Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
2.
Proc Natl Acad Sci U S A ; 120(4): e2218118120, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36652476

ABSTRACT

Although epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (EGFRi) are approved for treating EGFR-mutant lung adenocarcinoma (LUAD), emergence of acquired resistance limits their clinical benefits. Several mechanisms for acquired resistance to EGFRi in LUAD have been identified; however, the molecular basis for this resistance remains unknown in ~30% of LUAD. Chromatin and DNA modifiers and their regulators play important roles in determining response to anticancer therapies. Therefore, to identify nongenetic mechanisms of EGFRi resistance in LUAD, we performed an epigenome-wide shRNA screen targeting 363 human epigenetic regulator genes. This screen identified loss of the transcriptional repressor chromobox homolog 5 (CBX5) as a driver of EGFRi resistance in EGFR-mutant LUAD. Loss of CBX5 confers resistance to multiple EGFRi in both cell culture and mice. We found that CBX5 loss in EGFR-mutant LUAD cells leads to increased expression of the transcription factor E2F1, which in turn stimulates expression of the antiapoptotic gene BIRC5 (survivin). This E2F1-mediated upregulation of BIRC5 in CBX5-knockdown LUAD cells attenuates apoptosis induction following EGFRi treatment. Consistent with these results, knockdown of E2F1 or BIRC5 partly rescues CBX5-knockdown-induced EGFRi resistance in cell culture and mice. EGFRi-resistant LUAD cell lines show reduced CBX5 expression compared to parental lines; however, bromo- and extra-terminal (BET)-domain inhibitors (BETi) restore CBX5 expression in these cells and sensitize them to EGFRi/BETi combination therapy. Similarly, treatment with a BIRC5 inhibitor suppresses growth of EGFRi-resistant LUAD cells. Collectively, these studies identify CBX5 loss as a driver of EGFRi resistance and reveal therapeutic opportunities for treating EGFRi-resistant LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Animals , Mice , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , ErbB Receptors/metabolism , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor
3.
Mol Cell ; 67(4): 685-701.e6, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28803777

ABSTRACT

Metabolic deregulation is a hallmark of human cancers, and the glycolytic and glutamine metabolism pathways were shown to be deregulated in pancreatic ductal adenocarcinoma (PDAC). To identify new metabolic regulators of PDAC tumor growth and metastasis, we systematically knocked down metabolic genes that were overexpressed in human PDAC tumor samples using short hairpin RNAs. We found that p53 transcriptionally represses paraoxonase 2 (PON2), which regulates GLUT1-mediated glucose transport via stomatin. The loss of PON2 initiates the cellular starvation response and activates AMP-activated protein kinase (AMPK). In turn, AMPK activates FOXO3A and its transcriptional target, PUMA, which induces anoikis to suppress PDAC tumor growth and metastasis. Pharmacological or genetic activation of AMPK, similar to PON2 inhibition, blocks PDAC tumor growth. Collectively, our results identify PON2 as a new modulator of glucose transport that regulates a pharmacologically tractable pathway necessary for PDAC tumor growth and metastasis.


Subject(s)
Aryldialkylphosphatase/metabolism , Carcinoma, Pancreatic Ductal/enzymology , Cell Movement , Cell Proliferation , Energy Metabolism , Glucose Transporter Type 1/metabolism , Liver Neoplasms/enzymology , Lung Neoplasms/enzymology , Pancreatic Neoplasms/enzymology , AMP-Activated Protein Kinases/analysis , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Aryldialkylphosphatase/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Energy Metabolism/drug effects , Female , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Glucose Transporter Type 1/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Lung Neoplasms/genetics , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Mice, Nude , Mutation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , RNA Interference , Signal Transduction , Time Factors , Transcription, Genetic , Transfection , Tumor Burden , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
4.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: mdl-34301901

