Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Cell ; 175(5): 1244-1258.e26, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30454645

ABSTRACT

Cyclin-dependent kinase 9 (CDK9) promotes transcriptional elongation through RNAPII pause release. We now report that CDK9 is also essential for maintaining gene silencing at heterochromatic loci. Through a live cell drug screen with genetic confirmation, we discovered that CDK9 inhibition reactivates epigenetically silenced genes in cancer, leading to restored tumor suppressor gene expression, cell differentiation, and activation of endogenous retrovirus genes. CDK9 inhibition dephosphorylates the SWI/SNF protein BRG1, which contributes to gene reactivation. By optimization through gene expression, we developed a highly selective CDK9 inhibitor (MC180295, IC50 = 5 nM) that has broad anti-cancer activity in vitro and is effective in in vivo cancer models. Additionally, CDK9 inhibition sensitizes to the immune checkpoint inhibitor α-PD-1 in vivo, making it an excellent target for epigenetic therapy of cancer.


Subject(s)
Cyclin-Dependent Kinase 9/metabolism , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Methylation , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Cell ; 171(6): 1284-1300.e21, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-29195073

ABSTRACT

Combining DNA-demethylating agents (DNA methyltransferase inhibitors [DNMTis]) with histone deacetylase inhibitors (HDACis) holds promise for enhancing cancer immune therapy. Herein, pharmacologic and isoform specificity of HDACis are investigated to guide their addition to a DNMTi, thus devising a new, low-dose, sequential regimen that imparts a robust anti-tumor effect for non-small-cell lung cancer (NSCLC). Using in-vitro-treated NSCLC cell lines, we elucidate an interferon α/ß-based transcriptional program with accompanying upregulation of antigen presentation machinery, mediated in part through double-stranded RNA (dsRNA) induction. This is accompanied by suppression of MYC signaling and an increase in the T cell chemoattractant CCL5. Use of this combination treatment schema in mouse models of NSCLC reverses tumor immune evasion and modulates T cell exhaustion state towards memory and effector T cell phenotypes. Key correlative science metrics emerge for an upcoming clinical trial, testing enhancement of immune checkpoint therapy for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Drug Therapy, Combination , Lung Neoplasms/therapy , Tumor Escape/drug effects , Animals , Antigen Presentation/drug effects , Antineoplastic Agents/therapeutic use , Azacitidine/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Cell Line, Tumor , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Immunotherapy , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mice , T-Lymphocytes/immunology , Transcriptome , Tumor Microenvironment
3.
Cell ; 162(5): 974-86, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26317466

ABSTRACT

We show that DNA methyltransferase inhibitors (DNMTis) upregulate immune signaling in cancer through the viral defense pathway. In ovarian cancer (OC), DNMTis trigger cytosolic sensing of double-stranded RNA (dsRNA) causing a type I interferon response and apoptosis. Knocking down dsRNA sensors TLR3 and MAVS reduces this response 2-fold and blocking interferon beta or its receptor abrogates it. Upregulation of hypermethylated endogenous retrovirus (ERV) genes accompanies the response and ERV overexpression activates the response. Basal levels of ERV and viral defense gene expression significantly correlate in primary OC and the latter signature separates primary samples for multiple tumor types from The Cancer Genome Atlas into low versus high expression groups. In melanoma patients treated with an immune checkpoint therapy, high viral defense signature expression in tumors significantly associates with durable clinical response and DNMTi treatment sensitizes to anti-CTLA4 therapy in a pre-clinical melanoma model.


