., control without As4S4 treatment.

Fig. 2. Ubiquitination of BCR-ABL byc-CBL. (A) Schematic domain structureof human c-CBL. (B) BCR-ABL ubiq-uitination by c-CBL in in vitro ubiq-uitination assay. BCR-ABL and c-CBLwere transfected into 293T cells usingMigr1-BCR-ABL-GFP and pFlag-CMV4-c-CBL expression vectors and purifiedby using anti-GFP and anti-M2 beads,respectively. Ubiquitinated BCR-ABLwas detected by Western blot withanti-FK2 antibody (Upper) to showpolyubiquitinated protein and BCR-ABL (Lower). In vitro ubiquitination ofBCR-ABL using (i) wild-type c-CBL, (ii)mutant c-CBL, (iii) K48R mutant ubiq-uitin, and (iv) in vitro ubiquitinationassay using BCR-ABL or BSA as sub-strate immune-blotted with ubiquitinantibody (FK2 IB) or BCR antibody(BCR IB). (C) Wild-type or mutant (M1,G306E; M2, C381A) c-CBL was over-expressed in K562 cells. c-CBL andBCR-ABL proteins were examined byWestern blot. Cell morphology ofnontransfected (K562) and c-CBLtransfected K562 (c-CBL) cells waschecked with Wright’s staining undermicroscope (Lower). (D) IP using anti-BCR antibody in nontransfected K562 cells (K), scramble siRNA negative control (NC), and c-CBL siRNA stable K562 cell line(CBL). Ubiquitination of BCR-ABLwas examined byWestern blot using anti-FK2 antibody. (E) Immunofluorescence for colocalization of BCR-ABL and 20Sα6 afteradding As4S4 and proteasome inhibitor MG132 in K562 cells. (Scale bars, 10 μm.)

21684 | www.pnas.org/cgi/doi/10.1073/pnas.1016311108 Mao et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

1, 2

020

Page 3: As S targets RING-type E3 ligase c-CBL to induce ...BCR-ABL was observed (Fig. 2Bii). These data indicated that c-CBL should play a major role in BCR-ABL ubiquitination. We also determined

a K48R mutant ubiquitin, which exhibited diminished BCR-ABLubiquitination (Fig. 2Biii), suggesting that ubiquitin ligation ofBCR-ABL occurred mostly at K48 of ubiquitin, tagging BCR-ABLto the proteasome for degradation.When BCR-ABL was replacedbyBSA, aminimal level ofubiquitinationwasdetectable (Fig. 2Biv),indicating a relative substrate specificity of c-CBL.To confirm the E3 ligase function of c-CBL for endogenous

BCR-ABL, wild-type and mutant c-CBL constructs were trans-fected into K562 cells. Notably, cells expressing mutant c-CBLshowed reduced degradation of BCR-ABL as compared withthose with wild-type c-CBL expression. Apoptosis of K562 cellswas also observed with overexpression of wild-type c-CBL (Fig.2C). On the other hand, c-CBL knockdown in K562 cells led tomarked inhibition of BCR-ABL ubiquitination (Fig. 2D). Coloc-alization of BCR-ABL with 20S proteasome core α6 subunit wasdemonstrated in immunofluorescence assay (Fig. 2E), providingevidence that ubiquitinated BCR-ABL was indeed targeted to theproteasome for degradation.Next, we addressed the ubiquitination site(s) on BCR-ABL by

using IP-2D nano-HPLC-MS/MS in c-CBL siRNA or scramblesiRNA transfected K562 cells. The advantage of this approachcould be that the only differences between the two cell types shouldresult from the expression levels of c-CBL. Thus the ubiquitinationsite(s) modified in the presence of c-CBL but unaltered in the ab-

sence of c-CBL could be regarded as c-CBL specific. In K562 cellswith scramble siRNA, a peptide fragment containing K1517 ofBCR-ABL [P210, corresponding to K616 of human ABL (Uni-ProtKB/Swiss-Prot P00519)] gave a GG-K ubiquitin-modificationsignal (Fig. 3A). MS/MS analysis showed that K1517 of BCR-ABLwas the ubiquitinated residue (Fig. 3B). In support of this result, invitro assay showed that BCR-ABLK1517Rmutant but not the fourother lysine-arginine (K-R) mutants (K39R, K213R, K795R, andK1990R) generated abrogated ubiquitination, whereasmutation ofall five lysine residues (5M) displayed approximately the sameresults as did K1517R (Fig. 3C), suggesting K1517 as a major lysinetargeted by c-CBL.