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is a histone H3 lysine 27 methyltransferase that has been shown to function as an oncogene in some cancers. Previous reports have largely focused on the ability of EZH2 to regulate cell-intrinsic tumor regulatory pathways as its mechanism-of-oncogenic action. However, the role that EZH2-mediated immune suppression plays in its oncogenic activity is not fully known. In particular, the role of natural killer (NK) cells in EZH2-driven tumor growth remains incompletely understood. Here, we demonstrate that genetic or pharmacological inhibition of EZH2 induces reexpression of the chemokine CXCL10 in hepatic tumor cells. We find that histone deacetylase 10 (HDAC10) is necessary for EZH2 recruitment to the CXCL10 promoter, leading to CXCL10 transcriptional repression. Critically, CXCL10 is necessary and sufficient for stimulating NK cell migration, and EZH2's ability to inhibit NK cell migration via CXCL10 suppression is conserved in other EZH2-dependent cancers. NK cell depletion in an immunocompetent syngeneic mouse model of hepatic tumorigenesis reverses the tumor inhibitory effects of an EZH2 inhibitor (GSK343), and inhibitor-mediated reexpression of CXCL10 is required for its tumor suppressive effects in the same mouse model. Collectively, these results reveal a decisive role for NK cells and CXCL10 in mediating the oncogenic function of EZH2.


Subject(s)
Carcinoma, Hepatocellular/immunology , Chemokine CXCL10/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylases/metabolism , Animals , Benzopyrans/pharmacology , Cell Line, Tumor , Cell Movement , Chemokine CXCL10/genetics , Chemokines/genetics , Chemokines/metabolism , Decitabine/pharmacology , Enhancer of Zeste Homolog 2 Protein/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/immunology , Histone Deacetylases/genetics , Humans , Indazoles/pharmacology , Indoles/pharmacology , Killer Cells, Natural , Liver Neoplasms , Male , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/drug therapy , Phenols/pharmacology , Pyridones/pharmacology , Sulfones/pharmacology , Triazoles/pharmacology
5.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Article in English | MEDLINE | ID: mdl-33531346

ABSTRACT

Unlike other cell types, developing B cells undergo multiple rounds of somatic recombination and hypermutation to evolve high-affinity antibodies. Reflecting the high frequency of DNA double-strand breaks, adaptive immune protection by B cells comes with an increased risk of malignant transformation. B lymphoid transcription factors (e.g., IKZF1 and PAX5) serve as metabolic gatekeepers by limiting glucose to levels insufficient to fuel transformation. We here identified aberrant expression of the lactonase PON2 in B cell acute lymphoblastic leukemia (B-ALL) as a mechanism to bypass metabolic gatekeeper functions. Compared to normal pre-B cells, PON2 expression was elevated in patient-derived B-ALL samples and correlated with poor clinical outcomes in pediatric and adult cohorts. Genetic deletion of Pon2 had no measurable impact on normal B cell development. However, in mouse models for BCR-ABL1 and NRASG12D-driven B-ALL, deletion of Pon2 compromised proliferation, colony formation, and leukemia initiation in transplant recipient mice. Compromised leukemogenesis resulted from defective glucose uptake and adenosine triphosphate (ATP) production in PON2-deficient murine and human B-ALL cells. Mechanistically, PON2 enabled glucose uptake by releasing the glucose-transporter GLUT1 from its inhibitor stomatin (STOM) and genetic deletion of STOM largely rescued PON2 deficiency. While not required for glucose transport, the PON2 lactonase moiety hydrolyzes the lactone-prodrug 3OC12 to form a cytotoxic intermediate. Mirroring PON2 expression levels in B-ALL, 3OC12 selectively killed patient-derived B-ALL cells but was well tolerated in transplant recipient mice. Hence, while B-ALL cells critically depend on aberrant PON2 expression to evade metabolic gatekeeper functions, PON2 lactonase activity can be leveraged as synthetic lethality to overcome drug resistance in refractory B-ALL.


