34
1 Inhibition of Melanoma Growth by Small Molecules that Promote the Mitochondrial Localization of ATF2 Tal Varsano 1* , Eric Lau 1* , Yongmei Feng 1 , Marine Garrido 1 , Loribelle Milan 2 , Susanne Heynen-Genel 2 , Christian A. Hassig 2 , and Ze’ev A. Ronai 1 1 Signal Transduction Program and 2 Conrad Prebys Center for Chemical Genomics, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA *Equal contribution Running head: Small Molecules Promoting ATF2 Mitochondrial Localization Key words: ATF2, PKCε, melanoma, nuclear translocation, high content screen Abbreviations used: ATF2, activating transcription factor 2; PKCε, protein kinase C epsilon; FACS, fluorescence-activated cell sorting; DMSO, dimethyl sulfoxide Financial Support: Support by NIH grant CA099961 (to ZR) and a Melanoma Research Alliance grant (to ZR). EL supported in part by NIH T32 grant CA121949. Correspondence: Ze’ev A. Ronai, Signal Transduction Program, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd, La Jolla CA, 92037 USA, [email protected] Conflict of interest: There are no conflicts to disclose. Word count: 4641 excluding legends & abstracts Total number of figures and tables: 6 primary Figures, 6 Supplementary Figures, 2 Supplementary Tables Supplementary Methods, Legends for Supplementary Figures and Tables. Research. on June 25, 2021. © 2013 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

Inhibition of Melanoma Growth by Small Molecules that Promote the Mitochondrial ... · 2013. 4. 13. · 1 Inhibition of Melanoma Growth by Small Molecules that Promote the Mitochondrial

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

  • 1

    Inhibition of Melanoma Growth by Small Molecules that Promote the

    Mitochondrial Localization of ATF2

    Tal Varsano1*, Eric Lau1*, Yongmei Feng1, Marine Garrido1, Loribelle Milan2, Susanne Heynen-Genel2,

    Christian A. Hassig2, and Ze’ev A. Ronai1

    1Signal Transduction Program and 2Conrad Prebys Center for Chemical Genomics,

    Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA

    *Equal contribution

    Running head: Small Molecules Promoting ATF2 Mitochondrial Localization

    Key words: ATF2, PKCε, melanoma, nuclear translocation, high content screen

    Abbreviations used: ATF2, activating transcription factor 2; PKCε, protein kinase C epsilon; FACS, fluorescence-activated cell sorting; DMSO, dimethyl sulfoxide

    Financial Support: Support by NIH grant CA099961 (to ZR) and a Melanoma Research Alliance grant (to ZR). EL supported in part by NIH T32 grant CA121949.

    Correspondence: Ze’ev A. Ronai, Signal Transduction Program, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Rd, La Jolla CA, 92037 USA, [email protected]

    Conflict of interest: There are no conflicts to disclose.

    Word count: 4641 excluding legends & abstracts

    Total number of figures and tables: 6 primary Figures, 6 Supplementary Figures, 2 Supplementary Tables Supplementary Methods, Legends for Supplementary Figures and Tables.

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 2

    Translational Relevance

    The identification of new therapeutic modalities is among the most important priorities for improving

    cancer treatment. Among those is the need to halt oncogene addiction, which underlies tumor

    development, progression, and the development of resistance. We have recently demonstrated the

    oncogenic addiction of melanoma to ATF2 through PKCε, which mediates its confined nuclear

    localization. Here, we have developed a platform to identify small molecules that impair the oncogenic

    function of ATF2 by permitting its translocation from the nucleus to the mitochondria, thereby enabling

    its tumor suppressor activities to facilitate melanoma cell death. Our initial screen identified and

    characterized 2 compounds that sensitize melanoma cells to death, offering a proof-of-concept for the

    therapeutic paradigm whereby changing the subcellular localization of a protein can limit its oncogenic

    contributions and promote its tumor suppressor activities.

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 3

    ABSTRACT

    Purpose: Effective therapy for malignant melanoma, the leading cause of death from skin cancer,

    remains an area of significant unmet need in oncology. The elevated expression of PKCε in advanced

    metastatic melanoma results in the increased phosphorylation of the transcription factor ATF2 on

    threonine 52, which causes its nuclear localization and confers its oncogenic activities. The nuclear-to-

    mitochondrial translocation of ATF2 following genotoxic stress promotes apoptosis, a function that is

    largely lost in melanoma cells, due to its confined nuclear localization. Therefore, promoting the nuclear

    export of ATF2, which sensitizes melanoma cells to apoptosis, represents a novel therapeutic modality.

    Experimental Design: We conducted a pilot high-throughput screen of 3,800 compounds to identify

    small molecules that promote melanoma cell death by inducing the cytoplasmic localization of ATF2.

    The imaging-based ATF2 translocation assay was performed using UACC903 melanoma cells that stably

    express doxycycline-inducible GFP-ATF2.

    Results: We identified 2 compounds (SBI-0089410 and SBI-0087702) that promoted the cytoplasmic

    localization of ATF2, reduced cell viability, inhibited colony formation, cell motility, anchorage-free

    growth, and increased mitochondrial membrane permeability. SBI-0089410 inhibited the TPA-induced

    membrane tranlocation of PKC isoforms, whereas both compounds decreased ATF2 phosphorylation by

    PKCε and ATF2 transcriptional activity. Overexpression of either constitutively active PKCε or

    phosphomimic mutant ATF2T52E attenuated the cellular effects of the compounds.

    Conclusion: The imaging-based high-throughput screen provides a proof-of-concept for the

    identification of small molecules that block the oncogenic addiction to PKCε signaling by promoting

    ATF2 nuclear export, resulting in mitochondrial membrane leakage and melanoma cell death.

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 4

    INTRODUCTION

    The incidence of melanoma, the most deadly form of skin cancer, has steadily increased for the past

    30 years. Our current understanding of the mechanisms underlying the initiation and progression of

    melanoma has led to the development of specific inhibitors targeting the major signaling pathways that

    are known to be deregulated in melanoma, including B-Raf, PI3K, and MEK (1-3). Although clinical

    trials of B-Raf and MEK inhibitors in melanoma have produced promising results, the development of

    resistance to such monotherapies represents a barrier that has yet to be overcome (4, 5).

    ATF2, a member of the basic helix-loop-helix (HLH) family of transcription factors, is activated via

    phosphorylation by c-Jun N-terminal kinase or p38 in response to stimuli including stress and cytokines

    (6-8). ATF2 dimerizes with other members of the AP1 superfamily to activate the transcription of genes

    implicated in stress and DNA damage responses, growth, differentiation, and apoptosis (6-8). The genetic

    inactivation of ATF2 in melanocytes has been shown to abolish melanoma formation in the mutant N-

    Ras/Ink4a–/– genetic mouse model (9), indicating an oncogenic role for ATF2 in melanocyte

    transformation. Conversely, a tumor suppressor function for ATF2 was suggested by the increased

    incidence of papillomas (10) and mammary tumors (11) following the genetic inactivation of ATF2 in

    keratinocytes or mammary tissue, respectively. In our effort to understand the mechanisms underlying the

    opposing activities of ATF2, we discovered that the subcellular localization dictates the oncogenic or

    tumor suppressor function of ATF2. Whereas its nuclear localization is required for oncogenic activity,

    ATF2 must be localized to the cytoplasm to perform its tumor suppressor function. Analysis of tissue

    microarrays (TMAs) revealed that ATF2 exhibits cytosolic localization in basal cell carcinomas (BCC) or

    squamous cell carcinomas (SCC) (10) but is primarily nuclear in melanoma tumors, consistent with the

    constitutive transcriptional activity of ATF2 in these tumors (12). Notably, the nuclear localization of

    ATF2 is associated with poor prognosis in melanoma patients, suggesting that ATF2 localization might

    serve as a prognostic marker (12, 13).

    We recently found that the nuclear localization of ATF2 is dictated by its phosphorylation on

    threonine 52 (Thr52) by PKCε (14). Loss of Thr52 phosphorylation, as seen in several non-transformed

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 5

    or non-malignant cell lines following exposure to genotoxic stress, is required to enable the nuclear export

    and translocation of ATF2 to mitochondria, where it reduces mitochondrial membrane potential and

    promotes apoptosis. Elevated levels of PKCε, found in the more advanced metastatic melanomas, prevent

    the nuclear-to-mitochondrial translocation of ATF2 that enable its tumor suppressor function. Notably,

    the expression of peptides derived from ATF2 (amino acids 50–60 or 50–100) prevents the nuclear

    localization of ATF2 and sensitizes melanoma cells, but not melanocytes, to apoptosis (15-18). These

    effects were abolished by the mutation of the peptide at the PKCε phosphorylation site (Thr52) (15),

    suggesting that the native peptide functions by competitively inhibiting PKCε association

    with/phosphorylation of endogenous ATF2. Taken together, these findings suggest that small molecule

    modulators of ATF2 localization could attenuate its oncogenic addiction to PKCε signaling, thereby

    enhancing its pro-apoptotic functions. Because the nuclear-to-cytoplasmic export of ATF2 also sensitizes

    mutant B-Raf-expressing melanoma cells to apoptosis, agents that promote the nuclear export of ATF2

    are expected to represent a new therapeutic modality for drug-resistant melanomas.