Subject(s)
DNA Methylation/drug effects , Interferon Type I/immunology , Melanoma/immunology , Melanoma/therapy , Animals , Azacitidine/pharmacology , Cell Line, Tumor , DNA Modification Methylases/antagonists & inhibitors , Endogenous Retroviruses/genetics , Female , Humans , Immunotherapy , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , RNA, Double-Stranded/metabolism
6.
Nature ; 579(7798): 284-290, 2020 03.
Article in English | MEDLINE | ID: mdl-32103175

ABSTRACT

Cancer recurrence after surgery remains an unresolved clinical problem1-3. Myeloid cells derived from bone marrow contribute to the formation of the premetastatic microenvironment, which is required for disseminating tumour cells to engraft distant sites4-6. There are currently no effective interventions that prevent the formation of the premetastatic microenvironment6,7. Here we show that, after surgical removal of primary lung, breast and oesophageal cancers, low-dose adjuvant epigenetic therapy disrupts the premetastatic microenvironment and inhibits both the formation and growth of lung metastases through its selective effect on myeloid-derived suppressor cells (MDSCs). In mouse models of pulmonary metastases, MDSCs are key factors in the formation of the premetastatic microenvironment after resection of primary tumours. Adjuvant epigenetic therapy that uses low-dose DNA methyltransferase and histone deacetylase inhibitors, 5-azacytidine and entinostat, disrupts the premetastatic niche by inhibiting the trafficking of MDSCs through the downregulation of CCR2 and CXCR2, and by promoting MDSC differentiation into a more-interstitial macrophage-like phenotype. A decreased accumulation of MDSCs in the premetastatic lung produces longer periods of disease-free survival and increased overall survival, compared with chemotherapy. Our data demonstrate that, even after removal of the primary tumour, MDSCs contribute to the development of premetastatic niches and settlement of residual tumour cells. A combination of low-dose adjuvant epigenetic modifiers that disrupts this premetastatic microenvironment and inhibits metastases may permit an adjuvant approach to cancer therapy.


Subject(s)
Epigenesis, Genetic , Genetic Therapy , Myeloid-Derived Suppressor Cells/physiology , Neoplasms/therapy , Tumor Microenvironment , Animals , Azacitidine/pharmacology , Benzamides/pharmacology , Cell Differentiation , Cell Movement/drug effects , Chemotherapy, Adjuvant , Disease Models, Animal , Down-Regulation/drug effects , Mice , Myeloid-Derived Suppressor Cells/cytology , Neoplasm Metastasis/therapy , Neoplasms/surgery , Pyridines/pharmacology , Receptors, CCR2/genetics , Receptors, Interleukin-8B/genetics , Tumor Microenvironment/drug effects
7.
Am J Pathol ; 192(2): 195-207, 2022 02.
Article in English | MEDLINE | ID: mdl-34767812

ABSTRACT

To catalyze severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) research, including development of novel interventive and preventive strategies, the progression of disease was characterized in a robust coronavirus disease 2019 (COVID-19) animal model. In this model, male and female golden Syrian hamsters were inoculated intranasally with SARS-CoV-2 USA-WA1/2020. Groups of inoculated and mock-inoculated uninfected control animals were euthanized at 2, 4, 7, 14, and 28 days after inoculation to track multiple clinical, pathology, virology, and immunology outcomes. SARS-CoV-2-inoculated animals consistently lost body weight during the first week of infection, had higher lung weights at terminal time points, and developed lung consolidation per histopathology and quantitative image analysis measurements. High levels of infectious virus and viral RNA were reliably present in the respiratory tract at days 2 and 4 after inoculation, corresponding with widespread necrosis and inflammation. At day 7, when the presence of infectious virus was rare, interstitial and alveolar macrophage infiltrates and marked reparative epithelial responses (type II hyperplasia) dominated in the lung. These lesions resolved over time, with only residual epithelial repair evident by day 28 after inoculation. The use of quantitative approaches to measure cellular and morphologic alterations in the lung provides valuable outcome measures for developing therapeutic and preventive interventions for COVID-19 using the hamster COVID-19 model.