Subject(s)
Aryldialkylphosphatase/metabolism , B-Lymphocytes/metabolism , Carcinogenesis/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Adenosine Triphosphate/metabolism , Animals , Aryldialkylphosphatase/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Cells, Cultured , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Binding
6.
Cell ; 132(3): 363-74, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18267069

ABSTRACT

Expression of an oncogene in a primary cell can, paradoxically, block proliferation by inducing senescence or apoptosis through pathways that remain to be elucidated. Here we perform genome-wide RNA-interference screening to identify 17 genes required for an activated BRAF oncogene (BRAFV600E) to block proliferation of human primary fibroblasts and melanocytes. Surprisingly, we find a secreted protein, IGFBP7, has a central role in BRAFV600E-mediated senescence and apoptosis. Expression of BRAFV600E in primary cells leads to synthesis and secretion of IGFBP7, which acts through autocrine/paracrine pathways to inhibit BRAF-MEK-ERK signaling and induce senescence and apoptosis. Apoptosis results from IGFBP7-mediated upregulation of BNIP3L, a proapoptotic BCL2 family protein. Recombinant IGFBP7 (rIGFBP7) induces apoptosis in BRAFV600E-positive human melanoma cell lines, and systemically administered rIGFBP7 markedly suppresses growth of BRAFV600E-positive tumors in xenografted mice. Immunohistochemical analysis of human skin, nevi, and melanoma samples implicates loss of IGFBP7 expression as a critical step in melanoma genesis.


Subject(s)
Apoptosis , Cellular Senescence , Insulin-Like Growth Factor Binding Proteins/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Amino Acid Substitution , Animals , Autocrine Communication , Cell Line, Tumor , Cell Proliferation , Fibroblasts/cytology , Humans , Insulin-Like Growth Factor Binding Proteins/genetics , MAP Kinase Signaling System , Melanocytes/cytology , Melanocytes/metabolism , Melanoma/genetics , Melanoma/metabolism , Membrane Proteins/metabolism , Mice , Neoplasm Transplantation , Nevus, Pigmented/metabolism , Paracrine Communication , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf/genetics , RNA Interference , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transplantation, Heterologous , Tumor Suppressor Proteins/metabolism , Up-Regulation
7.
Proc Natl Acad Sci U S A ; 117(22): 12341-12351, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32430335

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with a dismal prognosis. Currently, there is no effective therapy for PDAC, and a detailed molecular and functional evaluation of PDACs is needed to identify and develop better therapeutic strategies. Here we show that the transcription factor Krüppel-like factor 7 (KLF7) is overexpressed in PDACs, and that inhibition of KLF7 blocks PDAC tumor growth and metastasis in cell culture and in mice. KLF7 expression in PDACs can be up-regulated due to activation of a MAP kinase pathway or inactivation of the tumor suppressor p53, two alterations that occur in a large majority of PDACs. ShRNA-mediated knockdown of KLF7 inhibits the expression of IFN-stimulated genes (ISGs), which are necessary for KLF7-mediated PDAC tumor growth and metastasis. KLF7 knockdown also results in the down-regulation of Discs Large MAGUK Scaffold Protein 3 (DLG3), resulting in Golgi complex fragmentation, and reduced protein glycosylation, leading to reduced secretion of cancer-promoting growth factors, such as chemokines. Genetic or pharmacologic activation of Golgi complex fragmentation blocks PDAC growth and metastasis similar to KLF7 inhibition. Our results demonstrate a therapeutically amenable, KLF7-driven pathway that promotes PDAC growth and metastasis by activating ISGs and maintaining Golgi complex integrity.


Subject(s)
Golgi Apparatus/metabolism , Kruppel-Like Transcription Factors/metabolism , Pancreatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Golgi Apparatus/genetics , Humans , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Knockout , Neoplasm Metastasis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/physiopathology , Transcription Factors/genetics , Transcription Factors/metabolism , Pancreatic Neoplasms
8.
PLoS Genet ; 15(10): e1008439, 2019 10.
Article in English | MEDLINE | ID: mdl-31589613