    MATERIALS AND METHODS Cell lines and culture conditions

    HEK293T and NIH3T3 cells were obtained from American Type Culture Collection (ATCC,

    Manassas, VA). Melanoma cell lines were kindly provided by Dr. Meenhard Herlyn (Wistar Institute).

    The melanoma cell lines UACC903 and 501Melwere kindly provided by Drs. Gavin Robertson (Penn

    State University) and Ruth Halaban (Yale University), respectively. The cells were maintained at 37°C in

    a humidified 5% CO2 atmosphere and cultured in DMEM supplemented with 10% fetal bovine serum

    (FBS), 30 U/ml penicillin, 30 μg/ml streptomycin, and 2 mM L-glutamine (Gibco-Life Technologies,

    Grand Island, NY). The human melanocytes (Hermes 3A) were maintained in 254 medium supplemented

    with 10% FBS and human melanocyte growth supplement (Gibco-Life Technologies). The Lenti-X Tet-

    Off Advanced lentiviral inducible expression system (Clontech Laboratories, Mountain View, CA) was

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 6

    used to generate a stable UACC903 melanoma cell line that could induce the expression of GFP-ATF2.

    For this purpose, we used the Lenti-X Tet-Off Advanced lentiviral inducible expression system which

    requires the following 2 lentiviral constructs for tetracycline-controlled expression of ATF2: pLVX-Tet-

    Off Advanced (which is under G418 selection) and pLVX-Tight into which GFP-ATF2 was cloned (and

    which is under puromycin selection). The UACC903 melanoma cells were co-transduced with the 2

    lentiviruses and selected by growth in G418- and puromycin-containing medium. The expression of GFP-

    ATF2 was repressed by the addition of the tetracycline analog doxycyline to the growth medium. The

    transfer of the cells into doxycycline-free medium then enabled the controlled expression of GFP-ATF2.

    Because melanocytes and melanoma cells are inherently resistant to G418 (19), we used FACS to further

    enrich for GFP-ATF2-positive cells after withdrawal from doxycycline. The UACC903 GFP-ATF2 Tet-

    Off (hereafter referred to as GFP-ATF2Tet-Off UACC903) cell line was maintained in DMEM

    supplemented with 10% FBS, 500 ng/ml doxycycline and 5 µg/ml puromycin, plus 1 mg/ml G418 to

    maintain the selection of stable lines.

    Reagents

    A 3,800 compound subset of the ChemBridge chemical library (ChemBridge, San Diego, CA) was

    provided by the Conrad Prebys Center for Chemical Genomics at our Institute. Dimethyl sulfoxide

    (DMSO), formaldehyde solution, crystal violet, Triton X-100 (TX-100), bovine serum albumin (BSA),

    and sucrose were purchased from Sigma-Aldrich; the PKC inhibitor Gö6850 was purchased from

    Calbiochem (Billerica, MA). MitoTracker Deep-Red FM was purchased from Invitrogen (Carlsbad, CA).

    Protease and phosphatase inhibitors cocktails (PhosSTOP and cOmplete, respectively) were purchased

    from Roche (Indianapolis, IN). The antibodies employed were purchased as follows: ATF2 (#20F1),

    Stat3 (#9132), Cox IV (3E11), pATF2-T69/71 (#9225), pan-pPKC T514 (#9379), P38 (#9216), pP38

    (#9212), pErk1/2 (#9106), Erk1/2 (#9102), PKC isoform sampler kit (#9960), pAkt (#9271), and Akt

    (#9272) from Cell Signaling Technologies (Danvers, MA); PKCε (C15), ATF2 (N96 & C19), β-tubulin

    (G8), p53 (FL393), pan-PKC (H300), and GFP (B2) from Santa Cruz Biotechnology, Inc. (Santa Cruz,

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 7

    CA); pS729 PKCε (44977G) from Invitrogen; HSP60 (24) and β-catenin (14) from BD Pharmingen, (San

    Diego, CA); Actin (ACTN05) from Thermo-Fisher (Pittsburgh, PA); pT52 ATF2 antibodies were

    produced by PhosphoSolutions Inc. (Aurora, CO). Secondary antibodies were goat anti-rabbit Alexa-680

    F(ab’)2, goat anti-rabbit Alexa-568 F(ab')2, goat anti-mouse Alexa-488 F(ab')2 (all Molecular Probes,

    Eugene, OR), and goat anti-mouse IRDye 800 F(ab’)2 (Rockland Immunochemicals, Gilbertsville, PA).

    Immunoblotting

    Cell lysates were prepared by lysis in RIPA buffer (150 mM NaCl, 1 mM EDTA, 1% NP40, 0.25%

    sodium deoxycholate, 0.1% SDS, 50 mM Tris HCl pH 7.4, freshly supplemented with phosphatase and

    protease inhibitors). Lysates were clarified by centrifugation, and proteins were separated by SDS-PAGE

    and transferred to PVDF membranes (Millipore, Billerica, MA). After blocking with PBS containing 5%

    non-fat milk, the membranes were incubated with primary antibodies (pT52-ATF2 (1:500); ATF2

    (#20F1; 1:1,000)) in PBS containing 5% BSA and 0.1% Tween-20, at room temperature for 1 h or at 4ºC

    overnight. The membranes were washed and incubated for 1 h at room temperature with goat anti-rabbit

    Alexa Fluor 680 F(ab’)2 or goat anti-mouse IRDye 800 F(ab’)2 diluted at 1:10,000. The blots were

    subjected to infrared imaging with 2-color detection using an Odyssey Infrared Imaging System (LiCor

    Biosciences, Lincoln, NE), and quantification of bands was performed according to the manufacturer’s

    protocols.

    Transfections

    Plasmid DNAs were transfected using JetPrime reagent (Polypus Transfection Inc., New York, NY)

    according to the manufacturer’s protocol.

    Immunofluorescence

    Cells were cultured on glass coverslips, subjected to the indicated treatments, and fixed with 4%

    formaldehyde in 100 mM phosphate buffer, pH 7.4, for 30 min. After fixation, cells were permeabilized

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 8

    with PBS containing 0.4% Triton X-100 (TX-100) and 0.4% Bovine Serum Albumin (BSA) for 20 min,

    and incubated for 1-18 h with primary antibodies in staining buffer (0.1% TX-100/0.1% BSA in PBS).

    Cells were washed 4-5 times with washing buffer (0.2% TX-100/0.2% BSA in PBS), followed by a 3 h

    incubation with goat anti-rabbit Alexa-568 and/or anti-mouse Alexa-488 F(ab')2 in staining buffer,

    followed by two additional washes in washing buffer. The coverslips were then mounted with Vectashield

    containing DAPI (Vector Laboratories, CA). Fluorescence images were acquired using either an Olympus

    IX-71 inverted microscope equipped with a digital ORCA-ER camera (Hamamatsu) or with an Olympus

    FluoView 1000 confocal microscope equipped with a CH350 CCD camera (Hamamatsu). All images

    were processed with Adobe Photoshop CS5 (Adobe Systems, Mountain View, CA). Fluorescence images

    were evaluated and quantified using ImageJ software (NIH, Bethesda, MD).

    Additional Methods can be found online at CCR web page.

    RESULTS

    ATF2 Translocation Assay

    We developed a cell-based imaging assay to monitor the subcellular localization of ATF2 by

    expressing GFP-ATF2 in melanoma cells. For this purpose, UACC903 cells were co-transduced with a

    tetracycline regulator lentivirus (pLVX Tet-Off Advanced) and a response lentivirus (pLVX-Tight puro)

    into which the GFP-ATF2 insert (GFP-ATF2Tet-Off UACC903 cells; see Methods and Materials) was

    cloned. We confirmed using fluorescence microscopy that GFP-ATF2 expression was prevented in

    cultures maintained in the presence of 500 ng/ml doxycycline but was induced and readily detectable in

    the nuclei within 36-48 h of cell transfer into doxycycline-free medium (Fig. 1A). We also monitored the

    expression of GFP-ATF2 by immunoblotting, because excessive GFP-ATF2 expression might interfere

    with the magnitude or kinetics of ATF2 export from the nuclei. As shown in Figure 1, 24-48 h after

    withdrawal of doxycycline, GFP-ATF2 was expressed at levels comparable to (within 1- to 5-fold as

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 9

    determined by immunoblotting analysis) the endogenous ATF2 levels (Fig. 1B). Taken together, these

    results demonstrate that in GFP-ATF2Tet-Off UACC903 melanoma cells, GFP-ATF2 was expressed at

    levels comparable to endogenous ATF2 and was similarly localized within the nucleus.