Subject(s)
COVID-19/pathology , Animals , COVID-19/virology , Cricetinae , Disease Models, Animal , Female , Lung/pathology , Male , Mesocricetus , SARS-CoV-2
8.
Genes Dev ; 28(5): 432-7, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24589775

ABSTRACT

Despite recent progress, the physiological role of Hippo signaling in mammary gland development and tumorigenesis remains poorly understood. Here we show that the Hippo pathway is functionally dispensable in virgin mammary glands but specifically required during pregnancy. In contrast to many other tissues, hyperactivation of YAP in mammary epithelia does not induce hyperplasia but leads to defects in terminal differentiation. Interestingly, loss of YAP causes no obvious defects in virgin mammary glands but potently suppresses oncogene-induced mammary tumors. The selective requirement for YAP in oncogenic growth highlights the potential of YAP inhibitors as molecular targeted therapies against breast cancers.


Subject(s)
Carcinogenesis/pathology , Cell Differentiation , Mammary Glands, Animal/cytology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Female , Hippo Signaling Pathway , Mammary Glands, Animal/growth & development , Mice, Inbred C57BL , Mice, Transgenic , Oncogenes/genetics , Pregnancy , Time
9.
Proc Natl Acad Sci U S A ; 115(6): E1239-E1248, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29367423

ABSTRACT

Triple-negative breast cancer (TNBC) is treated with cytotoxic chemotherapy and is often characterized by early relapse and metastasis. To form a secondary (recurrent and/or metastatic) tumor, a breast cancer cell must evade the innate and adaptive immune systems. CD47 enables cancer cells to evade killing by macrophages, whereas CD73 and PDL1 mediate independent mechanisms of evasion of cytotoxic T lymphocytes. Here, we report that treatment of human or murine TNBC cells with carboplatin, doxorubicin, gemcitabine, or paclitaxel induces the coordinate transcriptional induction of CD47, CD73, and PDL1 mRNA and protein expression, leading to a marked increase in the percentage of CD47+CD73+PDL1+ breast cancer cells. Genetic or pharmacological inhibition of hypoxia-inducible factors (HIFs) blocked chemotherapy-induced enrichment of CD47+CD73+PDL1+ TNBC cells, which were also enriched in the absence of chemotherapy by incubation under hypoxic conditions, leading to T cell anergy or death. Treatment of mice with cytotoxic chemotherapy markedly increased the intratumoral ratio of regulatory/effector T cells, an effect that was abrogated by HIF inhibition. Our results delineate an HIF-dependent transcriptional mechanism contributing to TNBC progression and suggest that combining chemotherapy with an HIF inhibitor may prevent countertherapeutic induction of proteins that mediate evasion of innate and adaptive antitumor immunity.


Subject(s)
5'-Nucleotidase/metabolism , Antineoplastic Agents/pharmacology , B7-H1 Antigen/metabolism , CD47 Antigen/metabolism , Hypoxia-Inducible Factor 1/metabolism , Triple Negative Breast Neoplasms/immunology , Tumor Escape/immunology , 5'-Nucleotidase/genetics , Animals , Apoptosis , B7-H1 Antigen/genetics , CD47 Antigen/genetics , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia-Inducible Factor 1/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Proc Natl Acad Sci U S A ; 114(51): E10981-E10990, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29203668

ABSTRACT

Ovarian cancer is the most lethal of all gynecological cancers, and there is an urgent unmet need to develop new therapies. Epithelial ovarian cancer (EOC) is characterized by an immune suppressive microenvironment, and response of ovarian cancers to immune therapies has thus far been disappointing. We now find, in a mouse model of EOC, that clinically relevant doses of DNA methyltransferase and histone deacetylase inhibitors (DNMTi and HDACi, respectively) reduce the immune suppressive microenvironment through type I IFN signaling and improve response to immune checkpoint therapy. These data indicate that the type I IFN response is required for effective in vivo antitumorigenic actions of the DNMTi 5-azacytidine (AZA). Through type I IFN signaling, AZA increases the numbers of CD45+ immune cells and the percentage of active CD8+ T and natural killer (NK) cells in the tumor microenvironment, while reducing tumor burden and extending survival. AZA also increases viral defense gene expression in both tumor and immune cells, and reduces the percentage of macrophages and myeloid-derived suppressor cells in the tumor microenvironment. The addition of an HDACi to AZA enhances the modulation of the immune microenvironment, specifically increasing T and NK cell activation and reducing macrophages over AZA treatment alone, while further increasing the survival of the mice. Finally, a triple combination of DNMTi/HDACi plus the immune checkpoint inhibitor α-PD-1 provides the best antitumor effect and longest overall survival, and may be an attractive candidate for future clinical trials in ovarian cancer.


Subject(s)
Epigenesis, Genetic/drug effects , Immunomodulation/drug effects , Interferon Type I/metabolism , Ovarian Neoplasms/etiology , Ovarian Neoplasms/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents, Immunological , Azacitidine/pharmacology , Cell Line, Tumor , Disease Models, Animal , Female , Histone Deacetylase Inhibitors/pharmacology , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Tumor Burden/drug effects , Tumor Burden/immunology , Xenograft Model Antitumor Assays
11.
Mol Cell ; 39(5): 661-3, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20832717

ABSTRACT

Stem-like cells in human cancer (CSCs) have multiple properties of embryonic stem cells (ESCs). In this issue, Iliopoulos et al. (2010) identify molecular mechanisms that link miRNAs to epigenetic states and drive CSCs, contributing to tumor aggressiveness.

13.
Mol Cancer ; 12: 90, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23919753

ABSTRACT

BACKGROUND: Recent epidemiological studies demonstrate that both active and involuntary exposure to tobacco smoke increase the risk of breast cancer. Little is known, however, about the molecular mechanisms by which continuous, long term exposure to tobacco smoke contributes to breast carcinogenesis because most previous studies have focused on short term treatment models. In this work we have set out to investigate the progressive transforming effects of tobacco smoke on non-tumorigenic mammary epithelial cells and breast cancer cells using in vitro and in vivo models of chronic cigarette smoke exposure. RESULTS: We show that both non-tumorigenic (MCF 10A, MCF-12A) and tumorigenic (MCF7) breast epithelial cells exposed to cigarette smoke acquire mesenchymal properties such as fibroblastoid morphology, increased anchorage-independent growth, and increased motility and invasiveness. Moreover, transplantation experiments in mice demonstrate that treatment with cigarette smoke extract renders MCF 10A cells more capable to survive and colonize the mammary ducts and MCF7 cells more prone to metastasize from a subcutaneous injection site, independent of cigarette smoke effects on the host and stromal environment. The extent of transformation and the resulting phenotype thus appear to be associated with the differentiation state of the cells at the time of exposure. Analysis by flow cytometry showed that treatment with CSE leads to the emergence of a CD44(hi)/CD24(low) population in MCF 10A cells and of CD44+ and CD49f + MCF7 cells, indicating that cigarette smoke causes the emergence of cell populations bearing markers of self-renewing stem-like cells. The phenotypical alterations induced by cigarette smoke are accompanied by numerous changes in gene expression that are associated with epithelial to mesenchymal transition and tumorigenesis. CONCLUSIONS: Our results indicate that exposure to cigarette smoke leads to a more aggressive and transformed phenotype in human mammary epithelial cells and that the differentiation state of the cell at the time of exposure may be an important determinant in the phenotype of the final transformed state.


Subject(s)
Breast Neoplasms/etiology , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Smoking/adverse effects , Animals , Breast/metabolism , Breast/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Stem Cells/metabolism
14.
Blood ; 117(23): 6297-303, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21447826

ABSTRACT

Epstein-Barr virus (EBV) is associated with a variety of lymphoid malignancies. Bortezomib activates EBV lytic gene expression. Bortezomib, a proteasome inhibitor, leads to increased levels of CCAAT/enhancer-binding proteinß (C/EBPß) in a variety of tumor cell lines. C/EBPß activates the promoter of the EBV lytic switch gene ZTA. Bortezomib treatment leads to increased binding of C/EBP to previously recognized binding sites in the ZTA promoter. Knockdown of C/EBPß inhibits bortezomib activation of EBV lytic gene expression. Bortezomib also induces the unfolded protein response (UPR), as evidenced by increases in ATF4, CHOP10, and XBP1s and cleavage of ATF6. Thapsigargin, an inducer of the UPR that does not interfere with proteasome function, also induces EBV lytic gene expression. The effects of thapsigargin on EBV lytic gene expression are also inhibited by C/EBPß knock-down. Therefore, C/EBPß mediates the activation of EBV lytic gene expression associated with bortezomib and another UPR inducer.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Burkitt Lymphoma , CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Viral/drug effects , Herpesvirus 4, Human/physiology , Pyrazines/pharmacology , Response Elements , Virus Activation/drug effects , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 6/metabolism , Bortezomib , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/virology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Regulatory Factor X Transcription Factors , Thapsigargin/pharmacology , Trans-Activators/biosynthesis , Transcription Factor CHOP/metabolism , Transcription Factors/metabolism , Unfolded Protein Response/drug effects , X-Box Binding Protein 1
15.
Cancer Res ; 83(23): 3956-3973, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37747726

ABSTRACT

NUT carcinoma is an aggressive carcinoma driven by the BRD4-NUT fusion oncoprotein, which activates chromatin to promote expression of progrowth genes. BET bromodomain inhibitors (BETi) are a promising treatment for NUT carcinoma that can impede BRD4-NUT's ability to activate genes, but the efficacy of BETi as monotherapy is limited. Here, we demonstrated that enhancer of zeste homolog 2 (EZH2), which silences genes through establishment of repressive chromatin, is a dependency in NUT carcinoma. Inhibition of EZH2 with the clinical compound tazemetostat potently blocked growth of NUT carcinoma cells. Epigenetic and transcriptomic analysis revealed that tazemetostat reversed the EZH2-specific H3K27me3 silencing mark and restored expression of multiple tumor suppressor genes while having no effect on key oncogenic BRD4-NUT-regulated genes. Indeed, H3K27me3 and H3K27ac domains were found to be mutually exclusive in NUT carcinoma cells. CDKN2A was identified as the only gene among all tazemetostat-derepressed genes to confer resistance to tazemetostat in a CRISPR-Cas9 screen. Combined inhibition of EZH2 and BET synergized to downregulate cell proliferation genes, resulting in more pronounced growth arrest and differentiation than either inhibitor alone. In preclinical models, combined tazemetostat and BETi synergistically blocked tumor growth and prolonged survival of NUT carcinoma-xenografted mice, with complete remission without relapse in one cohort. Identification of EZH2 as a dependency in NUT carcinoma substantiates the reliance of NUT carcinoma tumor cells on epigenetic dysregulation of functionally opposite, yet highly complementary, chromatin regulatory pathways to maintain NUT carcinoma growth. SIGNIFICANCE: Repression of tumor suppressor genes, including CDKN2A, by EZH2 provides a mechanistic rationale for combining EZH2 and BET inhibitors for the clinical treatment of NUT carcinoma. See related commentary by Kazansky and Kentsis, p. 3827.


Subject(s)
Carcinoma , Nuclear Proteins , Animals , Humans , Mice , Carcinoma/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromatin , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Genes, Tumor Suppressor , Histones/metabolism , Neoplasm Recurrence, Local/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
bioRxiv ; 2023 Aug 16.
Article in English | MEDLINE | ID: mdl-37645799

ABSTRACT

NUT carcinoma (NC) is an aggressive carcinoma driven by the BRD4-NUT fusion oncoprotein, which activates chromatin to promote expression of pro-growth genes. BET bromodomain inhibitors (BETi) impede BRD4-NUT's ability to activate genes and are thus a promising treatment but limited as monotherapy. The role of gene repression in NC is unknown. Here, we demonstrate that EZH2, which silences genes through establishment of repressive chromatin, is a dependency in NC. Inhibition of EZH2 with the clinical compound tazemetostat (taz) potently blocked growth of NC cells. Epigenetic and transcriptomic analysis revealed that taz reversed the EZH2-specific H3K27me3 silencing mark, and restored expression of multiple tumor suppressor genes while having no effect on key oncogenic BRD4- NUT-regulated genes. CDKN2A was identified as the only gene amongst all taz-derepressed genes to confer resistance to taz in a CRISPR-Cas9 screen. Combined EZH2 inhibition and BET inhibition synergized to downregulate cell proliferation genes resulting in more pronounced growth arrest and differentiation than either inhibitor alone. In pre-clinical models, combined taz and BETi synergistically blocked growth and prolonged survival of NC-xenografted mice, with all mice cured in one cohort. STATEMENT OF SIGNIFICANCE: Identification of EZH2 as a dependency in NC substantiates the reliance of NC tumor cells on epigenetic dysregulation of functionally opposite, yet highly complementary chromatin regulatory pathways to maintain NC growth. In particular, repression of CDKN2A expression by EZH2 provides a mechanistic rationale for combining EZH2i with BETi for the clinical treatment of NC.

17.
Cancer Cell ; 6(4): 387-98, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15488761

ABSTRACT

The gene hypermethylated in cancer 1 (HIC1) is epigenetically inactivated, but not mutated, in cancer. Here we show that cooperative loss of Hic1 with p53, but not INK4a, yields distinct tumor phenotypes in mice. Germline deletion of one allele of each gene on the opposite chromosome yields breast and ovarian carcinomas and metastatic osteosarcomas with epigenetic inactivation of the wild-type Hic1 allele. Germline deletion of the two genes on the same chromosome results in earlier appearance and increased prevalence and aggressiveness of osteosarcomas with genetic deletion of both wild-type genes. In human osteosarcomas, hypermethylation of HIC1 is frequent only in tumors with p53 mutations. Our results indicate the importance of genes altered only through epigenetic mechanisms in cancer progression in conjunction with genetically modified tumor suppressor genes.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Epigenesis, Genetic/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Chromosome Deletion , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation , Genes, Tumor Suppressor , Heterozygote , Humans , Immunohistochemistry , Kruppel-Like Transcription Factors , Mice , Mice, Knockout , Mutation/genetics , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Osteosarcoma/genetics , Osteosarcoma/pathology , Phenotype , Promoter Regions, Genetic , Transcription Factors/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
18.
Mol Cancer ; 10: 100, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21854628

ABSTRACT

BACKGROUND: The transcription factor, CCAAT enhancer binding protein-ß (C/EBPß), is expressed as several distinct protein isoforms (LAP1, LAP2 and LIP) that have opposing actions in cellular proliferation and differentiation. Increases in the ratio of LIP/LAP are associated with aggressive, metastatic breast cancer; however, little is known regarding the molecular mechanisms that regulate LIP expression or the biological actions of an increase in the LIP/LAP ratio. Metastasis is highly dependent upon the suppression of anoikis and the role of C/EBPß and LIP in this anchorage-independent, survival process is currently not known in mammary epithelial cells. IGF-1R signaling is important for the survival of breast cancer cells and crosstalk between IGF-1R and EGFR signaling pathways have been implicated in the development of more aggressive disease. We therefore evaluated in mammary epithelial cells whether IGF-1R signaling regulates the LIP/LAP ratio, analyzed the potential interplay between EGFR and IGF-1R signaling and addressed the biological significance of increased LIP expression in cellular survival and suppression of anoikis. RESULTS: Our data provide the first evidence that IGF-1R signaling regulates LIP expression in an EGFR independent manner to increase the LIP/LAP ratio in mammary epithelial cells. Although crosstalk between IGF-1R signaling and EGFR signaling is detectable in MCF10A cells, this crosstalk is not required for the IGF-1 mediated regulation of LIP expression. Rather, the critical regulator of IGF-1 induced LIP expression appears to be EGFR-independent, Akt activity. Our data also demonstrate that increases in LIP expression promote cell survival via suppression of anoikis. Likewise, knockdown of total C/EBPß leads to increased cell death and suggest that C/EBPß expression is important for survival and resistance to anoikis. IGF-1 treatment can partially rescue vector control cells from anoikis; however, cells with reduced C/EBPß expression do not survive anoikis. CONCLUSIONS: Taken together, our data demonstrate that IGF-1R signaling regulates LIP expression in an EGFR independent manner to increase the LIP/LAP ratio in mammary epithelial cells. C/EBPß expression and elevations in LIP play an important role in regulating cellular survival via suppression of anoikis, in an IGF-1R mediated context or in a manner independent of IGF-1R signaling.


Subject(s)
Anoikis , CCAAT-Enhancer-Binding Protein-beta/metabolism , Receptor, IGF Type 1/physiology , Signal Transduction , Breast Neoplasms , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Line, Tumor , Cell Survival , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Gene Expression , Genes, Reporter , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Receptor, ErbB-2/metabolism , Receptor, IGF Type 1/pharmacology
19.
Oncogene ; 40(34): 5314-5326, 2021 08.
Article in English | MEDLINE | ID: mdl-34247191

ABSTRACT

Despite advances in breast cancer treatment, residual disease driven by dormant tumor cells continues to be a significant clinical problem. Leukemia inhibitory factor receptor (LIFR) promotes a dormancy phenotype in breast cancer cells and LIFR loss is correlated with poor patient survival. Herein, we demonstrate that histone deacetylase inhibitors (HDACi), which are in phase III clinical trials for breast cancer, epigenetically induced LIFR and activated a pro-dormancy program in breast cancer cells. HDACi slowed breast cancer cell proliferation and reduced primary tumor growth. Primary breast tumors from HDACi-treated patients had increased LIFR levels and reduced proliferation rates compared to pre-treatment levels. Recent Phase II clinical trial data studying entinostat and azacitidine in metastatic breast cancer revealed that induction of several pro-dormancy genes post-treatment was associated with prolonged patient survival. Together, these findings suggest HDACi as a potential therapeutic avenue to promote dormancy, prevent recurrence, and improve patient outcomes in breast cancer.


Subject(s)
Histone Deacetylase Inhibitors , Receptors, OSM-LIF , Breast , Breast Neoplasms , Humans , Leukemia Inhibitory Factor Receptor alpha Subunit , Phenotype
20.
Expert Rev Mol Med ; 11: e12, 2009 Apr 08.
Article in English | MEDLINE | ID: mdl-19351437

ABSTRACT

The CCAAT/enhancer-binding proteins (C/EBPs) are a family of leucine-zipper transcription factors that regulate gene expression to control cellular proliferation, differentiation, inflammation and metabolism. Encoded by an intronless gene, C/EBPbeta is expressed as several distinct protein isoforms (LAP1, LAP2, LIP) whose expression is regulated by the differential use of several in-frame translation start sites. LAP1 and LAP2 are transcriptional activators and are associated with differentiation, whereas LIP is frequently elevated in proliferative tissue and acts as a dominant-negative inhibitor of transcription. However, emerging evidence suggests that LIP can serve as a transcriptional activator in some cellular contexts, and that LAP1 and LAP2 might also have unique actions. The LIP:LAP ratio is crucial for the maintenance of normal growth and development, and increases in this ratio lead to aggressive forms of breast cancer. This review discusses the regulation of C/EBPbeta activity by post-translational modification, the individual actions of LAP1, LAP2 and LIP, and the functions and downstream targets that are unique to each isoform. The role of the C/EBPbeta isoforms in breast cancer is discussed and emphasis is placed on their interactions with receptor tyrosine kinases.


Subject(s)
Breast Neoplasms/etiology , CCAAT-Enhancer-Binding Proteins/physiology , Receptor Protein-Tyrosine Kinases/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Female , Humans , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL