Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(31): e2201376119, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35878022

ABSTRACT

Relapse to anti-HER2 monoclonal antibody (mAb) therapies, such as trastuzumab in HER2+ breast cancer (BC), is associated with residual disease progression due to resistance to therapy. Here, we identify interferon-γ inducible protein 16 (IFI16)-dependent STING signaling as a significant determinant of trastuzumab responses in HER2+ BC. We show that down-regulation of immune-regulated genes (IRG) is specifically associated with poor survival of HER2+, but not other BC subtypes. Among IRG, IFI16 is identified as a direct target of EZH2, the underexpression of which leads to deficient STING activation and downstream CXCL10/11 expression in response to trastuzumab treatment. Dual inhibition of EZH2 and histone deacetylase (HDAC) significantly activates IFI16-dependent immune responses to trastuzumab. Notably, a combination of a novel histone methylation inhibitor with an HDAC inhibitor induces complete tumor eradication and long-term T cell memory in a HER2+ BC mouse model. Our findings demonstrate an epigenetic regulatory mechanism suppressing the expression of the IFI16-CXCL10/11 signaling pathway that provides a survival advantage to HER2+ BC to confer resistance to trastuzumab treatment.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Membrane Proteins , Nuclear Proteins , Phosphoproteins , Trastuzumab , Animals , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Cell Line, Tumor , Chemokine CXCL10 , Chemokine CXCL11 , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunity , Membrane Proteins/metabolism , Mice , Neoplasm Recurrence, Local/genetics , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Receptor, ErbB-2/genetics , Signal Transduction , Trastuzumab/pharmacology
2.
Nat Commun ; 13(1): 4118, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35840558

ABSTRACT

The hypoxic tumor microenvironment has been implicated in immune escape, but the underlying mechanism remains elusive. Using an in vitro culture system modeling human T cell dysfunction and exhaustion in triple-negative breast cancer (TNBC), we find that hypoxia suppresses immune effector gene expression, including in T and NK cells, resulting in immune effector cell dysfunction and resistance to immunotherapy. We demonstrate that hypoxia-induced factor 1α (HIF1α) interaction with HDAC1 and concurrent PRC2 dependency causes chromatin remolding resulting in epigenetic suppression of effector genes and subsequent immune dysfunction. Targeting HIF1α and the associated epigenetic machinery can reverse the immune effector dysfunction and overcome resistance to PD-1 blockade, as demonstrated both in vitro and in vivo using syngeneic and humanized mice models. These findings identify a HIF1α-mediated epigenetic mechanism in immune dysfunction and provide a potential strategy to overcome immune resistance in TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Epigenesis, Genetic , Humans , Hypoxia/genetics , Immunotherapy/methods , Mice , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/therapy , Tumor Microenvironment/genetics
3.
Nat Commun ; 12(1): 4441, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34290255

ABSTRACT

BRD4, a Bromodomain and Extraterminal (BET) protein family member, is a promising anti-cancer drug target. However, resistance to BET inhibitors targeting BRD4 is common in solid tumors. Here, we show that cancer-associated fibroblast (CAF)-activated stromal signaling, interleukin-6/8-JAK2, induces BRD4 phosphorylation at tyrosine 97/98 in colorectal cancer, resulting in BRD4 stabilization due to interaction with the deubiquitinase UCHL3. BRD4 phosphorylation at tyrosine 97/98 also displays increased binding to chromatin but reduced binding to BET inhibitors, resulting in resistance to BET inhibitors. We further show that BRD4 phosphorylation promotes interaction with STAT3 to induce chromatin remodeling through concurrent binding to enhancers and super-enhancers, supporting a tumor-promoting transcriptional program. Inhibition of IL6/IL8-JAK2 signaling abolishes BRD4 phosphorylation and sensitizes BET inhibitors in vitro and in vivo. Our study reveals a stromal mechanism for BRD4 activation and BET inhibitor resistance, which provides a rationale for developing strategies to treat CRC more effectively.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm , Transcription Factors/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cancer-Associated Fibroblasts/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Chromatin/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Janus Kinase 2/metabolism , Phosphorylation , Protein Domains , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription Factors/antagonists & inhibitors , Tumor Microenvironment , Ubiquitin Thiolesterase/metabolism
4.
Nat Commun ; 11(1): 5878, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33208750