As4S4 Inhibited c-CBL Self-ubiquitination and Degradation. BecauseAs4S4 up-regulated c-CBL,weexamined the catabolic properties ofthis E3 ligase using inhibitors against the caspases (Z-VAD-FMK),lysosome (chloroquine or NH4Cl), and proteasome (MG132)pathways. MG132, but not the other inhibitors, caused an obviousincrease of c-CBL (Fig. 4A and Fig. S3), suggesting c-CBL itselfcould be degradedmainly in the context of the proteasome system.Because arsenic was previously reported to activate proteasomeactivity (25), we checked the protein level of 20Sα6, the majorcomponent of proteasome complex. A slight increase of 20Sα6after arsenic treatment was observed (Fig. 4B), excluding the pos-

Fig. 3. Identification of ubiquitination site(s) in BCR-ABL. (A) MS/MS and database search identified a GG-K signal on the peptide fragment containing aminoacids 1511–1517 of BCR-ABL. (B) Identification by MSof BCR-ABL ubiquitination site mediated by c-CBL. (C)Upper: Schematic structure of BCR-ABL; arrowheadsindicate lysines that may conjugate ubiquitin mole-cules. (Lower) GFP-tagged BCR-ABL or its mutantswere transfected into 293T cells and purified by GFPantibody. Purified BCR-ABL or its mutant proteinswere used as substrate, and purified c-CBL as E3 ligasein in vitro ubiquitination assay. M1–5 indicate K-Rmutations at each site; 5M indicates mutation of all offive lysines.

Fig. 4. Inhibition of c-CBL self-ubiquitination uponAs4S4. (A) K562 cells were treated with lysosomeinhibitor chloroquine (100 μM), broad-spectrumcaspase inhibitor Z-VAD-FMK (50 μM), or protea-some inhibitor MG132 (5 μM), and 2 μM of As4S4. c-CBL protein was examined by Western blot. (B)Western blot of 20Sα6 after As4S4 treatment. (C)K562 cells were treated with cycloheximide (CHX,100 μg/mL) with or without As4S4 (2 μM) or MG132 (5μM) before treatment for 4 h. Western blot wasperformed with anti–c-CBL antibody. (D) pFlag-CMV4-c-CBL was transfected into 293T cells. Over-expressed c-CBL was purified and used as both sub-strate and E3 ligase. Self-ubiquitination of c-CBL waschecked by Western blot using anti-FK2 antibody.AS, As4S4 added into the in vitro ubiquitinationsystem; c-CBL+AS, As4S4 (2 μM for 4 h) added to thepFlag-CMV4-c-CBL transfected 293T cells. (E) pFlag-CMV4-c-CBL was transfected into 293T cells, whichwere treated with MG132 (5 μM for 4 h) and thenwith As4S4 (2 μM for 4 h) before harvesting. c-CBLwas purified from cell extract using anti-Flag (M2)beads, and Western blot was performed to checkubiquitination of c-CBL using anti-FK2 antibody. (F)K562 cells were treated with MG132 and As4S4 in a similar way to that for 293T cells. c-CBL was purified using c-CBL antibody. Western blot was performedusing anti-FK2 antibody. (G) MS/MS identified the ubiquitination site(s) on c-CBL. The peptide fragment containing amino acids 383–392 had a GG-K signal.(H) c-CBL or its mutants were used as substrate in in vitro ubiquitination assay with wild-type c-CBL. (I) Purified c-CBL or its mutant (K389R) were used as E3ligase, and purified BCR-ABL as substrate in in vitro ubiquitination assay. AS, As4S4; 382, K382R; 389, K389R; 392, K392R.

Mao et al. PNAS | December 14, 2010 | vol. 107 | no. 50 | 21685

MED

ICALSC

IENCE

S

Dow

nloa

ded

by g

uest

on

Aug

ust 3

1, 2

020

Page 4: As S targets RING-type E3 ligase c-CBL to induce ...BCR-ABL was observed (Fig. 2Bii). These data indicated that c-CBL should play a major role in BCR-ABL ubiquitination. We also determined

sibility that arsenic-induced c-CBL increasewasdue to inhibitionofthe proteasome. The protein synthesis inhibitor cycloheximide(CHX) blocked c-CBL synthesis and resulted in gradual decreaseof the protein, whereas As4S4 or MG132 stabilized c-CBL in thepresence of CHX (Fig. 4C). On the other hand, in vitro assaydemonstrated that c-CBL underwent self-ubiquitination, as pre-viously described (26, 27), and this process was inhibited by As4S4(Fig. 4D). Arsenic treatment of 293T cells overexpressing c-CBLand K562 cells provided further evidence supporting this effect ofarsenic (Fig. 4 E and F).We subsequently tried to identify the ubiquitination site(s) of

c-CBL. In purified c-CBL from 293T cells transfected with Flag-c-CBL, IP-2D nano-HPLC-MS/MS detected a peptide fragmentcontaining K389 of c-CBL with a GG-K signal (Fig. 4G). A seriesof c-CBL mutants—K382R, K389R, and K392R—was then gen-erated and tested in ubiquitination assays. Only K389R exhibitedabrogation of c-CBL self-ubiquitination in in vitro assay (Fig. 4H),consistent with MS analysis. Importantly, K389Rmutation did notaffect the E3 ligase activity of c-CBL toward BCR-ABL substrate(Fig. 4I). The positions ofK389 and the cysteines/histidine residuescoordinating metal binding in RF domain and the TKB domain ofc-CBL are shown in Fig. 5C. The structural simulation also dem-onstrates the interaction interfaces between c-CBL, the associatedE2 conjugase UbcH7, and the substrate ABL (Fig. 5C).

Arsenic Targeted c-CBL Through Direct Binding of RF Domain. It wasrecently reported that arsenic bound the RF domain of promye-locytic leukemia (PML), affecting its stability (28, 29). Thisprompted us to investigate whether As4S4 might target c-CBL ina similar manner. HeLa cells were transfected with EGFP-c-CBLconstruct and treated with ReAsH, a labeled organic arseniccompound. Confocal microscopy showed that c-CBL and ReAsHcolocalized in the cytoplasm (Fig. 5A). Using the organic arsenicp-aminophenylarsine oxide (PAPAO) labeled with biotin (Biotin-As) to treat 293T cells overexpressing c-CBL, followed by co-IPperformed with streptavidin-agarose beads, we showed a directbinding of arsenic with c-CBL (Fig. 5B). The specificity of thisbinding was confirmed through applying unlabeled arsenic com-pounds (Fig. 5B). The RF domain was indispensable for theinteraction because its deletion (ΔRF) completely abrogated ar-senic binding (Fig. 5B, Right). Mutation C381A in RF almost

abolished arsenic binding, suggesting C381 as a critical bindingsite, which was supported by modeling of the 3D structure of theRF domain of c-CBL (Fig. 5C). Control analysis showed absenceof arsenic binding to RARα but a binding to PML as previouslyreported (28).

Arsenic Modulated c-CBL and BCR-ABL in in Vivo CML Setting. Finally,we used a previously well-documented CML mouse model (11) toevaluate the in vivo effect of As4S4 on the fate of both c-CBL andBCR-ABL proteins. Migr1-BCR-ABL-IRES-GFP expressionplasmid was transfected into bone marrow (BM) cells from donormice pretreated with 5-fluorouracil. BCR-ABL–expressing BMcells were selected and transplanted to recipient. GFP expressionin peripheral blood (PB) was determined using flow cytometry tomonitor disease development (Fig. 6A). As4S4 (10 mg/kg per day)was administered on the fifth day after transplantation. c-CBL andBCR-ABL expression was examined in spleen cells when GFPlevels in PB cells showed significant differences between As4S4treatment and placebo (treated with normal saline) groups. c-CBLwas much lower in BCR-ABL–expressing cells compared withcontrol mice (Fig. 6B). Arsenic treatment increased c-CBL toa level comparable to that in control mice and a degradation ofBCR-ABL (Fig. 6B). Both c-CBL and BCR-ABL changes weredetected at the protein but not mRNA level. To clarify whether invivo changes of c-CBL and BCR-ABL occurred within leukemiccells or resulted from reduced number of BCR-ABL–expressingcells, in vitro culture of spleen cells from CMLmice and of mouse32D hematopoietic progenitor cells with stable transfection ofBCR-ABL was conducted (Fig. 6 C and D). After treatment withAs4S4, a significant increase of c-CBL and a reduction of BCR-ABL were observed in both culture systems. In addition, BMCD34+ cells were obtained from CML patients and normal indi-viduals with informed consent, cultured in vitro, and analyzedbefore and after As4S4 treatment. As shown in Fig. 6E, c-CBLexpression was lower in BM CD34+ cells from CML cases com-pared with controls, and arsenic treatment led to elevated c-CBLand decreased BCR-ABL.

