Heat Shock Protein 70 (Hsp70) Suppresses RIP1- Dependent Apoptotic and Necroptotic Cascades - Molecular Cancer Research

Page created by Morris Campos
 
CONTINUE READING
Published OnlineFirst September 28, 2017; DOI: 10.1158/1541-7786.MCR-17-0408

        Cell Death and Survival                                                                                                             Molecular
                                                                                                                                            Cancer
                                                                                                                                            Research
    Heat Shock Protein 70 (Hsp70) Suppresses RIP1-
    Dependent Apoptotic and Necroptotic Cascades
    Sharan R. Srinivasan1, Laura C. Cesa1, Xiaokai Li2, Olivier Julien2, Min Zhuang2,
    Hao Shao2, Jooho Chung3, Ivan Maillard3, James A. Wells2, Colin S. Duckett4,
    and Jason E. Gestwicki2

    Abstract
       Hsp70 is a molecular chaperone that binds to "client"                            model, MDA-MB-231 breast cancer cells treated with Hsp70
    proteins and protects them from protein degradation. Hsp70                          inhibitors underwent apoptosis, while cotreatment with z-
    is essential for the survival of many cancer cells, but it is not yet               VAD.fmk switched the cell death pathway to necroptosis. In
    clear which of its clients are involved. Using structurally distinct                addition, cell death in response to Hsp70 inhibitors was
    chemical inhibitors, we found that many of the well-known                           strongly suppressed by RIP1 knockdown or inhibitors. Thus,
    clients of the related chaperone, Hsp90, are not strikingly                         these data indicate that Hsp70 plays a previously unrecognized
    responsive to Hsp70 inhibition. Rather, Hsp70 appeared to be                        and important role in suppressing RIP1 activity.
    important for the stability of the RIP1 (RIPK1) regulators:
    cIAP1/2 (BIRC1 and BIRC3), XIAP, and cFLIPS/L (CFLAR).                              Implications: These findings clarify the role of Hsp70 in prosur-
    These results suggest that Hsp70 limits apoptosis and necrop-                       vival signaling and suggest IAPs as potential new biomarkers for
    tosis pathways downstream of RIP1. Consistent with this                             Hsp70 inhibition. Mol Cancer Res; 16(1); 58–68. 2017 AACR.

    Introduction                                                                        inhibitors, including VER-155008 (8), MAL3-101 (13), and JG-
                                                                                        98 (14). These molecules belong to distinct chemical families and
       Elevated expression of Hsp70 correlates with poor survival and
                                                                                        have nonoverlapping binding sites (15). For example, JG-98 is an
    resistance to chemotherapeutics (1–4). Hsp70 is generally
                                                                                        allosteric inhibitor that binds tightly to a deep pocket (16) that is
    thought to inhibit both the extrinsic and intrinsic pathways of
                                                                                        conserved in members of the Hsp70 family (14). Importantly,
    apoptosis (5) by protecting important "clients," such as the
                                                                                        JG-98 and its analogues have been found to be relatively selective
    oncoproteins Raf-1 and Akt-1, from degradation (6–8), However,
                                                                                        for members of the Hsp70 family, based on results from pull-
    this model is largely based on analogy to the related chaperone,
                                                                                        downs (17), overexpression, and point mutations (18–21). The
    Hsp90 (9, 10). Inhibitors of Hsp90 are well known to release
                                                                                        mechanism of JG-98 is to block a key allosteric transition in
    clients from that chaperone, leading to protein degradation and,
                                                                                        Hsp70 that favors degradation of some Hsp70-bound clients (19,
    ultimately, apoptotic cell death (11, 12). It is not clear whether
                                                                                        21). Other compounds bind different locations and have distinct
    Hsp700 s activity is restricted to these "Hsp90-like" functions or
                                                                                        mechanisms (22). For example, VER-155008 competes for bind-
    whether it plays a broader or even parallel role.
                                                                                        ing of nucleotide to Hsp70 (8), and MAL3-101 binds to a distinct
       The molecular roles of Hsp70 in cancer have been elusive, in
                                                                                        allosteric site (23). Although JG-98 is relatively nontoxic (EC50 >
    part, because of a lack of selective chemical inhibitors. A number
                                                                                        20 mmol/L) to normal mouse embryonic fibroblasts, it has anti-
    of recent reports have created the first generation of Hsp70
                                                                                        proliferative activity (EC50 ¼ 400 nmol/L) in multiple cancer cell
                                                                                        lines (14) and its analogues kill tamoxifen-resistant cells (24).
                                                                                        Similar selectivity for transformed cells is observed using Hsp70
    1
    Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan.           inhibitors belonging to other chemical series (8, 25). The consis-
    2
     Department of Pharmaceutical Chemistry, University of California San               tency of this result is important because parallel activity across
    Francisco, San Francisco, California. 3The Life Sciences Institute, University of   chemically distinct molecules often suggests that the activity is
    Michigan, Ann Arbor, Michigan. 4Department of Pathology, University of              mediated by the intended target. On the basis of all of these recent
    Michigan, Ann Arbor, Michigan.
                                                                                        findings, we envisioned JG-98 and other new Hsp70 inhibitors as
    Note: Supplementary data for this article are available at Molecular Cancer         promising chemical tools for better understanding the chaper-
    Research Online (http://mcr.aacrjournals.org/).                                     one's molecular roles in cancer.
    Current address for C.S. Duckett: Baylor Scott & White Research Institute,             Using multiple, structurally distinct Hsp70 inhibitors, we
    Department of Medicine, Section of Hematology and Oncology, Baylor College          found that Hsp90 clients, such as Akt or Raf1, are only weakly
    of Medicine, Dallas, Texas.
                                                                                        degraded after treatment. Rather, the stability of the RIP1 regu-
    Corresponding Author: Jason E. Gestwicki, University of California San              lators, IAP1/2, XIAP, and cFLIPS/L, seemed especially sensitive to
    Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158. Phone: 415-502-7121;    Hsp70 activity. Indeed, in MDA-MB-231 breast cancer cells, the
    Fax: 415-476-8386; E-mail: jason.gestwicki@ucsf.edu
                                                                                        kinetics of cell death correlated better with the loss of the RIP1
    doi: 10.1158/1541-7786.MCR-17-0408                                                  regulators than with degradation of Hsp90 clients. Consistent
    2017 American Association for Cancer Research.                                     with a role in limiting RIP1 activation, treatment with Hsp70

58 Mol Cancer Res; 16(1) January 2018

          Downloaded from mcr.aacrjournals.org on March 8, 2021. © 2018 American Association for Cancer Research.
Published OnlineFirst September 28, 2017; DOI: 10.1158/1541-7786.MCR-17-0408