    Identification of Hit Compounds in a High-Throughput Screen

    We performed a pilot study to determine whether the GFP-ATF2Tet-Off UACC903 system was

    amenable for the high-throughput screening of small molecules that promote ATF2 nuclear export. For

    this purpose, we selected approximately 3,800 compounds from the ChemBridge compound library,

    containing representatives of different scaffolds and structures. The cells were plated in twelve 384-well

    plates. Each plate included 32 wells treated with vehicle (0.2% DMSO) as negative controls and 32 wells

    treated with the PKC inhibitor Gö6850 (5 µM) as a positive control, whereas the remaining wells were

    treated with the library compounds (single compound, 10 µM per well). To determine if GFP-ATF2

    localization was affected by the compounds, we measured the ratio between the mean nuclear intensity

    and the mean cytoplasmic intensity of GFP fluorescence (Fig. 1C). Whereas in DMSO-treated cells, GFP-

    ATF2 was mainly localized to the nucleus, in Gö6850-treated cells, GFP-ATF2 was found both in the

    nucleus and in the cytosol (Fig. 2A, B). The average Z’-value (21) for this screen based on the positive

    and negative control wells was 0.47 and ranged between 0.4 and 0.56 between plates. As noted in the

    Materials and Methods section, the assay was designed to minimize influence of the autofluorescence of

    the positive control compound Gö6850. It is, however, possible that residual Gö6850 compound

    fluorescence might have artificially increased the reported Z’-value beyond the true assay window. As

    expected, most compounds were inactive and GFP-ATF2 localization was mostly nuclear, similar to that

    observed in the DMSO-treated cells. The automated analysis initially identified 20 wells with activity >

    50%, but further examination revealed that 14 of the 20 hits were due to autofluorescence or other

    artifacts, leaving six potential hits (0.16% hit rate). Two of the 6 compounds failed the PAINS

    cheminformatics filter (for known Promiscuous and Assay-Interfering Nuisance compounds). The

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 10

    remaining 4 compounds were selected for further analysis using independently ordered powders, which

    were confirmed in a set of dose-dependent assays (Fig. 2C and data not shown).

    Validation of Hit Compounds with Endogenous ATF2

    To confirm that the 4 hits identified in the GFP-ATF2Tet-Off UACC903 pilot screen are able to

    promote the cytoplasmic localization of endogenous ATF2, we ordered new batches of the compounds

    and tested their effects on endogenous ATF2 translocation in WM793 melanoma cell lines by

    immunofluorescence microscopy (Fig. 3A). The cells were incubated in the presence of the compounds

    for either 6 or 24 h. The images were analyzed and quantified using ImageJ software, which confirmed

    that treatment with hit 3 (SBI-0089410; N-[2-(1-adamantyl)ethyl]-4-

    [(dimethylamino)sulfonyl]benzamide) or hit 4 (SBI-0087702; N-[(4-methoxy-1-naphthyl)methyl]-2-(4-

    methoxyphenyl)ethanamine), but not the other 2 hits (hits 1 and 2), promoted cytoplasmic/mitochondrial

    localization of endogenous ATF2 (Fig. 3B). Partial colocalization between GFP-ATF2 and mitochondria

    (using HSP60 as surrogate marker of mitochondrial localization) was observed (arrowheads Fig. 3A)

    following treatment with both hits, thereby confirming their ability to promote ATF2 mitochondrial

    localization. SBI-0089410 and SBI-0087702 represent low molecular weight compounds that do not

    appear to be promiscuous based on a SciFinder search of each structure. These compounds do not contain

    undesirable or reactive functional groups and thus represent possible starting points for a hit-to-lead

    effort, and also illustrate that synthetically tractable hits can be obtained from a screen. Notably, the

    nuclear-to-cytoplasmic ratio of endogenous ATF2 was reduced to the same degree seen for the GFP-

    ATF2 (approximately 30-45% in SBI-0089410- or SBI-0087702-treated cells compared to control,

    DMSO-treated cells (Fig. 3B). Similar results were also observed in UACC903 melanoma cells (Fig. 3B).

    Interestingly, the effect of SBI-0087702 on ATF2 localization was detected only after incubation for 24 h,

    whereas SBI-0089410 induced earlier translocation (after 6 h), which was not sustained at later time

    points, suggesting that each of these compounds affects ATF2 translocation via a distinct

    mechanism/target(s). Consistent with the immunostaining data, subcellular biochemical fractionation

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 11

    revealed increased ATF2 in the purified VDAC1-containing mitochondrial fractions, as well as in the β-

    tubulin-containing cytosolic fraction of WM793 cells after 24 h treatment with SBI-0087702. Similar

    increase of ATF2 in the mitochondrial fraction was observed in cells treated for 6 h with SBI-0089410

    (Supplementary Fig. S1A, B).

    Notably, the treatment of UACC903 cells with DMSO, SBI-0089410, SBI-0087702, or Gö6850 did

    not affect the nuclear compartmentalization of p53, (Supplementary Fig. S1C), a transcription factor that

    partially localizes at the mitochondria following oxidative stress (22). Furthermore, neither SBI-0089410

    nor SBI-0087702 affected the localization of Stat3 and β-catenin, under conditions that induced ATF2

    translocation (Supplementary Fig. S2), suggesting that these compounds do not have pleiotropic effect on

    other transcripiton factors that can be also localized at the mitochondria.

    SBI-0089410 and SBI-0087702 Induce Apoptosis and Inhibit Melanoma Cell Growth in an ATF2-

    Dependent Manner

    To determine whether SBI-0089410 or SBI-0087702 affected melanoma cell growth, we measured

    the colony formation of 501Mel cells. Notably, although both compounds reduced colony formation, SBI-

    0087702 exhibited the greatest effect, reducing the number of colonies by ~90% (Fig. 4A). The inhibition

    of colony formation is likely an outcome of the cytotoxicity of these compounds. To verify that the cell

    death observed following treatment with SBI-0089410 and SBI-0087702 was mediated by ATF2

    translocation, we examined whether overexpression of a constitutively nuclear ATF2 mutant (ATFT52E)

    could rescue cell death. Indeed, ATFT52E-stably transduced UACC903 cells exhibited reduced degrees (up

    to 50%) of cell death upon treatment by SBI-0089410 or SBI-0087702, compared with control empty

    vector (EV)-transduced cells (Fig. 4B and 4C). Furthermore, similar to their effect on UACC903 cells

    (which harbor mutated B-Raf and wild-type N-Ras), SBI-0089410 and SBI-0087702 induced apoptosis in

    WM1346 melanoma cells (which harbor wild-type B-Raf and mutated N-Ras), which was dependent on

    ATF2 (Fig. 4B, 4C and Supplementary Fig. S3A-C). Notably, the combined treatment of PLX4032-

    resistant melanoma cell lines (501Mel & UACC903) with low dose (2 µM) PLX4032 and 10 µM of

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 12

    either of the 2 compounds suppressed their viability and colony-forming ability compared to PLX4032

    alone (see data below). Importantly, the two compounds did not induce apoptosis in melanocytes, even

    when used at a higher concentration (20 μM; Supplementary Fig. S3D), although in human foreskin (BJ)

    fibroblasts, they induced moderate levels of apoptosis (Supplementary Fig. S3D) relative to the melanoma

    cell lines. Treatment with SBI-0089410 (6 h) resulted in an increased G2/M phase population, whereas

    SBI-0087702 (24 h) reduced the G2/M population and increased sub-G1 (dead cell) content

    (Supplementary Fig. S3E), indicating that both compounds affect melanoma cells by distinct, albeit

    ATF2-dependent, mechanisms. Interestingly, the ability of ATF2T52E to attenuate the effect of SBI-

    0087702 was also accompanied by changes in cell morphology; ATF2T52E-expressing cells appeared more

    rounded and compact than EV-expressing cells (Supplementary Fig. S4A).

    We next examined the effect of these two compounds on migration, a major phenotype of melanoma

    cells. Both SBI-0089410 and SBI-0087702 inhibited the motility of WM1346 cells across a porous

    membrane in a Boyden chamber-type assay. Notably, the expression of ATF2T52E in the WM1346 cells

    effectively reversed the inhibitory effect of SBI-0089410 and to lesser extent the effect of SBI-0087702

    (Fig. 4D and Supplementary Fig. S4A-C). These results demonstrate that both compounds promote the

    apoptosis of melanoma cells and inhibit their migration in an ATF2-dependent manner.