ABSTRACT

HER2-targeted therapy has yielded a significant clinical benefit in patients with HER2+ breast cancer, yet disease relapse due to intrinsic or acquired resistance remains a significant challenge in the clinic. Here, we show that the protein phosphatase 2A (PP2A) regulatory subunit PPP2R2B is a crucial determinant of anti-HER2 response. PPP2R2B is downregulated in a substantial subset of HER2+ breast cancers, which correlates with poor clinical outcome and resistance to HER2-targeted therapies. EZH2-mediated histone modification accounts for the PPP2R2B downregulation, resulting in sustained phosphorylation of PP2A targets p70S6K and 4EBP1 which leads to resistance to inhibition by anti-HER2 treatments. Genetic depletion or inhibition of EZH2 by a clinically-available EZH2 inhibitor restores PPP2R2B expression, abolishes the residual phosphorylation of p70S6K and 4EBP1, and resensitizes HER2+ breast cancer cells to anti-HER2 treatments both in vitro and in vivo. Furthermore, the same epigenetic mechanism also contributes to the development of acquired resistance through clonal selection. These findings identify EZH2-dependent PPP2R2B suppression as an epigenetic control of anti-HER2 resistance, potentially providing an opportunity to mitigate anti-HER2 resistance with EZH2 inhibitors.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Nerve Tissue Proteins/metabolism , Protein Phosphatase 2/metabolism , Receptor, ErbB-2/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Humans , Mice , Mice, Nude , Nerve Tissue Proteins/genetics , Protein Phosphatase 2/genetics , Quinazolines/administration & dosage , Receptor, ErbB-2/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
5.
J Exp Med ; 215(11): 2833-2849, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30266800

ABSTRACT

PTEN deficiency in breast cancer leads to resistance to PI3K-AKT inhibitor treatment despite aberrant activation of this signaling pathway. Here, we report that genetic depletion or small molecule inhibition of KDM4B histone demethylase activates the unfolded protein response (UPR) pathway and results in preferential apoptosis in PTEN-deficient triple-negative breast cancers (TNBCs). Intriguingly, this function of KDM4B on UPR requires its demethylase activity but is independent of its canonical role in histone modification, and acts through its cytoplasmic interaction with eIF2α, a crucial component of UPR signaling, resulting in reduced phosphorylation of this component. Targeting KDM4B in combination with PI3K inhibition induces further activation of UPR, leading to robust synergy in apoptosis. These findings identify KDM4B as a therapeutic vulnerability in PTEN-deficient TNBC that otherwise would be resistant to PI3K inhibition.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Jumonji Domain-Containing Histone Demethylases/metabolism , PTEN Phosphohydrolase/deficiency , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/drug therapy , Unfolded Protein Response/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Female , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Unfolded Protein Response/genetics , Xenograft Model Antitumor Assays
6.
Mol Cancer Ther ; 17(9): 1973-1983, 2018 09.
Article in English | MEDLINE | ID: mdl-29925528