ABSTRACT

Metabolic alterations that are critical for cancer cell growth and metastasis are one of the key hallmarks of cancer. Here, we show that thymidine kinase 1 (TK1) is significantly overexpressed in tumor samples from lung adenocarcinoma (LUAD) patients relative to normal controls, and this TK1 overexpression is associated with significantly reduced overall survival and cancer recurrence. Genetic knockdown of TK1 with short hairpin RNAs (shRNAs) inhibits both the growth and metastatic attributes of LUAD cells in culture and in mice. We further show that transcriptional overexpression of TK1 in LUAD cells is driven, in part, by MAP kinase pathway in a transcription factor MAZ dependent manner. Using targeted and gene expression profiling-based approaches, we then show that loss of TK1 in LUAD cells results in reduced Rho GTPase activity and reduced expression of growth and differentiation factor 15 (GDF15). Furthermore, ectopic expression of GDF15 can partially rescue TK1 knockdown-induced LUAD growth and metastasis inhibition, confirming its important role as a downstream mediator of TK1 function in LUAD. Collectively, our findings demonstrate that TK1 facilitates LUAD tumor and metastatic growth and represents a target for LUAD therapy.


Subject(s)
Adenocarcinoma of Lung/genetics , Growth Differentiation Factor 15/metabolism , Lung Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Thymidine Kinase/metabolism , Adenocarcinoma of Lung/mortality , Adenocarcinoma of Lung/pathology , Adult , Aged , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , DNA-Binding Proteins/metabolism , Datasets as Topic , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung/pathology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Neoplasm Recurrence, Local/pathology , Prognosis , Survival Analysis , Thymidine Kinase/genetics , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
9.
Proc Natl Acad Sci U S A ; 116(10): 4583-4591, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30782837

ABSTRACT

Acquired resistance to BRAF kinase inhibitors (BRAFi) is the primary cause for their limited clinical benefit. Although several mechanisms of acquired BRAFi resistance have been identified, the basis for acquired resistance remains unknown in over 40% of melanomas. We performed a large-scale short-hairpin RNA screen, targeting 363 epigenetic regulators and identified Block of Proliferation 1 (BOP1) as a factor the loss of which results in resistance to BRAFi both in cell culture and in mice. BOP1 knockdown promoted down-regulation of the MAPK phosphatases DUSP4 and DUSP6 via a transcription-based mechanism, leading to increased MAPK signaling and BRAFi resistance. Finally, analysis of matched patient-derived BRAFi or BRAFi+MEKi pre- and progressed melanoma samples revealed reduced BOP1 protein expression in progressed samples. Collectively, our results demonstrate that loss of BOP1 and the resulting activation of the MAPK pathway is a clinically relevant mechanism for acquired resistance to BRAFi in melanoma.


Subject(s)
Drug Resistance, Neoplasm , Melanoma/genetics , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , RNA-Binding Proteins/genetics , Animals , Cell Line, Tumor , Dual Specificity Phosphatase 6/genetics , Dual Specificity Phosphatase 6/metabolism , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Female , Gene Deletion , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , Male , Melanoma/drug therapy , Melanoma/enzymology , Mice , Mice, Knockout , Mice, Nude , Mitogen-Activated Protein Kinase Phosphatases/genetics , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , RNA-Binding Proteins/metabolism
10.
Proc Natl Acad Sci U S A ; 115(15): E3509-E3518, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29581297

ABSTRACT

Natural killer (NK) cell-mediated tumor cell eradication could inhibit tumor initiation and progression. However, the factors that regulate NK cell-mediated cancer cell eradication remain unclear. We determined that hepatocellular carcinoma (HCC) cells exhibit transcriptional down-regulation of NK group 2D (NKG2D) ligands and are largely resistant to NK cell-mediated eradication. Because the down-regulation of NKG2D ligands occurred at the transcriptional level, we tested 32 chemical inhibitors of epigenetic regulators for their ability to re-express NKG2D ligands and enhance HCC cell eradication by NK cells and found that Enhancer of zeste homolog 2 (EZH2) was a transcriptional repressor of NKG2D ligands. The inhibition of EZH2 by small-molecule inhibitors or genetic means enhanced HCC cell eradication by NK cells in a NKG2D ligand-dependent manner. Collectively, these results demonstrate that EZH2 inhibition enhances HCC eradication by NK cells and that EZH2 functions, in part, as an oncogene by inhibiting immune response.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Killer Cells, Natural/immunology , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Small Molecule Libraries/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Methyltransferase 3A , Down-Regulation , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Ligands , Liver Neoplasms/genetics , Liver Neoplasms/pathology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Promoter Regions, Genetic
11.
Genes Dev ; 27(20): 2221-6, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24105743