    SBI-0089410 and SBI-0087702 Perturb Mitochondrial Membrane Integrity and Promote Leakage

    We previously showed that cytoplasmic ATF2 promotes cell death by binding to protein complexes at

    the mitochondrial membrane, resulting in reduced mitochondrial membrane potential, increased

    mitochondrial leakage, and subsequent apoptosis (14). Consistent with its effects on ATF2 localization,

    we found that SBI-0087702 compromised mitochondrial membrane potential and promoted

    mitochondrial leakage in WM793 and UACC903 cells (Fig. 5A, B and Supplementary Fig. S5,

    respectively). SBI-0089410 induced a transient decrease in mitochondrial membrane potential at 6 h,

    which is consistent with its transient effect on ATF2 cytoplasmic localization.

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 13

    To verify that the decrease in mitochondrial membrane potential observed following treatment with

    SBI-0089410 and SBI-0087702 was mediated by ATF2 translocation, we examined whether the

    overexpression of ATFT52E (the constitutively nuclear ATF2 mutant) would prevent mitochondrial leakage

    (Fig. 5C). Indeed, the ectopic expression of ATF2T52E effectively attenuated the loss of mitochondrial

    membrane potential induced by both SBI-0089410 and SBI-0087702, but not by the mitochondrial

    uncoupler CCCP, which promotes mitochondrial leakage in an ATF2-independent fashion.

    To determine the potential of SBI-0089410 and SBI-0087702 to inhibit tumor growth, we assessed

    their ability to inhibit 3D, anchorage-free growth using the spheroid growth assay (Fig. 4E, F). We found

    that after 8 days, SBI-0089410 and SBI-0087702 inhibited spheroid growth by >70% and >90%,

    respectively. These results suggest that these compounds are stable enough to affect long-term growth and

    are able to penetrate into 3D structures, characteristics required for effective therapy.

    SBI-0089410 and SBI-0087702 Inhibit Melanoma Cell Growth and Thr52 phosphorylation of ATF2 in

    a PKCε-Dependent Manner

    In cell viability assays, treatment with 10 µM SBI-0087702 was sufficient to inhibit the growth of

    501Mel cells by >90% (Fig. 5D, left). Significantly, the ectopic expression of a constitutively active form

    of PKCε (PKCε-CA) prevented the cell death induced by SBI-0087702 treatment, suggesting that this

    compound mediates its effect on ATF2 translocation and cell viability via inhibition of PKCε (Fig. 5D,

    right). Because PKCε-mediated phosphorylation of ATF2 on T52 controls its subcellular localization, we

    next investigated whether SBI-0089410 and SBI-0087702 affect the phosphorylation of endogenous

    ATF2 at Thr52. Lysates from DMSO-, SBI-0089410-, or SBI-0087702-treated NIH3T3 cells were

    subjected to immunoblot analysis for altered phosphorylation on T52, as well as on T69/71, which are the

    sites for JNK/p38 phosphorylation. SBI-0089410, and to a lesser degree SBI-0087702, reduced the level

    of ATF2 phosphorylation on T52 (SBI-0089410 exhibited comparable effects to that of the PKC inhibitor

    Gö6850; Fig. 6A). Interestingly, both Gö6850 and SBI-0087702 also attenuated the degree of ATF2

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 14

    phosphorylation on T69/71, indicative of the effect of these compounds on JNK/p38-mediated

    phosphorylation of ATF2. Unlike the broad PKC inhibitor Gö6850, which also reduced the total ATF2

    expression level, the selected hits were more selective and did not exhibit such effect (Fig. 6A). We

    further tested whether SBI-0089410 might elicit similar effects on ATF2 phosphorylation in UACC903

    cells (Fig. 6B). Indeed, SBI-0089410 inhibited ATF2 phosphorylation on Thr52 but to a lesser degree on

    Thr 69/71. Additionally, SBI-0089410 inhibited p38 phosphorylation. The inhibitory effect on

    phosphorylation was transient, peaking at 6 h after incubation with SBI-0089410. This transient effect on

    ATF2 phosphorylation is consistent with the effect on ATF2 translocation (Fig. 3) and on mitochondrial

    leakage (Fig. 5 and Supplementary Fig. S5). Notably, SBI-0089410 and SBI-0087702 did not inhibit

    other key signaling molecules including Erk1/2, Akt, pan-PKC (Supplementary Fig. S6A), suggesting

    that these compounds do not affect signal transduction pathways that are central in melanomagenesis.

    Since SBI-0089410 and SBI-0087702 affected ATF2 phosphorylation by PKCε but not the

    phosphorylation of PKC itself, we set to assess their possible effect on the recruitment of PKC to cellular

    membranes. Interestingly, SBI-0089410, but not SBI-0087702, inhibited the recruitment of PKCε and

    other PKC isoforms to the membrane within 3 min of stimulation with TPA (Fig. 6C, D and

    Supplementary Fig. S6B, C, D, and E). At 10 µM, SBI-0089410 inhibited PKC translocation, although

    this effect was stronger with increasing dose (10-50 µM) or at lower TPA concentration (Fig. 6C and

    S6E). Taken together, these results suggest that SBI-0089410 and SBI-0087702 inhibit ATF2

    phosphorylation on Thr52, thus preventing ATF2 nuclear translocation, and that the effect of SBI-

    0089410 is mediated by impairing PKC-to-membrane translocation.

    Mapping transcriptional programs that are affected by SBI-0087702

    To measure the effects of the 2 compounds on ATF2 transcriptional activities, we performed

    transcription-based reporter assays and a qPCR analysis of known ATF2 transcriptional target genes.

    Using an ATF2-targeted (Jun2) promoter-luciferase assay system, we evaluated how SBI-0089410 and

    SBI-0087702 affect the transcriptional activity of ATF2 (Fig. 6E). Consistent with the

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 15

    cytoplasmic/mitochondrial translocation effects of SBI-0089410 and SBI-0087702 on ATF2, these

    compounds reduced the transcriptional activity of ATF2 in UACC903 cells at 24 h by approximately 47%

    and 56%, respectively. Notably, the effect of the compound was more pronounced than a PKCε peptide

    inhibitor, which reduced ATF2 transcriptional activity by approximately 24%. Assessment of the Jun2-

    luciferase activity in another melanoma cell line, WM793, revealed that SBI-0089410 and SBI-0087702

    similarly reduced ATF2 transcriptional activity at 24 h by approximately 30% and 58%, respectively,

    whereas the PKCε peptide inhibitor reduced ATF2 transcriptional activity by approximately 37%

    (Supplementary Fig. S6F). Evaluation of the effects of these compounds on the activity of the c-Jun-

    targeted TRE promoter did not reveal inhibition, but rather slightly increased c-Jun activity

    (Supplementary Fig. S6G). To further define the specificity of these compounds, we examined the effects

    of SBI-0089410 and SBI-0087702 on the transcriptional activities of the heat shock response element

    (HSRE) and androgen receptor (AR), which are representative of independent transcriptional regulatory

    elements. As shown Supplementary Figure S6H and I, neither of the 2 compounds caused notable

    changes in the transcriptional activity from these unrelated promoters.

    In comparing the effect of the 2 compounds on ATF2 translocation, phosphorylation and

    sensitization of melanoma cells to apoptosis, SBI-0087702 appeared to be superior. Therefore, we further

    characterized the transcriptional changes elicited by this compound. To this end, we subjected WM793

    cells treated with either DMSO or 10 µM SBI-0087702 for 24 h to microarray-based expression analysis

    (see details in the Materials and Methods section; data deposited in GEO accession # GSE43135). The

    top 3 functional networks upregulated in response to SBI-0087702 (Fig. 6F) were as follows: (A)

    lipid/small molecule/vitamin and mineral metabolism, (B) cellular development, and (C)

    carbohydrate/small molecule/lipid metabolism. Notably, 2 of the 3 major networks that were upregulated

    are associated with lipid metabolism, consistent with the increase seen in mitochondrial mass

    (Supplementary Fig. 5B). The top 3 downregulated clusters were: (A) tumor morphology/cell growth and

    proliferation/development, (B) cell morphology, and (C) cellular movement. Here, all of the 3 major

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 16

    networks affected by SBI-0087702 are associated with cell morphology and growth, consistent with the

    effects elicited by this compound on the migratory behavior of the melanoma cells (Fig. 4 and

    Supplementary Fig. S4). The corresponding top 20 up- and downregulated genes are shown in

    Supplementary Tables S1 and S2.