DiscussionArsenic compounds were used in disease treatment thousands ofyears ago (30), but their therapeutic value has been revived only

Fig. 5. Arsenic binding of the RF domainof c-CBL. (A) Colocalization of c-CBL witharsenic in HeLa cells. EGFP-c-CBL or EGFP-c-CBL-ΔRF were introduced into HeLa cells,which were treated with MG132 (5 μM for4 h) before adding ReAsH. (Scale bars, 10μm.) (B)Wild-type c-CBL, c-CBL-ΔRF,or c-CBL(C381A) were transfected and treated withbiotin-labeled PAPAO (Biotin-As), while IPand IB were performed using streptavidinagarose and c-CBL antibody. As4S4 (AS),As2O3 (ATO), and PAPAO were unlabeled.PML and RARα were used as positive andnegative controls, respectively. (C) Structuresimulation for interaction of c-CBL (TKB andRF), ABL (TyrKC, SH2, and SH3), and UbcH7according to the structures of Protein DataBank ID 1OPL and 1FBV (43, 44). The TyrKCdomain of ABL is in deep cyan, SH2 domainin cyan, and SH3 domain in pale cyan. TheTKB domain of c-CBL is in green, RF domainin blue, the two zinc ions are shown as redspheres, C381 of c-CBL is shown in yellow,and K389 in orange. UbcH7 is in red. Right:TheRFdomainanditsadjacentproteinpartsare highlighted, with amino acid sequenceof the RF domain at the bottom. The cyste-ine/histidine residues coordinatingwith zincions are in light purple, K389 in orange.

21686 | www.pnas.org/cgi/doi/10.1073/pnas.1016311108 Mao et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

1, 2

020

Page 5: As S targets RING-type E3 ligase c-CBL to induce ...BCR-ABL was observed (Fig. 2Bii). These data indicated that c-CBL should play a major role in BCR-ABL ubiquitination. We also determined

recently owing to the success in APL (8, 9). As2O3 in combinationwith all-trans retinoic acid can achieve 5-year event-free survivalin 90% of APL patients (9). Studies have shown that arsenicinduces sumoylation and ubiquitination and subsequent degra-dation of the oncoprotein PML-RARα (31). Arsenic was alsoshown to induce down-regulation of BCR-ABL and apoptosis ofCML cells (10, 11). Here we demonstrated that arsenic treatmentled to up-regulation of c-CBL and degradation of BCR-ABLproteins at both cell (including K562-R cells) and organism levels.Furthermore, overexpression of c-CBL in CML cells alone in-duced degradation of BCR-ABL, whereas c-CBL knockdown ledto increased BCR-ABL, in agreement with the study in mouseembryonic fibroblasts (32). In c-CBL knockdownK562 cells, As4S4failed to induce ubiquitination and degradation of BCR-ABL,suggesting c-CBL as a mediator of the arsenic effect. In support ofthis, when c-CBL−/− genetic backgroundwas introduced intoBCR-ABL transgenic mice, CML animals exhibited a significantly lowersurvival rate (33).Arsenic up-regulated c-CBL to impair viability ofneoplastic cells not only in CMLbut also inAPL and gastric cancer(34). c-CBL mutations or defects in expression have been identi-fied in human AML (35, 36), whereas wild-type c-CBL could res-cue these defects, suggesting its tumor suppressor property.Imatinib and second-generation TKIs have been successful in

treating CML, but long-term use often leads to resistance andrelapse (6). Instead of inhibiting tyrosine kinase activity, arsenicexerts its effects through the ubiquitin–proteasome pathway thatultimately leads to degradation of the kinase. Imatinib mainlytargets dividing cells but not CML leukemia initiating cells (LICs),one of the reasons for relapse after withdrawal. Arsenic has beenshown to be able to target CML LICs by inducing PML degra-dation (37). Our previous study also suggested a synergistic effectof As4S4 and imatinib to induce apoptosis of CML cells, inhibitBCR-ABL tyrosine kinase activity, and increase survival time ofCML mice (10, 11). The combination of arsenic and TKIs mayhold promise for a more effective curative approach for CML.c-CBL exerts its functions either through E3 ligase activity or as