                                                                                                        Hsp70 Inhibition Activates Cell Death

inhibitors led to apoptotic cell death, but coadministration with        Immunoprecipitations
z-VAD-fmk switched the cells to a necroptotic pathway. Further-          Ripoptosome. Cells were pre-treated with z-VAD.fmk (20 mmol/L)
more, cell death in response to Hsp70 inhibitors required RIP1           to limit cleavage of RIP1. Following compound treatments, cells
activity, as shown using RIP1 knockdown and selective RIP1               were incubated for 24 hours before proceeding with lysis, immu-
kinase inhibitors. Thus, although Hsp70 is likely to have multiple       noprecipitation, and Western blots, as described previously (27).
clients, its activity on RIP1 seems to be especially important in cell
survival. These findings may help guide the selection of Hsp70-           Fluorescence and light microscopy
selective biomarkers and potentially accelerate the discovery of            Cells were visualized using an Olympus IX83 Inverted Micro-
clinical candidates.                                                     scope. Nuclear morphology was examined by Hoechst 33258
                                                                         staining (Sigma). For each experiment, at least 10 individual
Materials and Methods                                                    frames were examined and representative panels chosen for
Reagents and antibodies                                                  presentation. For quantification, we manually counted 200 cells
Inhibitors. The following reagents were purchased from                   from three independent experiments.
Sigma-Aldrich: necrostatin-1, bortezomib; Enzo: z-VAD.fmk;
Millipore: necrosulfonamide (NSA); LC Labs: 17-DMAG;                     Proteolytic profiling ("N-terminal degradomics")
StressMarq: VER-155008; and Teva Pharmaceuticals: etopo-                    Eight liters of T-cell leukemia Jurkat A cells were grown in four
side. JG-98, JG-84 and JG-258 were synthesized and charac-               separate 3 L spinner flasks to a density of 0.8  106 cells/mL (see
terized as described previously (14). All compounds were                 below). The cells were then treated for 16 hours with (i) 10 mmol/L
suspended in DMSO, and the final solvent concentration in                 JG98 alone; (ii) 10 mmol/L JG98 and 20 mmol/L NSA, (iii) 10
the assays was held to 1%.                                               mmol/L JG98 and 20 mmol/L of cell-permeable pan-caspase
                                                                         inhibitor zVAD(OMe)-fmk; or (iv) left untreated. For the com-
                                                                         bination treatments, the cells were pretreated with NSA or zVAD
Antibodies. The following antibodies were purchased from Enzo:
                                                                         for 1 hour prior to the addition of JG98. Cell growth was
Hsp72 (C92F3A-5), XIAP (ADI-AAM-050), c-IAP1 (ALX-803-
                                                                         monitored using a Scepter cell counter (Millipore) to estimate
335), caspase-8 (ALX-804-429); SCBT: GAPDH (sc-32233),
                                                                         the amount of cell death and cell morphology changes monitored
Hsp90 (sc-7947), Raf-1 (sc-133), caspase-8 (sc-6136), anti-rat
                                                                         under a Zeiss Observer Z1 microscope to confirm the expected
(sc-2006), goat IgG (sc-2028); Cell Signaling Technology: Akt-1
                                                                         phenotype. Free N-termini were detected as described previously
(2967), cleaved caspase-3 (9664), c-IAP2 (3130); BD Pharmin-
                                                                         (see below). In short, cell lysates were collected and the cells lysed
gen: RIP1 (610459), Cdk4 (559693), cytochrome c (556433),
                                                                         using 0.1% Trion X-100 (0.8–1.2  109 cells per sample), free N-
Bcl-xL (610746); Molecular Probes: COXIV (A21347); Alexis:
                                                                         termini biotinylated using a cleavable peptide ester (TEVest4B)
FLIP (ALX-804-428); Millipore: Smac (567365).
                                                                         and subtiligase, protein fragments captured on Neutravidin beads
                                                                         and trypsin-digested. Each sample was then fractionated by HPLC
Tissue culture
                                                                         into 12 fractions, for a total of 48 LC/MS-MS injections on
   MDA-MB-231 WT cells were maintained in DMEM (Invitro-
                                                                         an Orbitrap Velos Pro (Thermo Fisher Scientific). Peptide iden-
gen), supplemented with 10% FBS, 1% penicillin–streptomycin,
                                                                         tification was performed using Protein Prospector (v. 5.10.15 or
and nonessential amino acids. Jurkat cells were grown in
                                                                         higher; University of California, San Francisco, CA). All spectra
RPMI1640 (Corning), supplemented with GlutaMax. MDA-
                                                                         were searched using the full human SwissProt database (down-
MB-231 and Jurkat cells overexpressing Bcl-xL and RIP1 knockout
                                                                         loaded June 27, 2013) with random sequence database for
Jurkat cells were all created as described previously (26).
                                                                         FDR determination. Search parameters included: fixed modifica-
                                                                         tion cysteine carbamidomethylation; variable modifications
Cell line authentication
                                                                         N-terminal aminobutyric acid, methionine-loss (N-terminus)
  All cell lines were purchased from ATCC and experiments
                                                                         and methionine oxidation; up to one missed tryptic cleavages;
performed on cells that were passaged less than 20 times.
                                                                         C-terminal trypsin cleavage; nonspecific cleavage N-terminus;
                                                                         parent mass tolerance 30 ppm; fragment mass tolerance 0.5 Da.
Cell growth assays
                                                                         Expectation value cutoff was adjusted to maintain
Published OnlineFirst September 28, 2017; DOI: 10.1158/1541-7786.MCR-17-0408

    Srinivasan et al.

    Figure 1.
    Inhibition of Hsp70 causes rapid antiproliferative activity. A, Chemical structures of Hsp70 inhibitors (JG-98, JG-84, MAL3-101, and VER-155008), controls (JG-258)
    and inhibitors of Hsp90 (17-DMAG), and the proteasome (bortezomib). B, JG-98 (5 mmol/L) suppresses growth of MDA-MB-231 cancer cells with relatively
    rapid kinetics compared with 17-DMAG (5 mmol/L) or bortezomib (40 nmol/L), as monitored by MTT assays. Results are the average of two independent experiments
    performed in quintuplicates. Error bars, SEM. C, Hsp70 inhibitors induce rapid cell growth effects, as measured by CellTiter Glo. Results are the average of
    experiments performed in triplicate. Errors, SEM. D, Chaperone clients are degraded relatively late after treatment with JG-98 (10 mmol/L), after onset of effects on
    cell growth. Results are representative of experiments performed in duplicate.