    DISCUSSION

    Oncogene addiction is the phenomenon whereby the survival of cancer cells depends on constitutive

    oncogenic cues, often mediated by mutated, or otherwise activated oncogenes (23). Our recent studies

    have pointed to the oncogenic addiction of melanoma to ATF2, which is controlled by PKCε (14).

    Constitutively active PKCε-mediated phosphorylation of ATF2 impairs its nuclear export following

    cellular stress and damage, which under normal circumstances would promote the association of ATF2

    with the mitochondrial membrane proteins voltage-dependent anion channel 1 and hexokinase 1 (VDAC1

    and HK1), resulting in the loss of mitochondrial membrane integrity and the induction of apoptosis (14).

    In melanoma, ATF2 is largely confined to the nucleus, predominantly due to its constitutive

    phosphorylation by PKCε, which confers its oncogenic addiction and attenuates its role in genotoxic

    stress-induced cell death (14). We therefore sought to establish an imaging-based high-throughput screen

    to identify small molecules capable of inducing the nuclear export of ATF2 in melanoma cells. We report

    here on the establishment and the results of such proof-of-concept screen. The GFP-ATF2Tet-Off UACC903

    cell line was used to perform the initial screen of 3,800 compounds, from which we identified 2 small

    molecules that promote cytoplasmic/mitochondrial localization of ATF2 in melanoma cells. The

    compounds were confirmed to reduce mitochondrial membrane potential and to have concomitant effects

    on melanoma growth and colony formation. Importantly, the effects of the compounds could be partially

    blocked by the overexpression of a constitutively active form of PKCε or a phosphomimic ATF2 mutant

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 17

    that is constitutively active transcriptionally. These observations strongly suggest that the compounds

    mediate their inhibitory effects through PKCε and ATF2.

    Interestingly, SBI-0089410 reduced ATF2 phosphorylation on Thr52 by a similar degree as the PKCε

    inhibitor Gö6850, suggesting that this compound directly affects PKCε or its phosphorylation of ATF2.

    SBI-0087702 was effective in inhibiting ATF2 Thr52 phosphorylation, albeit to lesser degree compared

    with SBI-0089410, suggesting that its mechanism of action might be at least partially independent of the

    PKCε-ATF2 axis. Consistent with this possibility, the duration of the effects of these 2 compounds were

    different. SBI-0087702 elicited sustained ATF2 translocation, whereas the effect of SBI-0089410 was

    more rapid but transient, perhaps due to its reduced stability or alternate cellular target. Likely, SBI-

    0089410, and SBI-087702 elicit their effect on ATF2 and melanoma cells through different pathways.

    Consistent with the gene expression profiling, SBI-087702 reduces the expression of genes associated

    with growth and proliferation while increasing mitochondrial lipid biosynthesis, corresponding to the

    inhibition of melanoma growth and tumorigenicity seen in cultured cells. Arguably, each of the

    compounds identified in this screen exhibit a partial profile seen for Gö6850, pointing to a more selective

    effect. Among the 2 selected compounds that were characterized here, SBI-0087702 exhibits the more

    desired profile with respect to ATF2 translocation, phosphorylation and melanoma sensitization to

    apopotosis.

    Although we do not yet understand the mechanism by which the small molecules might induce ATF2

    nuclear export and function at the mitochondria, several possibilities exist. The compounds might inhibit

    PKCε activity (as appears to be the case for SBI-0089410), or they might activate a protein phosphatase,

    which would increase T52 dephosphorylation, thereby promoting ATF2 nuclear export. Alternatively, the

    compounds might modify ATF2 to facilitate its interaction with nuclear export factors. We cannot

    exclude the possibility that the compounds might elicit more indirect and global effects; for example, by

    influencing the function of import/export proteins in general. However, we note that the hit validation

    testing funnel was designed to exclude hits that affect ATF2 localization indirectly (by monitoring the

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 18

    subcellular partitioning of other transcription factors and by the ability to attenuate ATF2 translocation

    upon expression of ATF2T52E).

    Although we validated our observations on GFP-ATF2 by examining endogenous ATF2, it is

    possible that GFP-tagged ATF2 might not completely phenocopy the dynamics or the action of

    endogenous ATF2 co-expressed in the same cells. This issue could be circumvented by inhibiting

    endogenous ATF2 expression and by titrating doxycycline to limit the expression of exogenous GFP-

    ATF2. In this study, however, our secondary analysis confirmed that the hits indeed affected ATF2

    translocation and sensitized the melanoma cells to cell death, which was the ultimate purpose of the

    screen.

    Can small molecules that promote ATF2 nuclear export offer a novel therapeutic modality? Evidence

    from our previous work suggests an affirmative answer to this important question. We showed that

    ATF2-derived peptides that affected ATF2 subcellular localization and transcriptional activity also

    effectively sensitized the melanoma cells to death induced by chemotherapeutic drugs, and inhibited

    melanoma development and metastasis in human and mouse models (9, 15, 17). Supporting this notion,

    the treatment of melanoma cells that are resistant to PLX4032 with either SBI-0089410 or SBI-0087702

    and low-dose PLX4032 reduced their viability and colony-forming ability relative to PLX4032 alone

    (Supplementary Fig. S6J and S6K). However, given our current understanding of the melanoma

    therapeutic landscape, one might consider this as a complementary approach to existing therapies that

    target different components along the MAPK signaling pathways. Genetic evidence supports the

    importance of ATF2 in N-Ras melanoma development (9), pointing to the possibility that small molecules

    identified in these screens could be used to treat N-Ras melanomas, for which there is currently no

    effective therapy.

    Overall, this study provides proof-of-concept for the high-content screening of small molecules to

    promote the nuclear export of ATF2 in PKCε-addicted melanomas. This approach can be readily adapted

    to evaluate other transcription factors that elicit opposing functions dependent on their subcellular

    localization.

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 19

    ACKNOWLEDGMENTS

    We thank Drs. Meenhard Herlyn, Ruth Halaban, and Gavin Robertson for the melanoma cell lines used in

    this study. We also thank Dr. Edward Mosonov and the Cell-Imaging Facility for help with confocal

    microscopy, and Amy Cortez and the Flow Cytometry Facility for help with cell sorting.

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 20

    FIGURE LEGENDS

    Figure 1. The induction of GFP-ATF2 expression in the inducible GFP-ATF2Tet-Off UACC903 stable

    cell line and its application in an imaging-based HTP screen for inducers of ATF2 translocation.

    (A) GFP-ATF2 is expressed and readily detectable in GFP-ATF2Tet-Off UACC903 cells following removal

    of doxycycline from the medium. GFP-ATF2 expression was determined using direct fluorescence

    microscopy with an Olympus IX71 platform fitted with a 10x air objective lens (NA = 0.31). Scale bar =

    20 µm. Insets are further magnified areas of each image. (B) Levels of induced GFP-ATF2 and

    endogenous ATF2 are comparable (1- to 5-fold) in the GFP-ATF2Tet-Off UACC903 stable cell line.

    Expression of GFP-ATF2 and endogenous ATF2 was compared in GFP-ATF2Tet-Off UACC903 cells and

    the parental cell line (negative control). GFP-ATF2Tet-Off UACC903 cells were plated and cultured in the

    presence or absence of 500 ng/ml doxycycline to inhibit or promote expression of GFP-ATF2,

    respectively. Cells were cultured for 12-72 h and lysed in RIPA buffer. Lysates were subjected to

    immunoblot analysis using rabbit anti-ATF2 and mouse anti-GFP antibodies. Actin and Ponceau staining

    were used for loading control. (C) Imaging-based assay read-out for ATF2 translocation. UACC903 cells

    stably expressing ATF2-GFP and that were stained with DAPI were imaged with the Opera QEHS system

    using DAPI and GFP wavelengths (top left and middle respectively, top right shows pseudo-colored

    overlay). (1) Nuclei detection from DAPI image via thresholding and object splitting (nuclei outlines in

    red). (2) Cell detection from contrast-stretched DAPI images via thresholding combined with nuclei

    location information (cell outlines in green). (3) The creation of cytoplasmic regions by subtraction of

    nuclei regions from the cell regions (cytoplasm outlines in yellow). (4) The overlay of nuclei (red) and

    cytoplasm (yellow) regions on ATF2 image. (5) Quantify nuclear metrics from nuclei regions of DAPI

    images and ATF2-based nuclei and cytoplasmic region metrics from ATF2-GFP images on a cell-by-cell

    level. (6) Create GFP-positive cell sub-population by applying a minimum intensity threshold for whole

    cell GFP intensity. (7) Calculate well-level assay plate read-outs from cell sub-population statistics of

    each well. Image analysis was performed using Acapella/Columbus HCS image analysis software.