ABSTRACT

We previously identified 28 cofactors through which a RAS oncoprotein directs transcriptional silencing of Fas and other tumor suppressor genes (TSGs). Here we performed RNAi-based epistasis experiments and found that RAS-directed silencing occurs through a highly ordered pathway that is initiated by binding of ZFP354B, a sequence-specific DNA-binding protein, and culminates in recruitment of the DNA methyltransferase DNMT1. RNAi and pharmacological inhibition experiments reveal that silencing requires continuous function of RAS and its cofactors and can be rapidly reversed, which may have therapeutic implications for reactivation of silenced TSGs in RAS-positive cancers.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Tumor Suppressor , ras Proteins/metabolism , Animals , DNA Methylation , Epigenesis, Genetic , Mice , Models, Biological , NIH 3T3 Cells , Protein Binding , RNA Interference , Signal Transduction , fas Receptor/genetics , fas Receptor/metabolism , ras Proteins/genetics
12.
Blood ; 131(19): 2125-2137, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29572378

ABSTRACT

Resistance to targeted therapies has become increasingly prevalent. We noted that resistance to different targeted therapies occurs by largely common mechanisms. In this study, we used this information for identifying the mechanisms of resistance to enhancer of zeste homolog 2 (EZH2) inhibitors in diffuse large B-cell lymphoma (DLBCL) harboring EZH2 mutations. We discovered that EZH2 inhibitor-resistant DLBCL cells showed activation of the insulin-like growth factor 1 receptor (IGF-1R), MEK, and phosphoinositide-3-kinase (PI3K) pathways. Constitutive activation of IGF-1R, MEK, or PI3K pathways was sufficient to confer resistance to EZH2 inhibitors in DLBCL. The activation of the PI3K/AKT and MAPK pathways decreased TNFSF10 and BAD expression through a FOXO3-dependent mechanism, which was required for the antitumor effects of EZH2i GSK126. We also identified multiple acquired mutations in EZH2 inhibitor-resistant DLBCL cell lines. These mutations independently conferred resistance to EZH2 inhibitors. Mechanistically, cellular thermal shift assays revealed that the acquired EZH2 mutations that confer resistance to EZH2 inhibitors prevent EZH2 inhibitor binding to the EZH2 mutants. Notably, EZH2 inhibitor GSK126- and EPZ-6438-resistant DLBCL cells remained sensitive to the EZH2 inhibitor UNC1999 and embryonic ectoderm development protein inhibitor EED226, which provides an opportunity to treat DLBCLs that are resistant to these drugs. Collectively, our results underpin the importance for developing a unified approach for forestalling drug resistance by prospectively considering lessons learned from the use of different targeted therapeutic agents.


Subject(s)
Drug Resistance, Neoplasm , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Lymphoma, Large B-Cell, Diffuse/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Enhancer of Zeste Homolog 2 Protein/chemistry , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , MAP Kinase Signaling System , Mice , Models, Biological , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Protein Conformation , Receptor, IGF Type 1 , Receptors, Somatomedin/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Xenograft Model Antitumor Assays
14.
Nature ; 516(7529): 116-20, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25470042