    longer times, approximately 18 to 24 hours (Fig. 1B). To confirm                     apoptosis. This is an important step because MTT and CellTi-
    this result using a distinct cell growth measure, we also performed                 terGlo assays primarily report on proliferation and not neces-
    CellTiterGlo assays on treated MDA-MB-231 cells and found that                      sarily on cell death. Indeed, we found that JG-98 induced
    the kinetics in response to multiple, different Hsp70 inhibitors:                   classical apoptosis features, including morphologic changes
    JG-98, JG-84, VER-155008 or MAL3-101, was markedly faster                           consistent with programmed cell death (Supplementary Fig.
    than treatment with Hsp90 inhibitors (Fig. 1C). As mentioned                        S1A) and positive Annexin staining (Supplementary Fig. S1B).
    above, it was important to note that Hsp70 inhibitors from                          This result is consistent with previous reports that JG-98 acti-
    multiple chemical series showed similar responses, giving confi-                     vates caspase cleavage (14). To further probe this idea, we
    dence in the role of that target. It is well known that treatment with              overexpressed the Bcl-2 family member, Bcl-xL, a mitochondrial
    Hsp90 inhibitors leads to degradation of Hsp90 clients, such as                     antiapoptotic protein, in MDA-MB-231 cells. These cells are
    Akt, Raf-1, and Cdk4, and that the kinetics of that process parallels               considered type II cells that undergo apoptosis through cas-
    cell death (28, 29). To see whether these same clients were                         pase-8 and the mitochondrial pathway (30), so high Bcl-xL
    involved in Hsp70-mediated cell proliferation, we performed                         levels would be expected to block JG-98–mediated cell death.
    Western blots on JG-98–treated lysates. Surprisingly, we found                      Importantly, we also performed parallel studies in leukemia-
    that it took approximately 12 to 24 hours for the levels of Akt, Raf-               derived Jurkat cells, because it is known that overexpression of
    1, and Cdk4 to be reduced (Fig. 1D), which is relatively late into                  Bcl-xL only incompletely inhibits RIP1-depedent cell death in
    the response. This result suggested that the mechanism(s) of                        MDA-MB-231 cells (27). Surprisingly, we found that overex-
    Hsp70 inhibitors might not be simply explained by "Hsp90-like"                      pression of Bcl-xL did not impede cytotoxicity in response to JG-
    effects in these cells.                                                             98 in either cell type (Fig. 2A and B). As controls, we confirmed
                                                                                        that Bcl-xL overexpression partially suppressed cell death in
    Inhibitors of Hsp70 induce apoptosis, independent of                                response to the control molecules, 17-DMAG, bortezomib, or
    Bcl-xL status                                                                       etoposide, in either MDA-MB-231 (see Fig. 2A) or Jurkat
      To better understand the response to Hsp70 inhibition, we                         (see Fig. 2B) cells. To understand the impact of Bcl-xL over-
    then asked whether treated MDA-MB-231 cells underwent                               expression on the potency of the compounds, we performed

60 Mol Cancer Res; 16(1) January 2018                                                                                                   Molecular Cancer Research

          Downloaded from mcr.aacrjournals.org on March 8, 2021. © 2018 American Association for Cancer Research.
Published OnlineFirst September 28, 2017; DOI: 10.1158/1541-7786.MCR-17-0408

                                                                                                                   Hsp70 Inhibition Activates Cell Death

                                                                                     The mechanism of cell death did not seem to be affected,
                                                                                     because treatment of the Bcl-xL–overexpressing cells with JG-
                                                                                     98 led to morphologic features that were still consistent with
                                                                                     apoptosis (Supplementary Fig. S2A). On the basis of the mito-
                                                                                     chondrial retention of COX IV in treated MDA-MB-231 cells,
                                                                                     this apoptotic activity also seemed to occur without global
                                                                                     disruption of mitochondria (Supplementary Fig. S2B). Thus,
                                                                                     inhibition of Hsp70 appeared to initiate a mitochondrial death
                                                                                     pathway that was independent of Bcl-2 family status.

                                                                                     When caspase-mediated apoptosis is blocked, Hsp70 inhibitors
                                                                                     trigger an alternative, necroptosis pathway
                                                                                        The caspase inhibitor, z-VAD.fmk, is known to partially
                                                                                     suppress cell death in response to Hsp90 inhibitors (31).
                                                                                     However, we found that this compound did not block prolif-
                                                                                     eration induced by JG-98 (Fig. 3A). Rather, we observed that
                                                                                     51%  13% cells pretreated with z-VAD.fmk before addition of
                                                                                     JG-98 displayed a swollen cytoplasm containing numerous
                                                                                     granules (Fig. 3B). This morphology was not observed in the
                                                                                     absence of v-VAD.fmk. The swollen cytoplasm feature is rem-
                                                                                     iniscent of necrotic cell death, suggesting that JG-98 might be
                                                                                     activating an alternative pathway if apoptosis was blocked.
                                                                                     Consistent with this idea, we observed nuclear fragmentation
                                                                                     in only 4%  1% of cells that were pretreated with z-VAD.fmk
                                                                                     prior to JG-98, compared with 40%  16% in the absence of z-
                                                                                     VAD.fmk (Fig. 3C). To further explore whether JG-98 might be
                                                                                     activating an alternative death pathway, we profiled the peptide
                                                                                     fragments produced from proteolysis in cells treated with JG-98,
                                                                                     using a recently described mass spectrometry–based method
                                                                                     termed "N-terminal degradomics" (Fig. 4A; ref. 32). Briefly, this
                                                                                     method profiles the neopeptide termini that are produced
                                                                                     by proteolysis during cell death. Treatment of Jurkat cells with
                                                                                     JG-98 produced a fragment profile consistent with caspase
                                                                                     activation, as judged by the prominent aspartate in the P1
                                                                                     position (Fig. 4B and C; Supplementary Table S1). As expected,
                                                                                     treatment with z-VAD.fmk nearly completely blocked this sig-
                                                                                     nature. Moreover, when z-VAD.fmk was used as a cotreatment
                                                                                     with JG-98, it also suppressed the caspase response (Fig. 4B and
                                                                                     C), with the peptide profile resembling the untreated cells.
                                                                                     These results support the idea that treatment with JG-98
                                                                                     initiates caspase-mediated apoptosis, but that the cells die by
                                                                                     an alternative, necroptosis, pathway when caspases are inhib-
                                                                                     ited. Perhaps more broadly, these findings also show that
Figure 2.                                                                            necroptosis occurs in the absence of dramatic proteolysis, a
Hsp70 inhibitor activity is independent of Bcl status. A, Overexpression of Bcl-xL   feature of this alternative cell death pathway that had not
does not block JG-98 cytotoxicity. MDA-MB-231 cells were treated for 24 hours        previously been described.
with JG-98 (10 mmol/L), 17-DMAG (10 mmol/L), bortezomib (40 nmol/L), or
etoposide (20 mmol/L). MTT results are the mean average of triplicates, and          Hsp70 limits RIP1 activation by stabilizing E3 ligases
error bars represent SEM.  , P < 0.05. B, Jurkat cells overexpressing Bcl-xL were
                                                                                        RIP1 is an important signaling regulator that directs cells into
treated with indicated compounds for 24 hours. Viability was determined by
Trypan blue staining. Results are the average of two experiments performed in
                                                                                     either the apoptotic or necroptotic cascades (27, 33). Because
triplicate. Error, SEM.  , P < 0.01. C, Bcl-xL overexpression provides partial     JG-98 appeared to initiate both pathways, we hypothesized that
resistance to compounds, except JG-98. MDA-MB-231 cells were treated for 72          RIP1 might be important in Hsp70-mediated cell survival.
hours and cell growth determined by MTT assays. Results are the average of two       To test this idea, we treated Jurkat cells lacking RIP1. Strikingly,
independent experiments performed in triplicate. Error, SEM.                         JG-98 was not cytotoxic in RIP1 knockout Jurkat cells (Fig. 5A).
                                                                                     Similarly, necrostatin-1, an inhibitor of RIP10 s kinase activity
                                                                                     (34), protected MDA-MB-231 cells from treatment with JG-98
dose dependence studies in MDA-MB-231 cells and calculated                           (Fig. 5B). Conversely, treatment with 17-DMAG, bortezomib,
EC50 values. Consistent with the single-concentration data, we                       or etoposide was not affected by loss of RIP1 protein or RIP1
found that overexpression of Bcl-xL did not alter the EC50 for                       activity (see Fig. 5A and B). Thus, cell death by JG-98 appeared
JG-98 (Fig. 2C); whereas the EC50s of 17-DMAG, bortezomib,                           to be uniquely dependent on RIP1 kinase activity. RIP1 is
and etoposide were significantly increased (2.5- to 5.7-fold).                        constitutively ubiquitinated by the E3 ubiquitin ligases XIAP,

www.aacrjournals.org                                                                                                  Mol Cancer Res; 16(1) January 2018    61

      Downloaded from mcr.aacrjournals.org on March 8, 2021. © 2018 American Association for Cancer Research.
Published OnlineFirst September 28, 2017; DOI: 10.1158/1541-7786.MCR-17-0408

    Srinivasan et al.