an adaptor to recruit other molecules. In this work, we providedevidence that c-CBL was an E3 ligase for BCR-ABL. Of note,c-CBL also serves as E3 ligase for a number of receptor/proteintyrosine kinases, aberrant signaling of which is frequently involvedin malignancies (4, 5, 20, 38–40). Mutations of these tyrosinekinases disrupting interaction with c-CBL have been reported toplay important roles in malignant transformation (41). c-CBLmutations have also been identified inmultiple cancers (35, 36, 40).

These properties of c-CBL suggest it may serve as a unique ther-apeutic target for cancers associated with uncontrolled tyrosinekinase activity.The demonstration of a self-ubiquitination site at K389 of

c-CBL should be of interest in that K389R mutation abolishedself-ubiquitination but not the E3 ligase activity toward BCR-ABLsubstrate. Comparison of RF domains between PML and c-CBLindicates conservation of critical cysteines/histidine residues. Al-though arsenic binds the RF of c-CBL in a similar manner to whatit does in PML, the agent generates distinct outcomes of the twotarget proteins: ubiquitination of c-CBL is inhibited, contrary toPML, whose sumoylation/ubiquitination is promoted. Becausearsenic tends to coordinate with three cysteines, whereas zinc doeswith four cysteine/histidine residues, it is possible that binding ofc-CBL by arsenic leads to conformational alterations in localprotein structure that in turn may block ubiquitin conjugation atK389, which is located within a loop just between the two co-ordinating arsenic ions. Modeling of the protein complex con-taining ABL (TyrKC, SH2, and SH3), c-CBL (TKB and RF), andUbcH7 suggests that a consensus sequence in the kinase catalyticdomain of BCR-ABL makes direct contacts with the TKB, on theopposite side of the RF domain of c-CBL. This model providesa possible explanation that subtle structural changes of the RFdomain of c-CBL by arsenic binding are less likely to affect itsdirect interaction with BCR-ABL (Fig. 5C). On the other hand,interaction with E2 ubiquitin conjugating enzyme UbcH7 may bechanged because the interaction interface lies largely in the RFdomain (Fig. 5C). Enhanced interaction among components ofthis complex may promote ubiquitination of BCR-ABL. It is in-teresting to note that another RF domain-containing E3 ligase,SIAH1, was also capable of binding arsenic (Fig. S4). It is to beexplored whether other proteins with homologous RF may betargeted by arsenic, as well as how arsenic regulates functions ofthese proteins under different pathophysiological circumstances.

Materials and MethodsReagents and Antibodies. As4S4 and MG132 (Calbiochem) were prepared aspreviously described (10). Antibodies used for IP and/or Western blotting in-clude anti-ABL (k-12), anti-BCR (N-20) (Santa Cruz Biotechnology), anti-ABL(ab16905), anti-GFP (ab290) (Abcam), anti-ubiquitin (FK2) mouse monoclonalantibody (Affinity Research Products), and anti–c-CBL (BD Transduction Lab-oratories). Protein A and ProteinG Sepharose 4 fastflow (GEHealthcare) wereused to purify immunoprecipitates. ReAsH-EDT2 (ReAsH) was purchased fromSigma-Aldrich.

Fig. 6. Up-regulation of c-CBL in CML mice andCD34+ cells of CML patients by As4S4. (A) Whiteblood cell (WBC) counts, GFP-positive cells in PB, andliver/body or spleen/body weight ratios of micemeasured 13 d after As4S4 treatment. Control, micetransplanted with cells transfected with empty vec-tor (n = 4); Placebo, CML mice treated with normalsaline (n = 13); AS, CML mice treated with As4S4 (n =14). (B) RT-PCR and Western blot of c-CBL and BCR-ABL in spleen cells from mice with or without As4S4treatment. (C) Western blot of c-CBL and BCR-ABL inspleen cells from CML mice treated with As4S4 invitro. (D) RT-PCR (Upper) and Western blot (Lower)of BCR-ABL and c-CBL in 32D cells with stable BCR-ABL transfection. (E) Western blot of c-CBL and BCR-ABL in CML patients’ CD34+ cells treated with As4S4in vitro. SP, spleen; 32D, 32D cells without BCR-ABLtransfection; 32D-BA, 32D cells with stable BCR-ABLtransfection.

Mao et al. PNAS | December 14, 2010 | vol. 107 | no. 50 | 21687

MED

ICALSC

IENCE

S

Dow

nloa

ded

by g

uest

on

Aug

ust 3

1, 2

020

Page 6: As S targets RING-type E3 ligase c-CBL to induce ...BCR-ABL was observed (Fig. 2Bii). These data indicated that c-CBL should play a major role in BCR-ABL ubiquitination. We also determined

Cell Culture, Transfection, IP, and Western Blot. K562 cell line was cultured inRPMI 1640 supplemented with 10% FBS (HyClone). The imatinib-resistant cellline K562-Rwas described previously (42). 293T andHeLa cells fromATCCwerecultured in DMEM with 10% FBS. K562 cells were transfected by electro-poration using the Amaxa Nucleofector system. 293T and HeLa cells weretransfected using Superfect (Qiagen). Both K562 and 293T cells were subject toIP with anti-BCR and anti-GFP or anti-Flag (M2) antibodies. Western blot wasperformed with anti-FK2, anti–c-CBL, or anti-ABL antibodies as previouslydescribed (11).

IP-2D-Nano-HPLC-MALDI-MS/MS. The immune complex was incubated with100 mM DTT and 400 mM iodoacetamide for 30 min in dark. The sample wasprecipitated and digested overnight with trypsin at 37 °C. 2D-nano HPLC wasperformed using a workstation (Shimadzu) and analyzed using a 4700 massspectrometer (ABI).

In Vitro Ubiquitination Assay. 293T cells were transfected with BCR-ABL wildtype or its mutants, together with c-CBL and its mutants. The purified proteinswere used as substrate or E3 ligase in vitro ubiquitination assay. The reactionmixture contains 50 mM Tris-HCl (PH 7.4), 2 mM DTT, 2 mM ATP (Bos-tonBiochem), 50 ng E1 (Calbiochem), 400 ngE2 (UbcH7; BostonBiochem), 40 ngpurifiedc-CBLor itsmutant,50ngBCR-ABLorBSA,and2μgFlag-UborK48R-Ub(BostonBiochem). The reactionmixture was incubated at 37 °C for 3 to 4 h andterminated by boiling in 2× SDS loading buffer for 10 min and assayed usingWestern blot.

Immunofluorescence. K562 cells were treated with As4S4 andMG132, smearedon slides, fixed for 10 min, and incubated with anti-ABL and anti-20Sα6 anti-bodies. HeLa cells stably expressing EGFP-c-CBL or itsmutantwere treatedwithMG132 before adding 5 μMof ReAsH. Immunofluorescence was performed aspreviously described (28). Images were captured using a laser scanning con-focal microscope with LSM5 Pascal software (Zeiss, Jena, Germany).

Streptavidin Agarose Affinity Assay. Streptavidin agarose affinity assay wasperformed as previously described (28).

BCR-ABL Mouse Model and CD34+ BM Cells from CML Patients. Mouse CMLmodel was established as previously reported (11). Mice were randomized andtreated with As4S4 (10 mg/kg/d; vena caudalis injection) or normal saline. BMof CML patients who had not received treatment were obtained with in-formed consent according to institutional guidelines. CD34+ cells were se-lected (Miltenyi Biotech) and cultured as previously described (10).

ACKNOWLEDGMENTS. Migr1-BCR-ABL-GFP expression vector was kindlyprovided by Dr. Rui-Bao Ren (Brandeis University, Waltham, MA). Theimatinib-resistant cell line K562-R was kindly provided by Prof. Junia V.Melo (University of Adelaide, Australia). Biotin-As was kindly provided byDr. Xiao-Wei Zhang (Shanghai Institute of Hematology, Shanghai, China).This work was supported in part by Grant 863:2006AA02A405 from theNational High Tech Program for Biotechnology, Grant 973, 2010CB529202from the Chinese National Key Basic Research Project, Grants 30772744 and30821063 from the National Natural Science Foundation of China, GrantY0201 from the Key Discipline Program of Shanghai Municipal EducationCommission, and by Shanghai Charity Cancer Research Funds.