ABSTRACT

Despite showing promise against PIK3CA-mutant breast cancers in preclinical studies, PI3K/AKT pathway inhibitors demonstrate limited clinical efficacy as monotherapy. Here, we found that histone H3K27me3 demethylase KDM6B-targeted IGFBP5 expression provides a protective mechanism for PI3K/AKT inhibitor-induced apoptosis in breast cancer cells. We found that overexpression of KDM6B and IGFBP5 in luminal breast cancer are positively associated with poorer disease outcomes. Mechanistically, KDM6B promotes IGFBP5 expression by antagonizing EZH2-mediated repression, and pharmacologic inhibition of KDM6B augments apoptotic response to PI3K/AKT inhibitor treatment. Moreover, the IGFBP5 expression is upregulated upon acquired resistance to the PI3K inhibitor GDC-0941, which is associated with an epigenetic switch from H3K27me3 to H3K27Ac at the IGFBP5 gene promoter. Intriguingly, GDC-0941-resistant breast cancer cells remained sensitive to KDM6B or IGFBP5 inhibition, indicating the dependency on the KDM6B-IGFBP5 axis to confer the survival advantage in GDC-0941-resistant cells. Our study reveals an epigenetic mechanism associated with resistance to targeted therapy and demonstrates that therapeutic targeting of KDM6B-mediated IGFBP5 expression may provide a useful approach to mitigate both intrinsic and acquired resistance to the PI3K inhibitor in breast cancer. Mol Cancer Ther; 17(9); 1973-83. ©2018 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Enhancer of Zeste Homolog 2 Protein/genetics , Indazoles/pharmacology , Insulin-Like Growth Factor Binding Protein 5/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Sulfonamides/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Drug Resistance, Neoplasm/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 5/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Kaplan-Meier Estimate , MCF-7 Cells , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
7.
Proc Natl Acad Sci U S A ; 115(26): E5990-E5999, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29891662

ABSTRACT

Colorectal cancer patients often relapse after chemotherapy, owing to the survival of stem or progenitor cells referred to as cancer stem cells (CSCs). Although tumor stromal factors are known to contribute to chemoresistance, it remains not fully understood how CSCs in the hypoxic tumor microenvironment escape the chemotherapy. Here, we report that hypoxia-inducible factor (HIF-1α) and cancer-associated fibroblasts (CAFs)-secreted TGF-ß2 converge to activate the expression of hedgehog transcription factor GLI2 in CSCs, resulting in increased stemness/dedifferentiation and intrinsic resistance to chemotherapy. Genetic or small-molecule inhibitor-based ablation of HIF-1α/TGF-ß2-mediated GLI2 signaling effectively reversed the chemoresistance caused by the tumor microenvironment. Importantly, high expression levels of HIF-1α/TGF-ß2/GLI2 correlated robustly with the patient relapse following chemotherapy, highlighting a potential biomarker and therapeutic target for chemoresistance in colorectal cancer. Our study thus uncovers a molecular mechanism by which hypoxic colorectal tumor microenvironment promotes cancer cell stemness and resistance to chemotherapy and suggests a potentially targeted treatment approach to mitigating chemoresistance.


Subject(s)
Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Neoplasm Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Transforming Growth Factor beta2/biosynthesis , Tumor Microenvironment , Zinc Finger Protein Gli2/biosynthesis , Cell Hypoxia , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Transforming Growth Factor beta2/genetics , Zinc Finger Protein Gli2/genetics
8.
Nat Med ; 23(11): 1319-1330, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28967919

ABSTRACT

Tumor recurrence remains the main reason for breast cancer-associated mortality, and there are unmet clinical demands for the discovery of new biomarkers and development of treatment solutions to benefit patients with breast cancer at high risk of recurrence. Here we report the identification of chromosomal copy-number amplification at 1q21.3 that is enriched in subpopulations of breast cancer cells bearing characteristics of tumor-initiating cells (TICs) and that strongly associates with breast cancer recurrence. Amplification is present in ∼10-30% of primary tumors but in more than 70% of recurrent tumors, regardless of breast cancer subtype. Detection of amplification in cell-free DNA (cfDNA) from blood is strongly associated with early relapse in patients with breast cancer and could also be used to track the emergence of tumor resistance to chemotherapy. We further show that 1q21.3-encoded S100 calcium-binding protein (S100A) family members, mainly S100A7, S100A8, and S100A9 (S100A7/8/9), and IL-1 receptor-associated kinase 1 (IRAK1) establish a reciprocal feedback loop driving tumorsphere growth. Notably, this functional circuitry can be disrupted by the small-molecule kinase inhibitor pacritinib, leading to preferential impairment of the growth of 1q21.3-amplified breast tumors. Our study uncovers the 1q21.3-directed S100A7/8/9-IRAK1 feedback loop as a crucial component of breast cancer recurrence, serving as both a trackable biomarker and an actionable therapeutic target for breast cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Chromosomes, Human, Pair 1 , Neoplasm Recurrence, Local/genetics , Animals , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Bridged-Ring Compounds/therapeutic use , Cell-Free Nucleic Acids/genetics , Disease Progression , Female , Heterografts , Humans , In Situ Hybridization, Fluorescence , Mice , Polymerase Chain Reaction , Pyrimidines/therapeutic use , Treatment Outcome
9.
Proc Natl Acad Sci U S A ; 113(26): E3735-44, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27303043

ABSTRACT

Despite the established oncogenic function of Polycomb repressive complex 2 (PRC2) in human cancers, its role as a tumor suppressor is also evident; however, the mechanism underlying the regulation of the paradoxical functions of PRC2 in tumorigenesis is poorly understood. Here we show that hypoxia-inducible factor 1, α-subunit (HIFI-α) is a crucial modulator of PRC2 and enhancer of zeste 2 (EZH2) function in breast cancer. Interrogating the genomic expression of breast cancer indicates high HIF1A activity correlated with high EZH2 expression but low PRC2 activity in triple-negative breast cancer compared with other cancer subtypes. In the absence of HIFIA activation, PRC2 represses the expression of matrix metalloproteinase genes (MMPs) and invasion, whereas a discrete Ezh2 complexed with Forkhead box M1 (FoxM1) acts to promote the expression of MMPs. HIF1-α induction upon hypoxia results in PRC2 inactivation by selective suppression of the expression of suppressor of zeste 12 protein homolog (SUZ12) and embryonic ectoderm development (EED), leading to a functional switch toward Ezh2/FoxM1-dependent induction of the expression of MMPs and invasion. Our study suggests a tumor-suppressive function of PRC2, which is restricted by HIF1-α, and an oncogenic function of Ezh2, which cooperates with FoxM1 to promote invasion in triple-negative breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism
11.
Nat Commun ; 6: 8746, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26503059

ABSTRACT

Metastatic tumour recurrence due to failed treatments remains a major challenge of breast cancer clinical management. Here we report that interleukin-1 receptor-associated kinase 1 (IRAK1) is overexpressed in a subset of breast cancers, in particular triple-negative breast cancer (TNBC), where it acts to drive aggressive growth, metastasis and acquired resistance to paclitaxel treatment. We show that IRAK1 overexpression confers TNBC growth advantage through NF-κB-related cytokine secretion and metastatic TNBC cells exhibit gain of IRAK1 dependency, resulting in high susceptibility to genetic and pharmacologic inhibition of IRAK1. Importantly, paclitaxel treatment induces strong IRAK1 phosphorylation, an increase in inflammatory cytokine expression, enrichment of cancer stem cells and acquired resistance to paclitaxel treatment. Pharmacologic inhibition of IRAK1 is able to reverse paclitaxel resistance by triggering massive apoptosis at least in part through inhibiting p38-MCL1 pro-survival pathway. Our study thus demonstrates IRAK1 as a promising therapeutic target for TNBC metastasis and paclitaxel resistance.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Interleukin-1 Receptor-Associated Kinases/genetics , Paclitaxel/administration & dosage , Animals , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Female , Humans , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Mice, SCID , Neoplasm Metastasis , Phosphorylation
12.
PLoS One ; 10(4): e0122983, 2015.
Article in English | MEDLINE | ID: mdl-25928216

ABSTRACT

Aberrant epigenetic events contribute to tumorigenesis of all human cancers. Significant efforts are underway in developing new generation of epigenetic cancer therapeutics. Although clinical trials for agents targeting DNA hypermethylation and histone deacetylation have yielded promising results, developing agents that target histone methylation remains to be in the early stage. We and others have previously reported that 3-Deazaneplanocin A (DZNep) is a histone methylation inhibitor that has a wide range of anticancer effects in various human cancers. Here, focusing on acute myeloid leukemia (AML) as a model, we reported a less toxic analog of DZNep, named D9, which is shown to be efficacious in AML cell lines and patient-derived samples in vitro, as well as AML tumorigenesis in vivo. Gene expression analysis in a panel of AML cell lines treated with D9 identified a set of genes that is associated with D9 sensitivity and implicated in multiple oncogenic signaling pathways. Moreover, we show that D9 is able to deplete the leukemia stem cells (LSC) and abolish chemotherapy-induced LSC enrichment, leading to dramatic elimination of AML cell survival. Thus, D9 appears to be a robust epigenetic compound that may constitute a potential for AML therapy.


Subject(s)
Adenosine , Drug Delivery Systems , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Female , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
13.
ChemMedChem ; 10(1): 173-82, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25319940

ABSTRACT

3-Deazaneplanocin A (DzNep) is a potential epigenetic drug for the treatment of various cancers. DzNep has been reported to deplete histone methylations, including oncogenic EZH2 complex, giving rise to epigenetic modifications that reactivate many silenced tumor suppressors in cancer cells. Despite its promise as an anticancer drug, little is known about the structure-activity relationships of DzNep in the context of epigenetic modifications and apoptosis induction. In this study, a number of analogues of DzNep were examined for DzNep-like ability to induce synergistic apoptosis in cancer cells in combination with trichostatin A, a known histone deacetylase (HDAC) inhibitor. The structure-activity relationship data thus obtained provide valuable information on the structural requirements for biological activity. The studies identified three compounds that show similar activities to DzNep. Two of these compounds show good pharmacokinetics and safety profiles. Attempts to correlate the observed synergistic apoptotic activities with measured S-adenosylhomocysteine hydrolase (SAHH) inhibitory activities suggest that the apoptotic activity of DzNep might not be directly due to its inhibition of SAHH.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/chemistry , Adenosine/therapeutic use , Adenosine/toxicity , Adenosylhomocysteinase/antagonists & inhibitors , Adenosylhomocysteinase/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Structure-Activity Relationship , Transplantation, Heterologous
14.
Cell Rep ; 8(1): 204-16, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24953652

ABSTRACT

Although small-molecule targeting of EZH2 appears to be effective in lymphomas carrying EZH2 activating mutations, finding similar approaches to target EZH2-overexpressing epithelial tumors remains challenging. In MYC-driven, but not PI3K-driven prostate cancer, we show that interferon-γ receptor 1 (IFNGR1) is directly repressed by EZH2 in a MYC-dependent manner and is downregulated in a subset of metastatic prostate cancers. EZH2 knockdown restored the expression of IFNGR1 and, when combined with IFN-γ treatment, led to strong activation of IFN-JAK-STAT1 tumor-suppressor signaling and robust apoptosis. Pharmacologic depletion of EZH2 by the histone-methylation inhibitor DZNep mimicked the effects of EZH2 knockdown on IFNGR1 induction and delivered a remarkable synergistic antitumor effect with IFN-γ. In contrast, although they efficiently depleted histone Lysine 27 trimethylation, EZH2 catalytic inhibitors failed to mimic EZH2 depletion. Thus, EZH2-inactivated IFN signaling may represent a therapeutic target, and patients with advanced prostate cancer driven by MYC may benefit from the combination of EZH2 and IFN-γ-targeted therapy.


Subject(s)
Polycomb Repressive Complex 2/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/therapeutic use , Animals , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein , Humans , Interferon-gamma/therapeutic use , Janus Kinases/metabolism , Male , Mice , Mice, Nude , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/genetics , Prostatic Neoplasms/drug therapy , Receptors, Interferon/metabolism , STAT1 Transcription Factor/metabolism , Interferon gamma Receptor
15.
Cancer Discov ; 3(10): 1156-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23887393

ABSTRACT

UNLABELLED: Although 3-phosphoinositide-dependent protein kinase-1 (PDK1) has been predominately linked to the phosphoinositide 3-kinase (PI3K)-AKT pathway, it may also evoke additional signaling outputs to promote tumorigenesis. Here, we report that PDK1 directly induces phosphorylation of Polo-like kinase 1 (PLK1), which in turn induces MYC phosphorylation and protein accumulation. We show that PDK1-PLK1-MYC signaling is critical for cancer cell growth and survival, and small-molecule inhibition of PDK1/PLK1 provides an effective approach for therapeutic targeting of MYC dependency. Intriguingly, PDK1-PLK1-MYC signaling induces an embryonic stem cell-like gene signature associated with aggressive tumor behaviors and is a robust signaling axis driving cancer stem cell (CSC) self-renewal. Finally, we show that a PLK1 inhibitor synergizes with an mTOR inhibitor to induce synergistic antitumor effects in colorectal cancer by antagonizing compensatory MYC induction. These findings identify a novel pathway in human cancer and CSC activation and provide a therapeutic strategy for targeting MYC-associated tumorigenesis and therapeutic resistance. SIGNIFICANCE: This work identifies PDK1­PLK1-MYC signaling as a new oncogenic pathway driving oncogenic transformation and CSC self-renewal. Targeted inhibition of PDK1/PLK1 is robust in targeting MYC dependency in cancer cells. Thus, our findings provide important insights into cancer and CSC biology and have significant therapeutic implications.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic , Neoplastic Stem Cells/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , 3-Phosphoinositide-Dependent Protein Kinases/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Humans , Molecular Targeted Therapy , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Polo-Like Kinase 1
16.
Cancer Cell ; 18(5): 459-71, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21075311

ABSTRACT

The PP2A serine/threonine protein phosphatase serves as a critical cellular regulator of cell growth, proliferation, and survival. However, how this pathway is altered in human cancer to confer growth advantage is largely unknown. Here, we show that PPP2R2B, encoding the B55ß regulatory subunit of the PP2A complex, is epigenetically inactivated by DNA hypermethylation in colorectal cancer. B55ß-associated PP2A interacts with PDK1 and modulates its activity toward Myc phosphorylation. On loss of PPP2R2B, mTORC1 inhibitor rapamycin triggers a compensatory Myc phosphorylation in PDK1-dependent, but PI3K and AKT-independent manner, resulting in resistance. Reexpression of PPP2R2B, genetic ablation of PDK1 or pharmacologic inhibition of PDK1 abrogates the rapamycin-induced Myc phosphorylation, leading to rapamycin sensitization. Thus, PP2A-B55ß antagonizes PDK1-Myc signaling and modulates rapamycin sensitivity.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Colorectal Neoplasms/enzymology , Nerve Tissue Proteins/metabolism , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Sirolimus/therapeutic use , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cellular Senescence , Class I Phosphatidylinositol 3-Kinases , Cluster Analysis , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA Methylation , Drug Resistance, Neoplasm , Epigenesis, Genetic , Humans , Mice , Nerve Tissue Proteins/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Phosphatase 2/physiology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/physiology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Signal Transduction , Transplantation, Heterologous
17.
Cancer Cell ; 13(6): 529-41, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18538736

ABSTRACT

Genetic and epigenetic defects in Wnt/beta-catenin signaling play important roles in colorectal cancer progression. Here we identify DACT3, a member of the DACT (Dpr/Frodo) gene family, as a negative regulator of Wnt/beta-catenin signaling that is transcriptionally repressed in colorectal cancer. Unlike other Wnt signaling inhibitors that are silenced by DNA methylation, DACT3 repression is associated with bivalent histone modifications. Remarkably, DACT3 expression can be robustly derepressed by a pharmacological combination that simultaneously targets both histone methylation and deacetylation, leading to strong inhibition of Dishevelled (Dvl)-mediated Wnt/beta-catenin signaling and massive apoptosis of colorectal cancer cells. Our study identifies DACT3 as an important regulator of Wnt/beta-catenin signaling in colorectal cancer and suggests a potential strategy for therapeutic control of Wnt/beta-catenin signaling in colorectal cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colorectal Neoplasms/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histones/metabolism , Signal Transduction/genetics , Wnt Proteins/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosylhomocysteinase/antagonists & inhibitors , Adenosylhomocysteinase/metabolism , Apoptosis , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Methylation , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/metabolism , Decitabine , Dishevelled Proteins , Down-Regulation , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Oligonucleotide Array Sequence Analysis , Phosphoproteins/metabolism , Promoter Regions, Genetic , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transfection , Wnt Proteins/genetics , beta Catenin/genetics
18.
Cancer Res ; 67(23): 11317-26, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18056458

ABSTRACT

Activation of the p53 tumor suppressor upon DNA damage elicits either cell cycle arrest or apoptosis, and the precise mechanism governing cell fate after p53 response has not been well defined. Through genomic analysis, we have identified the ribosomal protein S27-like (RPS27L) as a novel p53 transcriptional target gene. Although RPS27L mRNA levels were consistently induced after diverse p53 activating signals, its change in protein level was stimuli-dependent: it was up-regulated when cells were arrested in response to DNA-damaging agents Adriamycin or VP16 but was down-regulated when cells underwent apoptosis in response to antimetabolite agent 5-fluorouracil. RPS27L is a nuclear protein that forms nuclear foci upon DNA damage. Depletion of RPS27L resulted in deficiency in DNA damage checkpoints, leading to conversion of DNA damage-induced p53 response from cell cycle arrest to apoptosis. We further show that RPS27L positively regulates p21 protein expression. Through this mechanism, RPS27L induction by p53 facilitates p21-mediated cell cycle arrest and protects against DNA damage-induced apoptosis. Thus, RPS27L modulates DNA damage response and functions as a part of the control switch to determine cell fate to DNA damage-p53 response.


Subject(s)
Apoptosis/physiology , Biomarkers, Tumor/metabolism , DNA Damage , Gene Expression Regulation, Neoplastic , Metalloproteins/genetics , Nuclear Proteins/genetics , RNA-Binding Proteins/genetics , Ribosomal Proteins/genetics , Tumor Suppressor Protein p53/metabolism , Biomarkers, Tumor/genetics , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Repair , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Genomic Instability , HCT116 Cells , Humans , Luciferases/metabolism , Metalloproteins/metabolism , Micronucleus Tests , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Zinc Fingers
19.
Genes Dev ; 21(9): 1050-63, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17437993

ABSTRACT

Polycomb-repressive complex 2 (PRC2)-mediated histone methylation plays an important role in aberrant cancer gene silencing and is a potential target for cancer therapy. Here we show that S-adenosylhomocysteine hydrolase inhibitor 3-Deazaneplanocin A (DZNep) induces efficient apoptotic cell death in cancer cells but not in normal cells. We found that DZNep effectively depleted cellular levels of PRC2 components EZH2, SUZ12, and EED and inhibited associated histone H3 Lys 27 methylation (but not H3 Lys 9 methylation). By integrating RNA interference (RNAi), genome-wide expression analysis, and chromatin immunoprecipitation (ChIP) studies, we have identified a prominent set of genes selectively repressed by PRC2 in breast cancer that can be reactivated by DZNep. We further demonstrate that the preferential reactivation of a set of these genes by DZNep, including a novel apoptosis affector, FBXO32, contributes to DZNep-induced apoptosis in breast cancer cells. Our results demonstrate the unique feature of DZNep as a novel chromatin remodeling compound and suggest that pharmacologic reversal of PRC2-mediated gene repression by DZNep may constitute a novel approach for cancer therapy.


Subject(s)
Apoptosis/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosylhomocysteinase/antagonists & inhibitors , Apoptosis/drug effects , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Female , Histones/metabolism , Humans , Methylation , Polycomb-Group Proteins , Promoter Regions, Genetic , RNA Interference , RNA Polymerase II/metabolism , RNA, Small Interfering/genetics , Repressor Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...