    Figure 3.
    Inhibition of caspases with z-VAD-fmk does not block JG-980 s activity. A, Inhibition of caspases by z-VAD.fmk does not suppress the activity of JG-98. Results are the
    average of three independent experiments performed in quintuplicate. Error bars, SEM. Cells were pretreated with z-VAD.fmk (40 mmol/L) for 1 hour prior
    to addition of compounds (same concentrations as in A and MTT assays performed after 24 hours.   , P < 0.01. ns, not significant. Error, SEM. B, Cells were treated
    as in A and visualized using an Olympus IX83 Inverted Microscope. Black arrows, apoptotic cells; white arrows, necrotic cells. C, Hoechst 33258 staining of
    MDA-MB-231 cells pretreated with z-VAD.fmk (40 mmol/L) for 1 hour prior to addition of JG-98 (10 mmol/L) for 24 hours. Cells pre-treated with z-VAD.fmk show no
    nuclear fragmentation (white arrows), even after coadministration of JG-98. Scale bar, 10 mm.

    c-IAP1, c-IAP2, and cFLIPS/L (35). Ubiquitination of RIP1 is                         Blocking caspase-mediated apoptosis and necroptosis prevents
    linked to turnover of the kinase, but also to nondegradation                         cell death in response to Hsp70 inhibitors
    pathways that modulate RIP1 activity (36). Indeed, ubiquitina-                          Next, we wondered whether blocking necroptosis would make
    tion of RIP1 is thought to protect against necroptosis (37).                         cells resistant to JG-98. However, we found that inhibition of this
    Hsp70 is known to protect a number of proteins from degra-                           pathway by NSA did not block JG-98 effects on proliferation (Fig.
    dation, so it seems possible that it may normally stabilize RIP1                     6A). Furthermore, the proteolytic fragment signature of these
    itself or the E3 ligases. To test this idea, we treated MDA-MB-                      treated cells suggested that they may still die by caspase-mediated
    231 cells with JG-98 and examined the levels of RIP1 and its E3                      apoptosis (see Fig. 4D). Then, to understand if blocking both
    ligases by Western blots. Although inhibition of Hsp70 did not                       apoptosis and necroptosis might limit JG-98 mediated activity, we
    significantly impact RIP1 protein levels under these conditions,                      pretreated MDA-MB-231 (Fig. 6B) or Jurkat cells (Fig. 6C) with
    it induced a striking loss of XIAP, c-IAP1/2, and cFLIPS/L (Fig.                     both z-VAD.fmk and NSA. We found that the combination
    5C). To test whether this pathway might be linked to JG-98                           partially blocked JG-98–mediated cell death. Importantly, a dis-
    treatment, we examined the kinetics by which the levels of c-                        tinct Hsp70 inhibitor, VER-155008, also required cotreatment
    IAP, XIAP, and cFLIPS/L are reduced. Indeed, loss of the E3                          with both z-VAD-fmk and NSA to protect against activity in
    ubiquitin ligases closely paralleled the kinetics of cell death,                     MDA-MB-231 cells (Supplementary Fig. S4A). To explore this
    with cFLIPS levels diminished within 1 to 3 hours and the levels                     phenomenon more broadly, we tested JG-98 in combination with
    of the other ligases beginning to decrease between 6 and 12                          v-VAD.fmk or z-VAD.fmk plus NSA in cancer cells derived from a
    hours (Fig. 5D). This result sharply contrasts with the delay in                     variety of tissues. Consistent with the results from the MDA-MB-
    Hsp90 client destabilization that we observed earlier (see Fig. 1B),                 231 and Jurkat cells, z-VAD.fmk alone was unable to protect cells
    suggesting that Hsp70 regulation of RIP1 may be more closely                         derived from breast (MCF7, SK-BR-3, T-47D), lung (A549) or
    linked to activity in these cells. To understand the mechanism of                    colon (HT-29; Supplementary Fig. S4B), while cervical-derived
    turnover, we deleted the RING domain from XIAP (XIAPDRING) and                       HeLa cells were partially protected (EC50 increased 2-fold). Next,
    compared its stability with full-length XIAP (XIAPFL) in the pres-                   we used the combination of z-VAD.fmk and NSA and found that
    ence of JG-98 in MDA-MB-231 cells (Supplementary Fig. S3A).                          both were required to protect MDA-MB-231, MCF7, SK-BR-3, and
    XIAP is known to degrade by self-ubiquitination through this                         Jurkat cells. Interestingly, A549, T47-D, and HT-29 cells were not
    domain (38, 39). We found that XIAPDRING was resistant to                            fully protected by this combination, and the effect of JG-98 may
    treatment with JG-98 (Supplementary Fig. S3B), consistent with                       have even been mildly exacerbated (see Supplementary Fig. S4B).
    activation of its normal turnover pathway through the ubiquitin                      These results suggest that Hsp70 may play additional roles in
    proteasome system. Finally, we wanted to test whether Hsp70                          some cell types, perhaps through additional mechanisms such as
    inhibitors from different structural classes also trigger loss of the E3             lysosomal cell death (40).
    ligases. These types of experiments are important for confirming
    on-target activity because, as mentioned above, the different inhi-                  Hsp70 blocks oligomerization and activation of RIP1
    bitors have distinct chemical structures, and they bind different                       Finally, we wanted to explore additional, possible mechanisms
    allosteric sites on the chaperone (15). Indeed, we found that                        by which Hsp70 could inhibit RIP1 activation. One major role for
    treatment with JG-84, VER-155008, or MAL3-101 also destabilized                      RIP1 is as a central scaffold in the TNF-receptor 1 (TNF-R1)
    c-IAP1 in MDA-MB-231 cells, while a structurally-related negative                    complex (33). It has been shown that autocrine TNF–producing
    control, JG-258, had no effect (Supplementary Fig. S3C).                             cells, such as MDA-MB-231, are sensitive to small-molecule

62 Mol Cancer Res; 16(1) January 2018                                                                                                    Molecular Cancer Research

          Downloaded from mcr.aacrjournals.org on March 8, 2021. © 2018 American Association for Cancer Research.
Published OnlineFirst September 28, 2017; DOI: 10.1158/1541-7786.MCR-17-0408

                                                                                                                           Hsp70 Inhibition Activates Cell Death

Figure 4.
Proteolytic signature of cells treated with JG-98 supports activation of both an apoptotic and alternative cell death pathway. A, Schematic overview of the proteolytic
profiling study (aka "N-terminal degradomics"). B, Overview of the peptide fragments produced in response to compound treatments. More than 45% of the
substrates identified after JG-98 treatment feature a P1 ¼ D, suggesting the activation of caspases. In general, there was minimal proteolysis in untreated
cells or cells treated with JG-98 and z-VAD. Aminobutyric acid (Abu). C, Logo plots show that JG-98 activated apoptosis in Jurkat cells, and NSA is unable to block this
activity. However, z-VAD reversed the proteolytic profile to one that approximated untreated cells. D, The caspase activation profile (P1 ¼ D peptides)
was similar between cells treated with JG-98 or JG-98 plus NSA, while the pattern of peptides (P1 ¼ all) was similar between JG98 þ zVAD and untreated cells.
Experiments repeated in biological duplicate and technical triplicate.

mimetics of the Smac/DIABLO protein and that this cytotox-                            inhibits the formation of amyloids formed from diverse pro-
icity proceeds through TNF-R1 (27). We investigated this pos-                         teins, such as amyloid beta, huntingtin, and tau (42). Thus, it
sibility for JG-98 and found that a neutralizing antibody against                     seemed logical that Hsp70 might counteract conversion of RIP1
TNF-R1 (Enbrel) blocked cell death by a Smac mimetic (SM),                            into its amyloid state, such that treatment with JG-98 might
but that it was unable to prevent JG-98–induced cell death (Fig.                      relieve this suppression. To test this idea, we treated MDA-MB-
7A). Thus, JG-98 appears to act independently of TNF-R1, but                          231 cells with JG-98 or the combination of JG-98 and z-VAD.
with a mechanism distinct from Smac mimetics. A cytosolic                             fmk (to induce necroptosis) and looked for stable, high molec-
death complex containing RIP1, termed the ripoptosome, has                            ular mass oligomers of RIP1. We found that treatment with
been also shown to direct both apoptosis and necroptosis (27).                        JG-98 promoted conversion of RIP1 to the oligomeric state,
A key event in formation of the ripoptosome is binding of RIP1                        concurrent with depletion of its binding partner, RIP3 (Fig.
to caspase-8. To determine whether treatment with JG-98                               7C). Similar effects were observed with the other Hsp70 inhi-
triggered this event, we immunoprecipitated caspase-8 and                             bitors, JG-84 and VER-155008 (Supplementary Fig. S5). Again,
performed Western blots with a RIP1 antibody. Although Smac                           this mechanism was distinct from that used by SM-164, which
mimetic gave the expected result, JG-98 did not promote the                           had no effect on RIP1 oligomerization in this system (see Fig.
interaction (Fig. 7B), suggesting that JG-98 does not trigger                         7D). Furthermore, RIP1 oligomerization did require kinase
formation of the ripoptosome. Finally, we tested whether                              activity in this context, as Nec-1 could block the process.
Hsp70 might play a role in RIP1 oligomerization. It has recently                      Importantly, treatment with 17-DMAG or bortezomib did not
been reported that RIP1 and RIP3 forms stable, oligomeric,                            impact RIP1 oligomerization (Fig. 7D). Together, these results
amyloid-like structures that are required for necroptosis (41),                       suggested that Hsp70 plays a key role in suppressing RIP1
which appear as stable, high molecular mass bands on SDS-                             activation, possibly through effects on the stability of its E3
PAGE. At the same time, it is well known that Hsp70 broadly                           ligases and on its self-association.

www.aacrjournals.org                                                                                                         Mol Cancer Res; 16(1) January 2018            63

      Downloaded from mcr.aacrjournals.org on March 8, 2021. © 2018 American Association for Cancer Research.
Published OnlineFirst September 28, 2017; DOI: 10.1158/1541-7786.MCR-17-0408

    Srinivasan et al.

    Figure 5.
    JG-98 activity occurs through a RIP1-dependent process. A, RIP1 KO Jurkat cells are resistant to JG-98. Viability was determined by Trypan blue exclusion. Cells were
    treated for 24 hours with JG-98 (10 mmol/L), 17-DMAG (10 mmol/L), bortezomib (40 nmol/L), etoposide (20 mmol/L). Results are the average of three independent
    experiments performed in triplicate. ns, not significant;  P < 0.05. B, JG-98 activity requires RIP1 kinase. MDA-MB-231 cells were pretreated with 20 mmol/L
    necrostatin-1 for 1 hour prior to addition of compounds. Viability was determined by three independent MTT assays performed in quintuplicate. Error, SEM. ns, not
    significant;    , P < 0.0001. C, JG-98 induces degradation of RIP1 modulators. MDA-MB-231 cells were treated for 24 hours. Results represent experiments
    performed in triplicate. D, RIP1 regulators are rapidly degraded in response to JG-98. MDA-MB-231 cells were treated with JG-98 (10 mmol/L). Results are
    representative of duplicates.

                                                                                        ment of Hsp70 inhibitors by multiple research groups. By anal-
    Discussion                                                                          ogy, the clients of Hsp90 were revealed after the serendipitous
       Knockdown of Hsp70 has been shown to reduce proliferation                        discovery of geldanamycin and the development of its more
    and enhance sensitivity to chemotherapy in multiple cancer                          potent analogues, such as 17-DMAG (46). Thus, given recent
    models (1–4). Because Hsp70 itself is not an oncogene, it was                       advances in Hsp70 inhibitor discovery, it seemed timely to ask
    presumed that it acts through a "nononcogene addiction" mech-                       whether Hsp70 might have its own clients. Here, we used a suite of
    anism, in which the chaperone stabilizes a subset of oncogenes to                   Hsp70 inhibitors with different binding sites and mechanisms to
    protect against cell death (6, 43). Hsp90 is known to play a similar                explore Hsp700 s prosurvival function. We focused on MDA-MB-
    functional role (12), yet more is known about the mechanisms by                     231 and Jurkat cells, because of existing genetic data on the
    which that chaperone creates a nononcogene addiction. Specif-                       importance of Hsp70 in these cells, but it is important to stress
    ically, Hsp90 is known to directly bind and stabilize a subset of                   that Hsp70 may play additional roles in other cells (see Supple-
    prosurvival clients, including Akt, Her-2, Raf-1, and Cdk4 (44).                    mentary Fig. S4B).
    Although individual clients of Hsp70 have been suggested (6–8)                         Our results suggest an unexpected model in which Hsp70
    and some shared clients seem likely (45), we wondered whether                       protects against cell death through suppressing RIP1 activation.
    these two chaperones might also have distinct clients. This ques-                   In part, this mechanism appears to involve Hsp70-mediated
    tion became possible to address, thanks to the recent develop-                      stabilization of the E3 ubiquitin ligases of RIP1, such as c-IAPs

64 Mol Cancer Res; 16(1) January 2018                                                                                                   Molecular Cancer Research

          Downloaded from mcr.aacrjournals.org on March 8, 2021. © 2018 American Association for Cancer Research.
Published OnlineFirst September 28, 2017; DOI: 10.1158/1541-7786.MCR-17-0408