1. Lugo TG, Pendergast AM, Muller AJ, Witte ON (1990) Tyrosine kinase activity andtransformation potency of bcr-abl oncogene products. Science 247:1079–1082.

2. Goldman JM, Melo JV (2003) Chronic myeloid leukemia—advances in biology andnew approaches to treatment. N Engl J Med 349:1451–1464.

3. Deininger MW, Goldman JM, Melo JV (2000) The molecular biology of chronicmyeloid leukemia. Blood 96:3343–3356.

4. Quintás-Cardama A, Kantarjian H, Cortes J (2009) Imatinib and beyond—exploringthe full potential of targeted therapy for CML. Nat Rev Clin Oncol 6:535–543.

5. Larson RA, et al.; IRIS (International Randomized Interferon vs STI571) Study Group(2008) Imatinib pharmacokinetics and its correlation with response and safety inchronic-phase chronic myeloid leukemia: A subanalysis of the IRIS study. Blood 111:4022–4028.

6. Jabbour E, et al. (2009) Long-term outcome of patients with chronic myeloid leukemiatreated with second-generation tyrosine kinase inhibitors after imatinib failure ispredicted by the in vitro sensitivity of BCR-ABL kinase domain mutations. Blood 114:2037–2043.

7. Kwong YL, Todd D (1997) Delicious poison: Arsenic trioxide for the treatment ofleukemia. Blood 89:3487–3488.

8. Chen GQ, et al. (1997) Use of arsenic trioxide (As2O3) in the treatment of acutepromyelocytic leukemia (APL): I. As2O3 exerts dose-dependent dual effects on APLcells. Blood 89:3345–3353.

9. Hu J, et al. (2009) Long-term efficacy and safety of all-trans retinoic acid/arsenictrioxide-based therapy in newly diagnosed acute promyelocytic leukemia. Proc NatlAcad Sci USA 106:3342–3347.

10. Yin T, et al. (2004) Combined effects of As4S4 and imatinib on chronic myeloidleukemia cells and BCR-ABL oncoprotein. Blood 104:4219–4225.

11. Zhang QY, et al. (2009) A systems biology understanding of the synergistic effects ofarsenic sulfide and Imatinib in BCR/ABL-associated leukemia. Proc Natl Acad Sci USA106:3378–3383.

12. Hershko A, Ciechanover A (1998) The ubiquitin system. Annu Rev Biochem 67:425–479.

13. Glickman MH, Ciechanover A (2002) The ubiquitin-proteasome proteolytic pathway:Destruction for the sake of construction. Physiol Rev 82:373–428.

14. Levkowitz G, et al. (1998) c-Cbl/Sli-1 regulates endocytic sorting and ubiquitination ofthe epidermal growth factor receptor. Genes Dev 12:3663–3674.

15. Miyake S, Lupher ML, Jr., Druker B, Band H (1998) The tyrosine kinase regulator Cblenhances the ubiquitination and degradation of the platelet-derived growth factorreceptor alpha. Proc Natl Acad Sci USA 95:7927–7932.

16. Lee PS, et al. (1999) The Cbl protooncoprotein stimulates CSF-1 receptormultiubiquitination andendocytosis, andattenuatesmacrophageproliferation.EMBO J18:3616–3628.

17. Yokouchi M, et al. (2001) Src-catalyzed phosphorylation of c-Cbl leads to theinterdependent ubiquitination of both proteins. J Biol Chem 276:35185–35193.

18. Zeng S, Xu Z, Lipkowitz S, Longley JB (2005) Regulation of stem cell factor receptorsignaling by Cbl family proteins (Cbl-b/c-Cbl). Blood 105:226–232.

19. Soubeyran P, Barac A, Szymkiewicz I, Dikic I (2003) Cbl-ArgBP2 complex mediatesubiquitination and degradation of c-Abl. Biochem J 370:29–34.

20. Klapper LN, Waterman H, Sela M, Yarden Y (2000) Tumor-inhibitory antibodies toHER-2/ErbB-2 may act by recruiting c-Cbl and enhancing ubiquitination of HER-2.Cancer Res 60:3384–3388.

21. Bhat A, et al. (1997) Interactions of CBL with BCR-ABL and CRKL in BCR-ABL-transformed myeloid cells. J Biol Chem 272:16170–16175.

22. Thien CB, Langdon WY (2001) Cbl: Many adaptations to regulate protein tyrosinekinases. Nat Rev Mol Cell Biol 2:294–307.

23. Swaminathan G, Tsygankov AY (2006) The Cbl family proteins: Ring leaders inregulation of cell signaling. J Cell Physiol 209:21–43.

24. Thien CB, Langdon WY (2005) c-Cbl and Cbl-b ubiquitin ligases: Substrate diversityand the negative regulation of signalling responses. Biochem J 391:153–166.

25. Stanhill A, et al. (2006) An arsenite-inducible 19S regulatory particle-associatedprotein adapts proteasomes to proteotoxicity. Mol Cell 23:875–885.

26. Joazeiro CA, et al. (1999) The tyrosine kinase negative regulator c-Cbl as a RING-type,E2-dependent ubiquitin-protein ligase. Science 286:309–312.

27. Pickart CM (2001) Mechanisms underlying ubiquitination. Annu Rev Biochem 70:503–533.

28. Zhang XW, et al. (2010) Arsenic trioxide controls the fate of the PML-RARalphaoncoprotein by directly binding PML. Science 328:240–243.

29. Jeanne M, et al. (2010) PML/RARA oxidation and arsenic binding initiate theantileukemia response of As2O3. Cancer Cell 18:88–98.

30. Waxman S, Anderson KC (2001) History of the development of arsenic derivatives incancer therapy. Oncologist 6(Suppl 2):3–10.

31. Lallemand-Breitenbach V, et al. (2008) Arsenic degrades PML or PML-RARalpha througha SUMO-triggered RNF4/ubiquitin-mediated pathway. Nat Cell Biol 10:547–555.

32. Dinulescu DM, et al. (2003) c-CBL is not required for leukemia induction by Bcr-Abl inmice. Oncogene 22:8852–8860.

33. Sanada M, et al. (2009) Gain-of-function of mutated C-CBL tumour suppressor inmyeloid neoplasms. Nature 460:904–908.

34. Li Y, et al. (2009) Arsenic trioxide induces apoptosis and G2/M phase arrest byinducing Cbl to inhibit PI3K/Akt signaling and thereby regulate p53 activation. CancerLett 284:208–215.

35. Abbas S, Rotmans G, Löwenberg B, Valk PJ (2008) Exon 8 splice site mutations in thegene encoding the E3-ligase CBL are associated with core binding factor acutemyeloid leukemias. Haematologica 93:1595–1597.

36. Caligiuri MA, et al. (2007) Novel c-CBL and CBL-b ubiquitin ligase mutations in humanacute myeloid leukemia. Blood 110:1022–1024.

37. Ito K, et al. (2008) PML targeting eradicates quiescent leukaemia-initiating cells.Nature 453:1072–1078.

38. Paez JG, et al. (2004) EGFR mutations in lung cancer: Correlation with clinical responseto gefitinib therapy. Science 304:1497–1500.

39. Marchetti A, et al. (2005) EGFR mutations in non-small-cell lung cancer: analysis ofa large series of cases and development of a rapid and sensitive method for diagnosticscreening with potential implications on pharmacologic treatment. J Clin Oncol 23:857–865.

40. Tan YH, et al. (2010) CBL is frequently altered in lung cancers: Its relationship tomutations in MET and EGFR tyrosine kinases. PLoS ONE 5:e8972.

41. Kato Y, et al. (2005) Selective activation of STAT5 unveils its role in stem cell self-renewal in normal and leukemic hematopoiesis. J Exp Med 202:169–179.

42. Mahon FX, et al. (2000) Selection and characterization of BCR-ABL positive cell lineswith differential sensitivity to the tyrosine kinase inhibitor STI571: Diversemechanisms of resistance. Blood 96:1070–1079.

43. Nagar B, et al. (2003) Structural basis for the autoinhibition of c-Abl tyrosine kinase.Cell 112:859–871.

44. Zheng N, Wang P, Jeffrey PD, Pavletich NP (2000) Structure of a c-Cbl-UbcH7 complex:RING domain function in ubiquitin-protein ligases. Cell 102:533–539.

21688 | www.pnas.org/cgi/doi/10.1073/pnas.1016311108 Mao et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

1, 2

020