ABSTRACT

The TRIM37 (also known as MUL) gene is located in the 17q23 chromosomal region, which is amplified in up to ∼ 40% of breast cancers. TRIM37 contains a RING finger domain, a hallmark of E3 ubiquitin ligases, but its protein substrate(s) is unknown. Here we report that TRIM37 mono-ubiquitinates histone H2A, a chromatin modification associated with transcriptional repression. We find that in human breast cancer cell lines containing amplified 17q23, TRIM37 is upregulated and, reciprocally, the major H2A ubiquitin ligase RNF2 (also known as RING1B) is downregulated. Genome-wide chromatin immunoprecipitation (ChIP)-chip experiments in 17q23-amplified breast cancer cells identified many genes, including multiple tumour suppressors, whose promoters were bound by TRIM37 and enriched for ubiquitinated H2A. However, unlike RNF2, which is a subunit of polycomb repressive complex 1 (PRC1), we find that TRIM37 associates with polycomb repressive complex 2 (PRC2). TRIM37, PRC2 and PRC1 are co-bound to specific target genes, resulting in their transcriptional silencing. RNA-interference-mediated knockdown of TRIM37 results in loss of ubiquitinated H2A, dissociation of PRC1 and PRC2 from target promoters, and transcriptional reactivation of silenced genes. Knockdown of TRIM37 in human breast cancer cells containing amplified 17q23 substantially decreases tumour growth in mouse xenografts. Conversely, ectopic expression of TRIM37 renders non-transformed cells tumorigenic. Collectively, our results reveal TRIM37 as an oncogenic H2A ubiquitin ligase that is overexpressed in a subset of breast cancers and promotes transformation by facilitating silencing of tumour suppressors and other genes.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins/genetics , Polycomb Repressive Complex 1/genetics , Animals , Female , Gene Expression Profiling , Gene Knockdown Techniques , Gene Silencing , Heterografts , Histones/metabolism , Humans , MCF-7 Cells , Mice , NIH 3T3 Cells , Oncogene Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases
15.
Exp Mol Pathol ; 105(1): 41-49, 2018 08.
Article in English | MEDLINE | ID: mdl-29852185

ABSTRACT

KIT Proto-Oncogene Receptor Tyrosine Kinase (KIT) is a transmembrane receptor tyrosine kinase which plays an important role in regulation of cell proliferation, survival and migration. Interestingly, the role of c-KIT in malignant transformation seems to be highly tissue-specific and it can act either as an oncogene or tumor suppressor gene. Here we analyzed the expression of c-KIT in normal breast tissues and tissues from different stages encompassing major steps of breast tumor development. Our study showed, that the c-KIT protein expression is gradually lost during the process of breast tissue transformation. The analysis of previously published datasets revealed that c-KIT expression in breast malignancies was downregulated at mRNA level. Because sequencing studies did not identify any recurrent mutations or copy number alterations, we proposed a potential epigenetic mechanism for the downregulation of c-KIT expression. In-silico analysis of the KIT promoter revealed the presence of CpG islands, therefore we performed bisulfite sequencing of normal breast epithelial tissues as well as breast tumor samples. We found, that KIT promoter is hypermethylated in breast tumors compared to normal breast tissues. Furthermore, treatment of breast cancer cell lines, that lack the expression of c-KIT, with methyltransferase inhibitor 5-Azacytidine (5Aza-2dC) resulted in increased expression of c-KIT mRNA. Collectively, our studies demonstrate that c-KIT expression is epigenetically downregulated during breast epithelium transformation and cancer development via KIT promoter hypermethylation.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Cell Transformation, Neoplastic , DNA Methylation , Proto-Oncogene Proteins c-kit/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Epigenesis, Genetic , Epithelium/metabolism , Epithelium/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Promoter Regions, Genetic , Proto-Oncogene Mas , Proto-Oncogene Proteins c-kit/metabolism
16.
Mol Cell ; 36(2): 219-30, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19854132

ABSTRACT

The RASSF1A tumor suppressor gene is epigenetically silenced in a variety of cancers. Here, we perform a genome-wide human shRNA screen and find that epigenetic silencing of RASSF1A requires the homeobox protein HOXB3. We show that HOXB3 binds to the DNA methyltransferase DNMT3B gene and increases its expression. DNMT3B, in turn, is recruited to the RASSF1A promoter, resulting in hypermethylation and silencing of RASSF1A expression. DNMT3B recruitment is facilitated through interactions with Polycomb repressor complex 2 and MYC, which is bound to the RASSF1A promoter. Mouse xenograft experiments indicate that the oncogenic activity of HOXB3 is due, at least in part, to epigenetic silencing of RASSF1A. Expression analysis in human lung adenocarcinoma samples reveals that RASSF1A silencing strongly correlates with overexpression of HOXB3 and DNMT3B. Analysis of human cancer cell lines indicates that the RASSF1A epigenetic silencing mechanism described here may be common in diverse cancer types.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Silencing , Homeodomain Proteins/metabolism , Tumor Suppressor Proteins/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Polycomb-Group Proteins , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Small Interfering/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , DNA Methyltransferase 3B
17.
Nucleic Acids Res ; 43(1): 259-71, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25488815