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 21

    Figure 2. A pilot screen of a 3,800 compound ChemBridge library to identify compounds that

    promote the nuclear export of GFP-ATF2. (A) Summary of the results of the complete HTS screen

    showing the mean ratio of nuclear and cytoplasmic GFP fluorescence intensity for triplicate wells of each

    compound. (B) Nuclear and cytoplasmic intensities were also evaluated separately for each hit to remove

    compounds that interfere with the fluorescent protein directly or spectrally as indicated by a significant

    increase (Z-Score > 5) or decrease (Z-Score < 5) in both regions’ intensities simultaneously. Some

    autofluorescent compounds are outside of the axes range displayed here. The assay was designed away

    from the autofluroescent wavelength (Gö6850 is strongly autofluorescent in the red channel) by using

    green secondary antibodies and emission filters that would minimize the autofluorescence of the Go

    compound. The average Z’ for this screen was 0.47 and ranged between 0.4 and 0.56 in different plates.

    Cells were imaged from twelve 384-well plates (4,357 valid total wells 3,669 valid compound wells)

    using the Opera acquisition system and analyzed with Acapella software to identify GFP-positive cells

    and delineate nuclear and cytoplasmic regions. DMSO was used as the negative control (blue circles) and

    the PKC inhibitor Gö6850, which promotes nuclear export of ATF2, was used as the positive control (red

    circles). As expected, most of the compounds were inactive and exhibited ratios similar to the negative

    controls. (C) The structure of the 4 selected hits.

    Figure 3. SBI-0089410 and SBI-0087702 promote the translocation of endogenous ATF2 to the

    cytoplasm/mitochondria. (A) Representative images of WM793 melanoma cells following treatment

    with DMSO, SBI-0089410, or SBI-0087702, showing increased localization of ATF2 to the cytoplasm

    and mitochondria (arrowheads). WM793 cells were cultured on glass coverslips and incubated with

    DMSO (0.2%, 24 h), SBI-0089410 (10 µM, 6 h), or SBI-0087702 (10 µM, 24 h). The cells were then

    fixed and stained with specific anti-ATF2 antibodies (green channel), anti-HSP60 antibodies to label

    mitochondria (red channel), and DAPI to label nuclei (blue channel). The cells were imaged using a

    FluoView1000 confocal microscope with an 100x oil immersion objective. Scale bar = 10 µm. Similar

    results were obtained with UACC903 cells (data not shown). (B) To measure ATF2 translocation, either

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 22

    the GFP fluorescence intensity of GFP-ATF2-expressing UACC903 (graph on left) or the ATF2

    immunofluorescence intensity in WM793 and UACC903 cells (graphs on right) in the nucleus and

    cytoplasm were quantified using ImageJ software. Between 6 and 17 cells were analyzed for each

    condition. Quantification reveals a reduction of approximately 40-50% in the nuclear-to-cytoplasmic

    ATF2 ratio.

    Figure 4. SBI-0089410 and SBI-0087702 inhibit colony formation, cell motility, anchorage-

    independent growth, and promote the apoptosis of melanoma cells in an ATF2-dependent manner.

    (A) Panels A-F in right image show representative wells containing colonies of 501Mel melanoma cells

    cultured in the presence of screen hits and controls as follows: (A) DMSO, (B) Gö6850, (C) hit 1, (D) hit

    2, (E) SBI-0089410, and (F) SBI-0087702. 501Mel cells were plated at low density (500 cells/well in 6-

    well plates) and were grown in medium containing the indicated compounds. The number of colonies

    formed after 7 days in culture was determined by crystal violet staining. SBI-0089410 (410; E) and SBI-

    0087702 (702; F) were the most potent inhibitors of colony formation compared with Gö6850 (G).

    Similar results were obtained with the UACC903 cell line (data not shown). (B) Representative FACS

    profiles for pBabe empty vector (EV) or pBabe ATF2T52E (ATF2T52E)-stably transduced UACC903 cells

    that were treated with DMSO, 10 µM SBI-0089410 (410) or 10 µM SBI-0087702 (702) for 24 h. The

    cells were harvested, stained with Annexin V and propidium iodide (PI), and subjected to FACS analysis.

    N = 10,000 cells per replicate sample. (C) The histogram represents data from 3 independent experiments

    in (B) (ATF2T52E = 52E). (D) Quantification of the effect of hit compounds on the motility of WM1346

    cell lines stably expressing phospho-mimic ATF2 mutant (ATF2 52E) or empty vector (EV). Expression

    of ATF2 enhanced cell motility by approximately 2-fold (right columns). Both hit compounds inhibited

    migration of EV cells but expression of ATF2 52E rescued the inhibitory effect. Cell motility was

    determined in a modified Boyden chamber assay using the Calcein-AM fluorecent staining as described

    in material and methods. The experiments were performed in triplicate wells and results show averages

    and standard deviations. (E) SBI-0089410 and SBI-0087702 inhibited the spheroid growth of SW1

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 23

    melanoma cells. Representative images demonstrating the effect of hit compounds on SW1 spheroids

    growth. Spheroids were generated by the hanging drop method and subsequently transferred to separate

    wells and treated with the indicated compounds (10 µM) or vehicle (DMSO 0.08%) every other day for 8

    days, in triplicates. Scale bar = 200 µm. (F) Quantification of spheroid growth showing average and

    standard deviation from 3 independent wells. Phase-contrast images of spheroids were obtained using an

    Olympus light microscope and spheroid diameter was measure using the SlideBook software. Similar

    results were obtained with UACC903 cells.

    Figure 5. SBI-0089410 and SBI-0087702 promote mitochondrial leakage and reduced viability,

    which is block by expression of constitutively active PKCε. (A) SBI-0089410 (410) and SBI-0087702

    (702) induce ATF2-dependent mitochondrial membrane leakage. WM793 cells were incubated DMSO

    alone, 10 µM hits 1 or 2 for 24 h, 10 µM SBI-0089410 for 6 and 24 h, 10 µM SBI-0087702 for 24 h, or

    with 50 µM carbonyl cyanide 3-chlorophenylhydrazone (CCCP) for 45 min. Cell were then labeled with

    TMRE (250 µM) or NAO (10 nM) and analyzed by FACS. Samples were gated on whole cells by

    forward and side scatter and 10,000 gated cells were analyzed per sample. (B) Red FACS profiles

    represent TMRE uptake (reflective of mitochondrial membrane potential), whereas the green profiles

    represent NAO uptake (mitochondrial mass). Leftward peak shifts indicate decreased TMRE or NAO

    uptake, whereas rightward peak shifts indicate increased TMRE or NaO uptake. The dashed line indicates

    the median uptake values for the DMSO-treated samples. (C) WM793 cells were transiently transfected

    with control empty vector (EV) or ATF2T52E (pEF-HA-ATF2T52E) for 48 h. Cells were then treated with

    DMSO alone, 10 µM SBI-0089410 (410) for 6 h, 10 µM SBI-0087702 (702) for 24 h, or 50 µM CCCP

    for 45 min. The cells were labeled and analyzed as described for (A). Inset in (C): Western blot analysis

    showing HA-ATF2 expression. Histograms for (A) and (C) show the mean TMRE/NAO ratio values ±

    S.D. of 3 independent experiments. (D) Transient expression of a constitutively active form of PKCε

    (PKCε-CA) confers resistance to SBI-0087702-induced cell death. 501Mel melanoma cells were

    transiently transfected with either control GFP or PKCε-CA and incubated with the indicated compounds

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 24

    (10 µM final concentration) for 3 days before cell viability was measured using the CellTiter-Blue

    fluorescence assay (Promega). SBI-0087702-induced cell death was prevented by the expression of

    PKCε-CA.

    Figure 6. SBI-0089410 and SBI-0087702 inhibit melanoma cell growth, ATF2 transcriptional

    activity, and affect the expression of ATF2 target genes in a PKCε-dependent manner.

    (A) NIH3T3 cells were incubated with DMSO, 10 µM SBI-0089410 (410) for 6 h, 10 μM SBI-0087702

    (702) for 24 h, or with 10 μM of the PKCε inhibitor Gö6850. Anisomycin was then added for 30 min to

    prevent de novo protein synthesis and to activate cellular stress responses. Cells were lysed in RIPA

    buffer and analyzed by western blotting with the indicated antibodies. Protein bands were quantified

    using ImageJ software (numbers below). (B) SBI-0089410 (410) transiently inhibited ATF2 and p38

    phosphorylation in UACC903 melanoma cells. Cells were pretreated with SBI-0089410 for 3-9 h, before

    stimulation with anisomycin. SBI-0089410 inhibited ATF2 phosphorylation at Thr52 and p38

    phosphorylation with maximum activity after 6 h incubation. (C) SBI-0089410 inhibited the TPA-induced

    membrane translocation of PKC. WM793 cells were incubated in the presence of SBI-008410 or vehicle

    (DMSO) for 6 h, then stimulated with 100 nM TPA for 3 min, washed in cold PBS, lysed in lysis buffer,

    and the membrane and cytosol fractions were isolated according to the protocol detailed in the Material

    and Methods section. The different fractions were analyzed by Western blot using the antibodies

    indicated. (D) WM793 cells were treated with compounds and TPA as in (C) and subsequently fixed with

    4% formaldehyde prior to processing for immunofluorescence using anti-PKCε primary and FITC-

    conjugated goat anti-rabbit-IgG secondary antibodies. Imaging was conducted using an Olympus IX-71

    fluorescent microscope. Arrowheads point to PKCε staining at the plasma membrane and the insets show

    enlarged region near the plasma membrane. Scale-bar = 10 µm. (E) Luciferase assay evaluation of the

    effects of SBI-0089410 and SBI-0087702 on ATF2 transcriptional activity. UACC903 cells were stably

    transfected with a secreted 3X Jun2-(Gaussia) Luciferase construct. The cells were then transiently

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 25

    transfected with pCMV-Cypridina luciferase (normalization control), followed by treatment with DMSO,

    10 µM SBI-0089410, 10 μM SBI-0087702, or with 10 μM PKCε translocation peptide inhibitor (PKCε-

    i). At the indicated time-points (0, 1, 6, 24 h = T0, T1, T6, T24), Gaussia and Cypridina luciferase

    activities were measured, and Gaussia luciferase activity was normalized to Cypridina luciferase activity.

    The graph represents the mean Jun2-luciferase activities ± S.D. relative to DMSO of 3 replicate

    experiments. (F) IPA analysis of gene expression changes observed upon treatment with SBI-0087702

    identifies main clusters that are up- or downregulated. The lists of top 20 genes for each cluster are

    provided in Supplementary Tables 1 and 2. The original data were deposited into GEO (GSE43135).

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 26

    REFERENCES

    1. Ko JM, Fisher DE. A new era: melanoma genetics and therapeutics. The Journal of pathology. 2011;223:241-50.

    2. Shepherd C, Puzanov I, Sosman JA. B-RAF inhibitors: an evolving role in the therapy of malignant melanoma. Current oncology reports. 2010;12:146-52.

    3. Smalley KS. Understanding melanoma signaling networks as the basis for molecular targeted therapy. The Journal of investigative dermatology. 2010;130:28-37.

    4. Villanueva J, Vultur A, Lee JT, Somasundaram R, Fukunaga-Kalabis M, Cipolla AK, et al. Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer cell. 2010;18:683-95.

    5. Bollag G, Hirth P, Tsai J, Zhang J, Ibrahim PN, Cho H, et al. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature. 2010;467:596-9.

    6. Lopez-Bergami P, Lau E, Ronai Z. Emerging roles of ATF2 and the dynamic AP1 network in cancer. Nat Rev Cancer. 2010;10:65-76.

    7. Bhoumik A, Lopez-Bergami P, Ronai Z. ATF2 on the double - activating transcription factor and DNA damage response protein. Pigment Cell Res. 2007;20:498-506.

    8. Lau E, Ronai ZA. ATF2 - at the crossroad of nuclear and cytosolic functions. Journal of cell science. 2012;125:2815-24.

    9. Shah M, Bhoumik A, Goel V, Dewing A, Breitwieser W, Kluger H, et al. A role for ATF2 in regulating MITF and melanoma development. PLoS Genet. 2010;6:e1001258.

    10. Bhoumik A, Fichtman B, Derossi C, Breitwieser W, Kluger HM, Davis S, et al. Suppressor role of activating transcription factor 2 (ATF2) in skin cancer. Proc Natl Acad Sci U S A. 2008;105:1674-9.

    11. Maekawa T, Shinagawa T, Sano Y, Sakuma T, Nomura S, Nagasaki K, et al. Reduced levels of ATF-2 predispose mice to mammary tumors. Mol Cell Biol. 2007;27:1730-44.

    12. Berger AJ, Kluger HM, Li N, Kielhorn E, Halaban R, Ronai Z, et al. Subcellular localization of activating transcription factor 2 in melanoma specimens predicts patient survival. Cancer research. 2003;63:8103-7.

    13. Gould Rothberg BE, Berger AJ, Molinaro AM, Subtil A, Krauthammer MO, Camp RL, et al. Melanoma prognostic model using tissue microarrays and genetic algorithms. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009;27:5772-80.

    14. Lau E, Kluger H, Varsano T, Lee K, Scheffler I, Rimm DL, et al. PKCepsilon promotes oncogenic functions of ATF2 in the nucleus while blocking its apoptotic function at mitochondria. Cell. 2012;148:543-55.

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • 27

    15. Bhoumik A, Jones N, Ronai Z. Transcriptional switch by activating transcription factor 2-derived peptide sensitizes melanoma cells to apoptosis and inhibits their tumorigenicity. Proc Natl Acad Sci U S A. 2004;101:4222-7.

    16. Bhoumik A, Ivanov V, Ronai Z. Activating transcription factor 2-derived peptides alter resistance of human tumor cell lines to ultraviolet irradiation and chemical treatment. Clinical cancer research : an official journal of the American Association for Cancer Research. 2001;7:331-42.

    17. Bhoumik A, Huang TG, Ivanov V, Gangi L, Qiao RF, Woo SL, et al. An ATF2-derived peptide sensitizes melanomas to apoptosis and inhibits their growth and metastasis. J Clin Invest. 2002;110:643-50.

    18. Bhoumik A, Gangi L, Ronai Z. Inhibition of melanoma growth and metastasis by ATF2-derived peptides. Cancer research. 2004;64:8222-30.

    19. Halaban R, Alfano FD. Selective elimination of fibroblasts from cultures of normal human melanocytes. In vitro. 1984;20:447-50.

    20. Kelm JM, Timmins NE, Brown CJ, Fussenegger M, Nielsen LK. Method for generation of homogeneous multicellular tumor spheroids applicable to a wide variety of cell types. Biotechnology and bioengineering. 2003;83:173-80.

    21. Zhang JH, Chung TD, Oldenburg KR. A Simple Statistical Parameter for Use in Evaluation and Validation of High Throughput Screening Assays. Journal of biomolecular screening. 1999;4:67-73.

    22. Vaseva AV, Marchenko ND, Ji K, Tsirka SE, Holzmann S, Moll UM. p53 Opens the Mitochondrial Permeability Transition Pore to Trigger Necrosis. Cell. 2012;149:1536-48.

    23. Weinstein IB, Joe A. Oncogene addiction. Cancer research. 2008;68:3077-80; discussion 80.

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • A

    BTetOff

    +Doxycycline Doxycycline-Free 40 h

    GF

    P

    P

    ha

    se

    co

    ntr

    as

    t

    Actin

    anti-GFP

    GFP-ATF2

    ATF2

    UA

    CC

    903

    Dox Free + Dox

    UACC903 GFP-ATF2

    24 h 36 h 48 h 60 h 72 h 12 h 24 h

    Ponceau

    100

    100

    70

    40

    40

    anti-ATF2 (N96)

    GFP-ATF2

    ATF2

    DAPI ATF2 Overlay

    2 1

    2

    3

    4

    5

    56

    7

    Nuclear Metrics:

    ATF2 Metrics:

    Cell

    1

    2

    ---

    Area

    148

    146

    ---

    Int.

    817

    1070

    ---

    ---

    ---

    ---

    ---

    Cell

    1

    2

    ---

    Cyt I

    17

    16

    ---

    Nuc I

    267

    195

    ---

    ---

    ---

    ---

    ---

    Create Sub-Population

    GFP Int > Thresh

    Well Statistics:

    Mean, Std. Dev.,

    Count

    C

    Figure 1

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • A

    C

    Well number

    hit4

    Figure 2

    Nu

    cle

    ar-

    to-C

    yto

    pla

    sm

    ic M

    ea

    n In

    ten

    sit

    y R

    ati

    o

    [% A

    cti

    vit

    y, 0

    % =

    Ve

    hic

    le O

    nly

    , 1

    00

    % =

    10

    uM

    68

    50

    ]

    0

    50

    0

    10

    00

    15

    00

    20

    00

    25

    00

    30

    00

    35

    00

    40

    00

    45

    00

    250

    200

    150

    100

    50

    0

    -50

    -100

    NegControl

    PosControl

    SBI-0087702

    SBI-0089410

    (Other)

    SBI-0087702

    SBI-0089410

    Me

    an

    AT

    F2

    In

    ten

    sit

    y o

    f N

    uc

    lea

    r R

    eg

    ion

    [Z-S

    co

    re, b

    as

    ed

    on

    ne

    ga

    tiv

    e c

    on

    tro

    l w

    ells

    ]

    20

    15

    10

    5

    0

    -5

    -10

    -10 -5 0 5 10 15 20

    Mean ATF2 Intensity of Cytoplasmic Region

    [Z-Score, based on negative control wells]

    SBI-0089410

    SBI-0087702

    B

    SBI-0087702

    SBI-0089410

    HIT 1

    HIT 2

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • A

    AT

    F2

    (2

    0F

    1 Ig

    G)

    AT

    F2

    /HS

    P6

    0/D

    AP

    I

    DMSO

    B

    Nucle

    ar -to-c

    yto

    pla

    sm

    ic ratio

    GFP-ATF2

    UACC903

    WM793 UACC903

    GFP-ATF2 fluorescence Endogenous ATF2 immunofluorescence

    Nucle

    ar-

    to-c

    yto

    pla

    sm

    ic ratio

    TetOff

    Figure 3

    SBI-0089410SBI-0087702

    SBI-0

    0894

    10

    6 h

    SBI-0

    0877

    02

    24 h

    DM

    SO

    SBI-0

    0894

    10

    6 h

    SBI-0

    0877

    02

    24 h

    DM

    SO

    SBI-0

    0894

    10

    6 h

    SBI-0

    0877

    02

    24 h

    DM

    SO

    4

    3.5

    3

    2.5

    2

    1.5

    1

    0.5

    0

    3.5

    3

    2.5

    2

    1.5

    1

    0.5

    0

    3

    2.5

    2

    1.5

    1

    0.5

    0

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • G DM

    SO G

    oHit1

    Hit2 41

    070

    2

    A

    Figure 4

    DMSO SBI-0087702 SBI-0089410

    EV

    AT

    F2

    T5

    2E

    EV 52E EV 52E EV 52E% A

    nn

    ex

    in V

    +P

    I-p

    os

    itiv

    e c

    ells

    16

    14

    12

    10

    8

    6

    4

    2

    0

    B

    p = 0.0114

    p = 0.0039

    DMSO 702 410

    DC

    T0

    4 d

    ays

    8 d

    ays

    DMSO SBI-0087702 SBI-0089410

    0

    2

    4

    6

    8

    10

    12

    0 2 4 6 8

    Sp

    he

    re v

    olu

    me

    (m

    m3 )

    Time (Days)

    DMSO

    SBI-0087702

    SBI-0089410

    E F

    G

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • Figure 5

    A B

    C

    TM

    RE

    :NA

    O R

    ati

    os

    Re

    lati

    ve

    to

    DM

    SO

    DM

    SO

    410

    - 6 h

    702

    - 24

    h

    410

    - 24

    hHit 1

    Hit 2

    CCCP

    p = 0.0024

    TM

    RE

    :NA

    O R

    ati

    os

    Re

    lati

    ve

    to

    DM

    SO

    DM

    SO

    410

    - 6 h

    702

    - 24

    h

    CCCP

    DM

    SO

    410

    - 6 h

    702

    - 24

    h

    CCCP

    EV ATF2T52E

    !"

    #!"

    $!"

    %!"

    &!"

    '!!"

    '#!"

    '$!"

    !"

    #!"

    $!"

    %!"

    &!"

    '!!"

    '#!"

    '$!"

    501Mel-GFP 501Mel-PKCe-CA

    DM

    SO

    SBI-0

    0894

    10

    Gö6

    850

    SBI-0

    0877

    02

    DM

    SO

    SBI-0

    0894

    10

    Gö6

    850

    SBI-0

    0877

    02

    140

    120

    100

    80

    60

    40

    20

    0

    Via

    bili

    ty (

    % o

    f co

    ntr

    ol)

    140

    120

    100

    80

    60

    40

    20

    0

    Via

    bili

    ty (

    % o

    f co

    ntr

    ol)

    100

    84

    73

    3

    100 99104 101

    DMSO

    Hit 1

    Hit 2

    SBI-0089410, 24 h

    SBI-0089410, 6 h

    SBI-0087702

    CCCP

    TMRE Uptake NAO Uptake

    100 101 102 103 100 101 102 103

    DEV

    HA-T

    52E

    -55

    -55

    HA-ATF2 >

    ß-Tubulin >

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • Anisomycin:

    Compound:

    pATF2 (T52)

    pATF2 (T69/71)

    ATF2 (N96)

    p-p38

    p38

    Tubulin

    V V+ + + +

    41

    0 -

    3 h

    41

    0 -

    6 h

    41

    0 -

    6 h

    41

    0 -

    9 h

    UACC903

    0.0 0.4 1.0 0.8 0.4 0.7

    0.2 0.2 1.0 1.0 0.9 1.1

    0.5 0.4 1.0 0.8 0.4 0.7

    0.3 0.2 1.0 1.0 0.6 0.9

    0.6 0.7 1.0 1.0 1.0 1.0

    1.0 0.8 1.0 1.0 0.9 1.0

    D

    Figure 6

    PK

    DMSO DMSO + TPA SBI-0089410 + TPA

    Merg

    e +

    DA

    PI

    E

    A

    Anisomycin:

    Compound:

    pATF2 (T52)

    pATF2 (T69/71)

    ATF2 (N96)

    Tubulin

    V V Gö6850

    + + + +

    41

    0

    70

    2

    0.0 1.0 0.4 0.6 0.3

    0.0 1.0 1.0 0.9 0.6

    0.4 1.0 1.0 0.9 0.5

    1.3 1.0 1.0 0.9 1.0

    NIH3T3

    B C

    1.2

    1

    0.8

    0.6

    0.4

    0.2

    0T0 T1 T6 T24

    DMSO

    SBI-0089410

    SBI-0087702

    PKCε-i

    p = 0.0398

    Ju

    n2

    -Lu

    cif

    era

    se

    Ac

    tiv

    ity

    Re

    lati

    ve

    to

    DM

    SO

    Three most upregulated

    clusters

    (1) Lipid/small molecule/

    vitamin & mineral metabolism

    (2) Cellular development

    (3) Carbohydrate/

    small molecule/lipid

    metabolism

    Three most downregulated

    clusters

    (1) Tumor morphology/

    cell growth & proliferation/

    development

    (2) Cellular morphology

    (3) Cellular movement

    F

    PKCε

    PKCδ

    PKCα

    78

    78

    115

    90

    PKCμ

    Cy

    tos

    ol

    40Actin

    TPA - + - + - +

    DMSO 50 µM 25 µM

    SBI-0089410

    1.0 0.3 1.5 1.3 1.2 1.6

    1.0 0.5 1.2 1.3 1.2 1.6

    1.0 0.6 1.2 1.5 1.8 2.2

    1.0 0.1 1.1 1.0 1.2 1.8

    1.0 1.6 1.2 2.0 1.1 2.0

    Me

    mb

    . &

    Nu

    cl.

    Wh

    ole

    ce

    ll

    PKCε

    78

    90

    PKCδ

    pPKC

    (pan/T514)78

    Actin 40

    Actin 40

    100

    1.0 1.8 1.4 1.4 1.6 1.8

    1.0 1.0 0.9 0.8 0.8 0.8

    1.0 1.4 1.0 1.0 0.7 1.2

    1.0 1.0 0.8 0.9 0.8 0.9

    1.0 1.6 0.9 0.9 0.7 1.1

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/

  • Published OnlineFirst April 15, 2013.Clin Cancer Res Tal Varsano, Eric Lau, Yongmei Feng, et al. the Mitochondrial Localization of ATF2Inhibition of Melanoma Growth by Small Molecules that Promote

    Updated version

    10.1158/1078-0432.CCR-12-2689doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2013/04/17/1078-0432.CCR-12-2689.DC1

    Access the most recent supplemental material at:

    Manuscript

    Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

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

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://clincancerres.aacrjournals.org/content/early/2013/04/13/1078-0432.CCR-12-2689To request permission to re-use all or part of this article, use this link

    Research. on June 25, 2021. © 2013 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on April 15, 2013; DOI: 10.1158/1078-0432.CCR-12-2689

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-12-2689http://clincancerres.aacrjournals.org/content/suppl/2013/04/17/1078-0432.CCR-12-2689.DC1http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/early/2013/04/13/1078-0432.CCR-12-2689http://clincancerres.aacrjournals.org/

    Article FileFigure `Figure 2Figure 3Figure 4Figure 5Figure 6