                                                                                                     Hsp70 Inhibition Activates Cell Death

Figure 6.
JG-98 activity requires both apoptosis
and necroptosis. A, Inhibition of
necroptosis alone does not restore
growth of cells treated with JG-98.
Cells were pretreated with NSA (20
mmol/L) for 1 hour prior to addition of
compounds. Error, SEM. B, Inhibition
of both apoptosis and necroptosis is
necessary to fully suppress JG-98
activity. Cells were pretreated with
both 40 mmol/L z-VAD.fmk and 20
mmol/L NSA for 1 hour prior to addition
of compounds. Cell growth was
determined by three independent
MTT assays performed in
quintuplicate.  , P < 0.05;   , P < 0.01;

    , P < 0.001; ns, not significant. C,
Jurkat cells were pretreated with
z-VAD.fmk (40 mmol/L), NSA, both,
or necrostatin-1 for 1 hour prior to
addition of JG-98 (10 mmol/L).
Inactivation of both apoptosis and
necroptosis was necessary to prevent
activity. Cell viability was determined
by Trypan blue exclusion. Results are
the average of three independent
experiments performed in triplicate.

and XIAP (see Fig. 5). Importantly, we found that the kinetics of      key upstream "hub" of multiple cell death pathways. Indeed, our
cell death correlate better with loss of these E3 ligases than with    results suggest that, at least in MDA-MB-231 and Jurkat cells, this
the turnover of the classical Hsp90 clients, such as Raf-1. This       mechanism might be particularly important because RIP1 knock-
result could be particularly important for the future of Hsp70         down or cotreatment with necrostatin was sufficient to completely
inhibitor development, as these E3 ligases might be biomarkers         suppress JG-980 s activity. However, many important mysteries
for Hsp70 engagement. Again, an analogy to Hsp90 is illuminat-         remain. For example, we observed that cell death in response to
ing because loss of the Hsp90 clients, such as Raf-1 and Akt, is       Hsp70 inhibitor was independent of Bcl-xL, and it was coupled
often used to estimate engagement of that target. Another mech-        with an unusual relationship to cytochrome c release. Further-
anism of Hsp70-mediated cell survival appeared to involve sup-         more, it has been shown that RIP1 itself is a client of Hsp90 (48)
pression of RIP1/3 amyloid formation. Specifically, we found that       and that Hsp90 inhibitors can impact necroptosis in fibrosarcoma
treatment with JG-98 or other Hsp70 inhibitors relieved this           cells (49). Thus, it seems compelling to envision that Hsp90 and
suppression and allowed RIP1 oligomers to form (see Fig. 7). In        Hsp70 may cooperate in suppressing the RIP1 pathway, in a
retrospect, this finding might have been expected, based on the         currently unknown way. It is even possible that combinations
widespread role of Hsp70 in preventing amyloid formation (47).         of Hsp70 and Hsp90 inhibitors could be synergistic in this setting.
Although this mechanism has been best described in the neuro-             Finally, this work may accelerate the search for drug candidates
degenerative disease literature, it seems logical that it would also   that target Hsp70. On the basis of expression data, biochemical
play this part in cancer.                                              studies and knockdowns, members of the Hsp70 family appear to
   Hsp70 has been linked to a wide number of cell survival             be compelling, potential drug targets (50, 51). However, progress
aspects. We hypothesize that some of these activities might be         has been slowed by lack of mechanistic knowledge. Our results are
consolidated and understood through the functions on RIP1, a           potentially important in that context because they suggest that IAP

www.aacrjournals.org                                                                                   Mol Cancer Res; 16(1) January 2018    65

       Downloaded from mcr.aacrjournals.org on March 8, 2021. © 2018 American Association for Cancer Research.
Published OnlineFirst September 28, 2017; DOI: 10.1158/1541-7786.MCR-17-0408

    Srinivasan et al.

    Figure 7.
    Hsp70 limits RIP1/3 oligomerization. A, JG-98 toxicity does not depend on TNF signaling. MDA-MB-231 cells were pretreated with Enbrel (5 mg/mL) for 1 hour prior to
    addition of compounds. Viability was determined by MTT assay. Results are the average of three independent experiments performed in quintuplicate. Error,
    SEM.   , P < 0.01. B, JG-98 does not induce formation of a RIP1–caspase-8 complex. MDA-MB-231 cells were treated for 24 hours with indicated compounds.
    SM-164 was used at 100 nmol/L. C, Treatment with JG-98 (10 mmol/L) favored formation of a high molecular mass oligomer of RIP1 and a reduction of RIP3.
    Cotreatment with z-VAD-fmk exacerbated this effect. Necrostatin could block the effects of JG-98. Results are representative of experiments performed
    in triplicate. D, Treatment with 17-DMAG or bortezomib (BTZ) did not change RIP1 oligomerization.

    family members may be biomarkers of Hsp70 inhibition. If that                      Authors' Contributions
    finding is found to be robust across many cell types and by                         Conception and design: S.R. Srinivasan, L.C. Cesa, X. Li, J.E. Gestwicki
    different groups, it would be expected to boost efforts to discover,               Development of methodology: S.R. Srinivasan, H. Shao
    optimize, and deploy clinical candidates to test the central                       Acquisition of data (provided animals, acquired and managed patients, pro-
                                                                                       vided facilities, etc.): S.R. Srinivasan, L.C. Cesa, X. Li, O. Julien, H. Shao, I. Maillard,
    hypothesis of Hsp70 as an anticancer target.                                       M. Zhuang, J. Chung
                                                                                       Analysis and interpretation of data (e.g., statistical analysis, biostatistics,
    Disclosure of Potential Conflicts of Interest                                       computational analysis): S.R. Srinivasan, L.C. Cesa, X. Li, O. Julien, M. Zhuang,
       No potential conflicts of interest were disclosed.                               H. Shao, J.E. Gestwicki

66 Mol Cancer Res; 16(1) January 2018                                                                                                        Molecular Cancer Research

          Downloaded from mcr.aacrjournals.org on March 8, 2021. © 2018 American Association for Cancer Research.
Published OnlineFirst September 28, 2017; DOI: 10.1158/1541-7786.MCR-17-0408

                                                                                                                        Hsp70 Inhibition Activates Cell Death

Writing, review, and/or revision of the manuscript: S.R. Srinivasan, L.C. Cesa,       The authors would like to thank the members of the Gestwicki and Duckett
X. Li, H. Shao, C.S. Duckett, J.E. Gestwicki                                       laboratories for assistance. MAL3-101 was supplied by Jeff Brodsky (U. Pittsburgh).
Administrative, technical, or material support (i.e., reporting or organizing
data, constructing databases): S.R. Srinivasan,                                      The costs of publication of this article were defrayed in part by the pay-
Study supervision: J.E. Gestwicki, J.A. Wells, C.S. Duckett                        ment of page charges. This article must therefore be hereby marked advertise-
                                                                                   ment in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
Acknowledgments
  Funding for this work was provided by the NIH (NS059690) and the                   Received July 27, 2017; revised September 10, 2017; accepted September 22,
University of Michigan's Comprehensive Cancer Center (CA046592).                   2017; published OnlineFirst September 28, 2017.

References
 1. Nylandsted J, Brand K, Jaattela M. Heat shock protein 70 is required for the   20. Young ZT, Rauch JN, Assimon VA, Jinwal UK, Ahn M, Li X, et al. Stabilizing
    survival of cancer cells. Ann N Y Acad Sci 2000;926:122–5.                         the Hsp70-tau complex promotes turnover in models of tauopathy. Cell
 2. Nanbu K, Konishi I, Mandai M, Kuroda H, Hamid AA, Komatsu T, et al.                Chem Biol 2016;23:992–1001.
    Prognostic significance of heat shock proteins HSP70 and HSP90 in               21. Wang AM, Miyata Y, Klinedinst S, Peng HM, Chua JP, Komiyama T, et al.
    endometrial carcinomas. Cancer Detect Prev 1998;22:549–55.                         Activation of Hsp70 reduces neurotoxicity by promoting polyglutamine
 3. Rohde M, Daugaard M, Jensen MH, Helin K, Nylandsted J, J€a€attel€a M.              protein degradation. Nat Chem Biol 2013;9:112–8.
    Members of the heat-shock protein 70 family promote cancer cell growth         22. Li X, Shao H, Taylor IR, Gestwicki JE. Targeting allosteric control mechan-
    by distinct mechanisms. Genes Dev 2005;19:570–82.                                  isms in heat shock protein 70 (Hsp70). Curr Top Med Chem 2016;
 4. Ciocca DR, Calderwood SK. Heat shock proteins in cancer: diagnostic,               16:2729–40.
    prognostic, predictive, and treatment implications. Cell Stress Chaperones     23. Wisen S, Bertelsen EB, Thompson AD, Patury S, Ung P, Chang L, et al.
    2005;10:86–103.                                                                    Binding of a small molecule at a protein-protein interface regulates
 5. Brodsky JL, Chiosis G. Hsp70 molecular chaperones: emerging roles in               the chaperone activity of hsp70-hsp40. ACS Chem Biol 2010;5:
    human disease and identification of small molecule modulators. Curr Top             611–22.
    Med Chem 2006;6:1215–25.                                                       24. Koren J 3rd, Miyata Y, Kiray J, O'Leary JC 3rd, Nguyen L, Guo J, et al.
 6. Davenport EL, Zeisig A, Aronson LI, Moore HE, Hockley S, Gonzalez D,               Rhodacyanine derivative selectively targets cancer cells and overcomes
    et al. Targeting heat shock protein 72 enhances Hsp90 inhibitor-induced            tamoxifen resistance. PLoS One 2012;7:e35566.
    apoptosis in myeloma. Leukemia 2010;24:1804–7.                                 25. Taldone T, Kang Y, Patel HJ, Patel MR, Patel PD, Rodina A, et al. Heat shock
 7. Powers MV, Clarke PA, Workman P. Dual targeting of HSC70 and HSP72                 protein 70 inhibitors. 2. 2,50 -thiodipyrimidines, 5-(phenylthio)pyrimi-
    inhibits HSP90 function and induces tumor-specific apoptosis. Cancer Cell           dines, 2-(pyridin-3-ylthio)pyrimidines, and 3-(phenylthio)pyridines as
    2008;14:250–62.                                                                    reversible binders to an allosteric site on heat shock protein 70. J Med
 8. Williamson DS, Borgognoni J, Clay A, Daniels Z, Dokurno P, Drysdale                Chem 2014;57:1208–24.
    MJ, et al. Novel adenosine-derived inhibitors of 70 kDa heat shock             26. Galban S, Hwang C, Rumble JM, Oetjen KA, Wright CW, Boudreault A, et al.
    protein, discovered through structure-based design. J Med Chem 2009;               Cytoprotective effects of IAPs revealed by a small molecule antagonist.
    52:1510–3.                                                                         Biochem J 2009;417:765–71.
 9. da Silva VC, Ramos CH. The network interaction of the human cytosolic 90       27. Tenev T, Bianchi K, Darding M, Broemer M, Langlais C, Wallberg F, et al.
    kDa heat shock protein Hsp90: a target for cancer therapeutics. J Proteomics       The Ripoptosome, a signaling platform that assembles in response to
    2012;75:2790–802.                                                                  genotoxic stress and loss of IAPs. Mol Cell 2011;43:432–48.
10. Jolly C, Morimoto RI. Role of the heat shock response and molecular            28. Chandarlapaty S, Sawai A, Ye Q, Scott A, Silinski M, Huang K, et al.
    chaperones in oncogenesis and cell death. J Natl Cancer Inst 2000;92:              SNX2112, a synthetic heat shock protein 90 inhibitor, has potent antitu-
    1564–72.                                                                           mor activity against HER kinase-dependent cancers. Clin Cancer Res
11. Matts RL, Brandt GE, Lu Y, Dixit A, Mollapour M, Wang S, et al. A systematic       2008;14:240–8.
    protocol for the characterization of Hsp90 modulators. Bioorg Med Chem         29. Caldas-Lopes E, Cerchietti L, Ahn JH, Clement CC, Robles AI, Rodina A,
    2011;19:684–92.                                                                    et al. Hsp90 inhibitor PU-H71, a multimodal inhibitor of malignancy,
12. Whitesell L, Lindquist SL. HSP90 and the chaperoning of cancer. Nat Rev            induces complete responses in triple-negative breast cancer models. Proc
    2005;5:761–72.                                                                     Natl Acad Sci U S A 2009;106:8368–73.
13. Fewell SW, Smith CM, Lyon MA, Dumitrescu TP, Wipf P, Day BW, et al.            30. Scaffidi C, Fulda S, Srinivasan A, Friesen C, Li F, Tomaselli KJ, et al. Two
    Small molecule modulators of endogenous and co-chaperone-stimulated                CD95 (APO-1/Fas) signaling pathways. EMBO J 1998;17:1675–87.
    Hsp70 ATPase activity. J Biol Chem 2004;279:51131–40.                          31. Georgakis GV, Li Y, Rassidakis GZ, Martinez-Valdez H, Medeiros LJ,
14. Li X, Srinivasan SR, Connarn J, Ahmad A, Young ZT, Kabza AM, et al.                Younes A. Inhibition of heat shock protein 90 function by 17-allyla-
    Analogs of the allosteric heat shock protein 70 (Hsp70) inhibitor, MKT-            mino-17-demethoxy-geldanamycin in Hodgkin's lymphoma cells
    077, as anti-cancer agents. ACS Med Chem Lett 2013;4:1042–7.                       down-regulates Akt kinase, dephosphorylates extracellular signal-regu-
15. Assimon VA, Gillies AT, Rauch JN, Gestwicki JE. Hsp70 protein complexes            lated kinase, and induces cell cycle arrest and cell death. Clin Cancer Res
    as drug targets. Curr Pharm Des 2013;19:404–17.                                    2006;12:584–90.
16. Rousaki A, Miyata Y, Jinwal UK, Dickey CA, Gestwicki JE, Zuiderweg ER.         32. Agard NJ, Mahrus S, Trinidad JC, Lynn A, Burlingame AL, Wells JA. Global
    Allosteric drugs: the interaction of antitumor compound MKT-077 with               kinetic analysis of proteolysis via quantitative targeted proteomics. Proc
    human Hsp70 chaperones. J Mol Biol 2011;411:614–32.                                Natl Acad Sci U S A 2012;109:1913–8.
17. Wadhwa R, Sugihara T, Yoshida A, Nomura H, Reddel RR, Simpson R, et al.        33. Christofferson DE, Li Y, Yuan J. Control of life-or-death decisions by RIP1
    Selective toxicity of MKT-077 to cancer cells is mediated by its binding to        kinase. Annu Rev Physiol 2014;76:129–50.
    the hsp70 family protein mot-2 and reactivation of p53 function. Cancer        34. Degterev A, Hitomi J, Germscheid M, Ch'en IL, Korkina O, Teng X, et al.
    Res 2000;60:6818–21.                                                               Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat
18. Fontaine SN, Rauch JN, Nordhues BA, Assimon VA, Stothert AR, Jinwal UK,            Chem Biol 2008;4:313–21.
    et al. Isoform-selective genetic inhibition of constitutive cytosolic Hsp70    35. Feoktistova M, Geserick P, Kellert B, Dimitrova DP, Langlais C, Hupe M,
    activity promotes client tau degradation using an altered co-chaperone             et al. cIAPs block Ripoptosome formation, a RIP1/caspase-8 containing
    complement. J Biol Chem 2015;290:13115–27.                                         intracellular cell death complex differentially regulated by cFLIP isoforms.
19. Taguwa S, Maringer K, Li X, Bernal-Rubio D, Rauch JN, Gestwicki JE, et al.         Mol Cell 2011;43:449–63.
    Defining Hsp70 subnetworks in dengue virus replication reveals key              36. Wertz IE, Dixit VM. Signaling to NF-kappaB: regulation by ubiquitination.
    vulnerability in flavivirus infection. Cell 2015;163:1108–23.                       Cold Spring Harb Perspect Biol 2010;2:a003350.

www.aacrjournals.org                                                                                                       Mol Cancer Res; 16(1) January 2018            67

      Downloaded from mcr.aacrjournals.org on March 8, 2021. © 2018 American Association for Cancer Research.
Published OnlineFirst September 28, 2017; DOI: 10.1158/1541-7786.MCR-17-0408

    Srinivasan et al.

    37. Moquin DM, McQuade T, Chan FK. CYLD deubiquitinates RIP1 in the                45. Pratt WB, Morishima Y, Murphy M, Harrell M. Chaperoning of glucocor-
        TNFalpha-induced necrosome to facilitate kinase activation and pro-                ticoid receptors. Handb Exp Pharmacol 2006;172:111–38.
        grammed necrosis. PLoS One 2013;8:e76841.                                      46. Schulte TW, Neckers LM. The benzoquinone ansamycin 17-allylamino-
    38. Galban S, Duckett CS. XIAP as a ubiquitin ligase in cellular signaling. Cell       17-demethoxygeldanamycin binds to HSP90 and shares important
        Death Differ 2010;17:54–60.                                                        biologic activities with geldanamycin. Cancer Chemother Pharmacol
    39. Yang Y, Fang S, Jensen JP, Weissman AM, Ashwell JD. Ubiquitin protein              1998;42:273–9.
        ligase activity of IAPs and their degradation in proteasomes in response to    47. Bukau B, Weissman J, Horwich A. Molecular chaperones and protein
        apoptotic stimuli. Science 2000;288:874–7.                                         quality control. Cell 2006;125:443–51.
    40. Kirkegaard T, Roth AG, Petersen NH, Mahalka AK, Olsen OD, Moilanen I,          48. Lewis J, Devin A, Miller A, Lin Y, Rodriguez Y, Neckers L, et al. Disruption of
        et al. Hsp70 stabilizes lysosomes and reverts Niemann-Pick disease-                hsp90 function results in degradation of the death domain kinase, recep-
        associated lysosomal pathology. Nature 2010;463:549–53.                            tor-interacting protein (RIP), and blockage of tumor necrosis factor-
    41. Li J, McQuade T, Siemer AB, Napetschnig J, Moriwaki K, Hsiao YS, et al. The        induced nuclear factor-kappaB activation. J Biol Chem 2000;275:
        RIP1/RIP3 necrosome forms a functional amyloid signaling complex                   10519–26.
        required for programmed necrosis. Cell 2012;150:339–50.                        49. Vanden Berghe T, Kalai M, van Loo G, Declercq W, Vandenabeele P.
    42. Muchowski PJ, Wacker JL. Modulation of neurodegeneration by molecular              Disruption of HSP90 function reverts tumor necrosis factor-induced
        chaperones. Nat Rev Neurosci 2005;6:11–22.                                         necrosis to apoptosis. J Biol Chem 2003;278:5622–9.
    43. Sabnis AJ, Guerriero CJ, Olivas V, Sayana A, Shue J, Flanagan J, et al.        50. Powers MV, Jones K, Barillari C, Westwood I, van Montfort RL, Workman P.
        Combined chemical-genetic approach identifies cytosolic HSP70 depen-                Targeting HSP70: the second potentially druggable heat shock protein and
        dence in rhabdomyosarcoma. Proc Natl Acad Sci U S A 2016;113:9015–20.              molecular chaperone? Cell Cycle 2010;9:1542–50.
    44. Taipale M, Krykbaeva I, Koeva M, Kayatekin C, Westover KD, Karras GI,          51. Rodina A, Taldone T, Kang Y, Patel PD, Koren J 3rd, Yan P, et al. Affinity
        et al. Quantitative analysis of HSP90-client interactions reveals principles       purification probes of potential use to investigate the endogenous Hsp70
        of substrate recognition. Cell 2012;150:987–1001.                                  interactome in cancer. ACS Chem Biol 2014;9:1698–705.

68 Mol Cancer Res; 16(1) January 2018                                                                                                   Molecular Cancer Research

          Downloaded from mcr.aacrjournals.org on March 8, 2021. © 2018 American Association for Cancer Research.
Published OnlineFirst September 28, 2017; DOI: 10.1158/1541-7786.MCR-17-0408

Heat Shock Protein 70 (Hsp70) Suppresses RIP1-Dependent
Apoptotic and Necroptotic Cascades
Sharan R. Srinivasan, Laura C. Cesa, Xiaokai Li, et al.

Mol Cancer Res 2018;16:58-68. Published OnlineFirst September 28, 2017.

 Updated version         Access the most recent version of this article at:
                         doi:10.1158/1541-7786.MCR-17-0408

     Cited articles      This article cites 51 articles, 15 of which you can access for free at:
                         http://mcr.aacrjournals.org/content/16/1/58.full#ref-list-1

    Citing articles      This article has been cited by 7 HighWire-hosted articles. Access the articles at:
                         http://mcr.aacrjournals.org/content/16/1/58.full#related-urls

      E-mail alerts      Sign up to receive free email-alerts related to this article or journal.

     Reprints and        To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at
    Subscriptions        pubs@aacr.org.

      Permissions        To request permission to re-use all or part of this article, use this link
                         http://mcr.aacrjournals.org/content/16/1/58.
                         Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)
                         Rightslink site.

                Downloaded from mcr.aacrjournals.org on March 8, 2021. © 2018 American Association for Cancer Research.
You can also read