ABSTRACT

Serine/threonine kinase 11 (STK11, also known as LKB1) functions as a tumor suppressor in many human cancers. However, paradoxically loss of LKB1 in mouse embryonic fibroblast results in resistance to oncogene-induced transformation. Therefore, it is unclear why loss of LKB1 leads to increased predisposition to develop a wide variety of cancers. Here, we show that LKB1 protects cells from genotoxic stress. Cells lacking LKB1 display increased sensitivity to irradiation, accumulates more DNA double-strand breaks, display defective homology-directed DNA repair (HDR) and exhibit increased mutation rate, compared with that of LKB1-expressing cells. Conversely, the ectopic expression of LKB1 in cells lacking LKB1 protects them against genotoxic stress-induced DNA damage and prevents the accumulation of mutations. We find that LKB1 post-transcriptionally stimulates HDR gene BRCA1 expression by inhibiting the cytoplasmic localization of the RNA-binding protein, HU antigen R, in an AMP kinase-dependent manner and stabilizes BRCA1 mRNA. Cells lacking BRCA1 similar to the cell lacking LKB1 display increased genomic instability and ectopic expression of BRCA1 rescues LKB1 loss-induced sensitivity to genotoxic stress. Collectively, our results demonstrate that LKB1 is a crucial regulator of genome integrity and reveal a novel mechanism for LKB1-mediated tumor suppression with direct therapeutic implications for cancer prevention.


Subject(s)
BRCA1 Protein/genetics , Protein Serine-Threonine Kinases/physiology , Recombinational DNA Repair , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Animals , BRCA1 Protein/metabolism , Cell Cycle , Cell Line , DNA Breaks, Double-Stranded , DNA Damage , ELAV Proteins/metabolism , Genome , Humans , Mice , Mutation , RNA Stability , RNA, Messenger/metabolism
18.
Proc Natl Acad Sci U S A ; 111(35): 12591-8, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136103

ABSTRACT

X-chromosome inactivation (XCI), the random transcriptional silencing of one X chromosome in somatic cells of female mammals, is a mechanism that ensures equal expression of X-linked genes in both sexes. XCI is initiated in cis by the noncoding Xist RNA, which coats the inactive X chromosome (Xi) from which it is produced. However, trans-acting factors that mediate XCI remain largely unknown. Here, we perform a large-scale RNA interference screen to identify trans-acting XCI factors (XCIFs) that comprise regulators of cell signaling and transcription, including the DNA methyltransferase, DNMT1. The expression pattern of the XCIFs explains the selective onset of XCI following differentiation. The XCIFs function, at least in part, by promoting expression and/or localization of Xist to the Xi. Surprisingly, we find that DNMT1, which is generally a transcriptional repressor, is an activator of Xist transcription. Small-molecule inhibitors of two of the XCIFs can reversibly reactivate the Xi, which has implications for treatment of Rett syndrome and other dominant X-linked diseases. A homozygous mouse knockout of one of the XCIFs, stanniocalcin 1 (STC1), has an expected XCI defect but surprisingly is phenotypically normal. Remarkably, X-linked genes are not overexpressed in female Stc1(-/-) mice, revealing the existence of a mechanism(s) that can compensate for a persistent XCI deficiency to regulate X-linked gene expression.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Glycoproteins/genetics , Methyl-CpG-Binding Protein 2/genetics , RNA, Long Noncoding/genetics , Rett Syndrome/genetics , X Chromosome Inactivation/genetics , Animals , Cerebral Cortex/cytology , Chromones/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Embryonic Stem Cells/physiology , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/genetics , Female , Fibroblasts/cytology , Fibroblasts/physiology , Gene Library , Genetic Therapy/methods , Humans , Mammals , Mice , Mice, Knockout , Morpholines/pharmacology , Neurons/cytology , Neurons/physiology , Pyrazoles/pharmacology , RNA, Small Interfering/genetics , Rett Syndrome/therapy , Sulfonamides/pharmacology , Transcriptome , X Chromosome Inactivation/drug effects
19.
Proc Natl Acad Sci U S A ; 111(30): E3062-71, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25024194

ABSTRACT

Tumor suppressor p53 plays an important role in mediating growth inhibition upon telomere dysfunction. Here, we show that loss of the p53 target gene cyclin-dependent kinase inhibitor 1A (CDKN1A, also known as p21(WAF1/CIP1)) increases apoptosis induction following telomerase inhibition in a variety of cancer cell lines and mouse xenografts. This effect is highly specific to p21, as loss of other checkpoint proteins and CDK inhibitors did not affect apoptosis. In telomerase, inhibited cell loss of p21 leads to E2F1- and p53-mediated transcriptional activation of p53-upregulated modulator of apoptosis, resulting in increased apoptosis. Combined genetic or pharmacological inhibition of telomerase and p21 synergistically suppresses tumor growth. Furthermore, we demonstrate that simultaneous inhibition of telomerase and p21 also suppresses growth of tumors containing mutant p53 following pharmacological restoration of p53 activity. Collectively, our results establish that inactivation of p21 leads to increased apoptosis upon telomerase inhibition and thus identify a genetic vulnerability that can be exploited to treat many human cancers containing either wild-type or mutant p53.


Subject(s)
Apoptosis , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Neoplasms, Experimental/metabolism , Telomerase/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Mice , Mice, Nude , Mutation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Telomerase/genetics , Telomerase/metabolism , Tumor Suppressor Protein p53/genetics
20.
Nature ; 459(7247): 722-5, 2009 Jun 04.
Article in English | MEDLINE | ID: mdl-19412162

ABSTRACT

In response to DNA damage, eukaryotic cells initiate a complex signalling pathway, termed the DNA damage response (DDR), which coordinates cell cycle arrest with DNA repair. Studies have shown that oncogene-induced senescence, which provides a barrier to tumour development, involves activation of the DDR. Using a genome-wide RNA interference (RNAi) screen, we have identified 17 factors required for oncogenic BRAF to induce senescence in primary fibroblasts and melanocytes. One of these factors is an F-box protein, FBXO31, a candidate tumour suppressor encoded in 16q24.3, a region in which there is loss of heterozygosity in breast, ovarian, hepatocellular and prostate cancers. Here we study the cellular role of FBXO31, identify its target substrate and determine the basis for its growth inhibitory activity. We show that ectopic expression of FBXO31 acts through a proteasome-directed pathway to mediate the degradation of cyclin D1, an important regulator of progression from G1 to S phase, resulting in arrest in G1. Cyclin D1 degradation results from a direct interaction with FBXO31 and is dependent on the F-box motif of FBXO31 and phosphorylation of cyclin D1 at Thr 286, which is known to be required for cyclin D1 proteolysis. The involvement of the DDR in oncogene-induced senescence prompted us to investigate the role of FBXO31 in DNA repair. We find that DNA damage induced by gamma-irradiation results in increased FBXO31 levels, which requires phosphorylation of FBXO31 by the DDR-initiating kinase ATM. RNAi-mediated knockdown of FBXO31 prevents cells from undergoing efficient arrest in G1 after gamma-irradiation and markedly increases sensitivity to DNA damage. Finally, we show that a variety of DNA damaging agents all result in a large increase in FBXO31 levels, indicating that induction of FBXO31 is a general response to genotoxic stress. Our results reveal FBXO31 as a regulator of the G1/S transition that is specifically required for DNA damage-induced growth arrest.


Subject(s)
Cyclin D1/metabolism , DNA Damage/genetics , F-Box Proteins/metabolism , G1 Phase/physiology , Tumor Suppressor Proteins/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , DNA Damage/drug effects , DNA-Binding Proteins/metabolism , Humans , Melanoma/genetics , Melanoma/physiopathology , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcriptional Activation , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL