Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Mol Cancer Res ; 22(2): 181-196, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37889141

RESUMO

Irradiation (IR) is a highly effective cancer therapy; however, IR damage to tumor-adjacent healthy tissues can result in significant comorbidities and potentially limit the course of therapy. We have previously shown that protein kinase C delta (PKCδ) is required for IR-induced apoptosis and that inhibition of PKCδ activity provides radioprotection in vivo. Here we show that PKCδ regulates histone modification, chromatin accessibility, and double-stranded break (DSB) repair through a mechanism that requires Sirtuin 6 (SIRT6). Overexpression of PKCδ promotes genomic instability and increases DNA damage and apoptosis. Conversely, depletion of PKCδ increases DNA repair via nonhomologous end joining (NHEJ) and homologous recombination (HR) as evidenced by increased formation of DNA damage foci, increased expression of DNA repair proteins, and increased repair of NHEJ and HR fluorescent reporter constructs. Nuclease sensitivity indicates that PKCδ depletion is associated with more open chromatin, while overexpression of PKCδ reduces chromatin accessibility. Epiproteome analysis reveals increased chromatin associated H3K36me2 in PKCδ-depleted cells which is accompanied by chromatin disassociation of KDM2A. We identify SIRT6 as a downstream mediator of PKCδ. PKCδ-depleted cells have increased SIRT6 expression, and depletion of SIRT6 reverses changes in chromatin accessibility, histone modification and DSB repair in PKCδ-depleted cells. Furthermore, depletion of SIRT6 reverses radioprotection in PKCδ-depleted cells. Our studies describe a novel pathway whereby PKCδ orchestrates SIRT6-dependent changes in chromatin accessibility to regulate DNA repair, and define a mechanism for regulation of radiation-induced apoptosis by PKCδ. IMPLICATIONS: PKCδ controls sensitivity to irradiation by regulating DNA repair.


Assuntos
Proteínas F-Box , Sirtuínas , Humanos , Montagem e Desmontagem da Cromatina , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Cromatina/genética , Sirtuínas/genética , Sirtuínas/metabolismo , Reparo do DNA por Junção de Extremidades , Proteínas F-Box/genética , Histona Desmetilases com o Domínio Jumonji/genética
2.
J Biol Chem ; 299(4): 104601, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36907441

RESUMO

Plant homeodomain (PHD) fingers are structurally conserved zinc fingers that selectively bind unmodified or methylated at lysine 4 histone H3 tails. This binding stabilizes transcription factors and chromatin-modifying proteins at specific genomic sites, which is required for vital cellular processes, including gene expression and DNA repair. Several PHD fingers have recently been shown to recognize other regions of H3 or histone H4. In this review, we detail molecular mechanisms and structural features of the noncanonical histone recognition, discuss biological implications of the atypical interactions, highlight therapeutic potential of PHD fingers, and compare inhibition strategies.


Assuntos
Histonas , Dedos de Zinco PHD , Proteínas de Ligação a DNA/metabolismo , Histonas/química , Histonas/metabolismo , Ligação Proteica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Camundongos , Neoplasias/genética , Neoplasias/fisiopatologia
3.
Cells ; 12(6)2023 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-36980293

RESUMO

(1) Background: Metal homeostasis is an important part of cellular programs and is disrupted when cells are exposed to carcinogenic heavy metals. Metal response is mediated by the metal response element transcription factor MTF-1. However, where MTF-1 binds and how that binding changes in response to heavy metals, such as cadmium, remains unknown. (2) Methods: To investigate the effects of prolonged cadmium exposure on the genomic distribution of MTF-1, we performed MTF-1 CUT&RUN, RNA-seq and ATAC-seq on control and cadmium-resistant cells. (3) Results: Changes in MTF-1 binding primarily occur distal to the transcription start sight. Newly occupied MTF-1 sites are enriched for FOS/JUN DNA binding motifs, while regions that lose MTF-1 binding in cadmium are enriched for the FOX transcription factor family member DNA binding sites. (4) Conclusions: Relocalization of MTF-1 to new genomic loci does not alter the accessibility of these locations. Our results support a model whereby MTF-1 is relocalized to accessible FOS/JUN-bound genomic locations in response to cadmium.


Assuntos
Cádmio , Proteínas de Ligação a DNA , Metais Pesados , Fatores de Transcrição , Feminino , Humanos , Neoplasias da Mama/metabolismo , Cádmio/metabolismo , Cádmio/farmacologia , Cádmio/toxicidade , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Genômica , Metais Pesados/metabolismo , Metais Pesados/farmacologia , Metais Pesados/toxicidade , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Zinco/metabolismo , Fator MTF-1 de Transcrição
4.
Cancers (Basel) ; 14(9)2022 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-35565439

RESUMO

Extrachromosomal circular DNA has emerged as a frequent genomic alteration in tumors. High numbers of circular DNAs correspond to poor prognosis suggesting an important function in tumor biology. However, despite mounting evidence supporting the importance of circular DNA, little is known about their production, maintenance, or selection. To provide insight into these processes, we analyzed circular DNA elements computationally identified in 355 TCGA tumors spanning 22 tumor types. Circular DNAs originated from common genomic loci irrespective of cancer type. Genes found in circularized genomic regions were more likely to be expressed and were enriched in cancer-related pathways. Finally, in support of a model for circle generation through either a homology or microhomology-mediated process, circles exhibit homology near their breakpoint. These breakpoints are also enriched in specific DNA motifs. Our analysis supports a model where gene-containing circles emerge from common, highly transcribed regions through a homology-mediated process.

5.
Cell Rep ; 38(5): 110323, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35108532

RESUMO

Rhabdomyosarcoma (RMS) is a pediatric muscle sarcoma characterized by expression of the myogenic lineage transcription factors (TFs) MYOD1 and MYOG. Despite high expression of these TFs, RMS cells fail to terminally differentiate, suggesting the presence of factors that alter their functions. Here, we demonstrate that the developmental TF SIX1 is highly expressed in RMS and critical for maintaining a muscle progenitor-like state. SIX1 loss induces differentiation of RMS cells into myotube-like cells and impedes tumor growth in vivo. We show that SIX1 maintains the RMS undifferentiated state by controlling enhancer activity and MYOD1 occupancy at loci more permissive to tumor growth over muscle differentiation. Finally, we demonstrate that a gene signature derived from SIX1 loss correlates with differentiation status and predicts RMS progression in human disease. Our findings demonstrate a master regulatory role of SIX1 in repression of RMS differentiation via genome-wide alterations in MYOD1 and MYOG-mediated transcription.


Assuntos
Proteínas de Homeodomínio/metabolismo , Desenvolvimento Muscular/genética , Rabdomiossarcoma/genética , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Diferenciação Celular/genética , Regulação Neoplásica da Expressão Gênica/genética , Camundongos , Desenvolvimento Muscular/fisiologia , Proteína MyoD/metabolismo , Miogenina/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma Embrionário , Peixe-Zebra
6.
Nat Commun ; 11(1): 5466, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33122719

RESUMO

Human Microrchidia 4 (MORC4) is associated with acute and chronic pancreatitis, inflammatory disorders and cancer but it remains largely uncharacterized. Here, we describe the structure-function relationship of MORC4 and define the molecular mechanism for MORC4 activation. Enzymatic and binding assays reveal that MORC4 has ATPase activity, which is dependent on DNA-binding functions of both the ATPase domain and CW domain of MORC4. The crystal structure of the ATPaseCW cassette of MORC4 and mutagenesis studies show that the DNA-binding site and the histone/ATPase binding site of CW are located on the opposite sides of the domain. The ATPase and CW domains cooperate in binding of MORC4 to the nucleosome core particle (NCP), enhancing the DNA wrapping around the histone core and impeding binding of DNA-associated proteins, such as transcription factors, to the NCP. In cells, MORC4 mediates formation of nuclear bodies in the nucleus and has a role in the progression of S-phase of the cell cycle, and both these functions require CW and catalytic activity of MORC4. Our findings highlight the mechanism for MORC4 activation, which is distinctly different from the mechanisms of action observed in other MORC family members.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas Nucleares , Sítios de Ligação , Ciclo Celular , Cristalografia por Raios X , DNA/metabolismo , Células HEK293 , Histonas/metabolismo , Humanos , Corpos de Inclusão Intranuclear/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Nucleossomos/metabolismo , Ligação Proteica , Domínios Proteicos/fisiologia , Pontos de Checagem da Fase S do Ciclo Celular , Espectrometria de Fluorescência , Fatores de Transcrição/metabolismo , Nucleases de Dedos de Zinco/química , Nucleases de Dedos de Zinco/metabolismo
7.
Mol Oncol ; 14(10): 2471-2486, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32697014

RESUMO

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children and young adults. RMS exists as two major disease subtypes, oncofusion-negative RMS (FN-RMS) and oncofusion-positive RMS (FP-RMS). FP-RMS is characterized by recurrent PAX3/7-FOXO1 driver oncofusions and is a biologically and clinically aggressive disease. Recent studies have revealed FP-RMS to have a strong epigenetic basis. Epigenetic mechanisms represent potential new therapeutic vulnerabilities in FP-RMS, but their complex details remain to be defined. We previously identified a new disease-promoting epigenetic axis in RMS, involving the chromatin factor KDM3A and the Ets1 transcription factor. In the present study, we define the KDM3A and Ets1 FP-RMS transcriptomes and show that these interface with the recently characterized PAX3/FOXO1-driven gene expression program. KDM3A and Ets1 positively control numerous known and candidate novel PAX3/FOXO1-induced RMS-promoting genes, including subsets under control of PAX3/FOXO1-associated superenhancers (SE), such as MEST. Interestingly, KDM3A and Ets1 also positively control a number of known and candidate novel FP-RMS-promoting, but not PAX3/FOXO1-dependent, genes. Epistatically, Ets1 is downstream of, and exerts disease-promoting effects similar to, both KDM3A and PAX3/FOXO1. MEST also manifests disease-promoting properties in FP-RMS, and KDM3A and Ets1 each impacts activation of the PAX3/FOXO1-associated MEST SE. Taken together, our studies show that the KDM3A/Ets1 epigenetic axis plays an important role in disease promotion in FP-RMS, and provide insight into potential new ways to target aggressive phenotypes in this disease.


Assuntos
Epigênese Genética , Proteína Forkhead Box O1/metabolismo , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Fator de Transcrição PAX3/metabolismo , Proteína Proto-Oncogênica c-ets-1/metabolismo , Rabdomiossarcoma/genética , Linhagem Celular Tumoral , Elementos Facilitadores Genéticos/genética , Humanos , Fenótipo , Regiões Promotoras Genéticas/genética , Transcriptoma/genética
8.
Genes Cancer ; 11(1-2): 53-65, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32577157

RESUMO

Rhabdomyosarcoma (RMS) is the most common soft tissue malignancy of childhood. RMS exists as two major disease subtypes, with oncofusion-positive RMS (FP-RMS) typically carrying a worse prognosis than oncofusion-negative RMS (FN-RMS), in part due to higher propensity for metastasis. Epigenetic mechanisms have recently emerged as critical players in the pathogenesis of pediatric cancers, as well as potential new therapeutic vulnerabilities. Herein, we show that the epigenetic regulator KDM3A, a member of the Jumonji-domain histone demethylase (JHDM) family, is overexpressed, potently promotes colony formation and transendothelial invasion, and activates the expression of genes involved in cell growth, migration and metastasis, in both FN-RMS and FP-RMS. In mechanistic studies, we demonstrate that both RMS subtypes utilize a KDM3A/Ets1/MCAM disease-promoting axis recently discovered in Ewing Sarcoma, another aggressive pediatric cancer of distinct cellular and molecular origin. We further show that KDM3A depletion in FP-RMS cells inhibits both tumor growth and metastasis in vivo, and that RMS cells are highly sensitive to colony growth inhibition by the pan-JHDM inhibitor JIB-04. Together, our studies reveal an important role for the KDM3A/Ets1/MCAM axis in pediatric sarcomas of distinct cellular and molecular ontogeny, and identify new targetable vulnerabilities in RMS.

9.
Nucleic Acids Res ; 48(10): e58, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32239215

RESUMO

Changes in gene copy number contribute to genomic instability, the onset and progression of cancer, developmental abnormalities and adaptive potential. The origins of gene amplifications have remained elusive; however, DNA rereplication has been implicated as a source of gene amplifications. The inability to determine which sequences are rereplicated and under what conditions have made it difficult to determine the validity of the proposed models. Here we present Rerep-Seq, a technique that selectively enriches for rereplicated DNA in preparation for analysis by DNA sequencing that can be applied to any species. We validated Rerep-Seq by simulating DNA rereplication in yeast and human cells. Using Rerep-Seq, we demonstrate that rereplication induced in Saccharomyces cerevisiae by deregulated origin licensing is non-random and defined by broad domains that span multiple replication origins and topological boundaries.


Assuntos
Replicação do DNA , Análise de Sequência de DNA/métodos , Linhagem Celular , Humanos , Saccharomyces cerevisiae/genética
10.
Cancer Discov ; 10(2): 179-181, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32024681

RESUMO

Response rates to immune checkpoint blockade (ICB) in KRAS-mutant lung adenocarcinoma remain poor. In this issue of Cancer Discovery, Li and colleagues report an in vivo CRISPR screen of epigenetic regulators of the tumor immune microenvironment that uncovers Asf1a as a tumor-intrinsic suppressor of ICB through suppression of GM-CSF expression.See related article by Li et al., p. 270.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Proteínas de Ciclo Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Epigênese Genética , Humanos , Imunoterapia , Chaperonas Moleculares , Proteínas Proto-Oncogênicas p21(ras) , Microambiente Tumoral
11.
Arch Pathol Lab Med ; 143(2): 197-205, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30168727

RESUMO

CONTEXT.­: Duchenne muscular dystrophy is a rare, progressive, and fatal neuromuscular disease caused by dystrophin protein loss. Common investigational treatment approaches aim at increasing dystrophin expression in diseased muscle. Some clinical trials include assessments of novel dystrophin production as a surrogate biomarker of efficacy, which may predict a clinical benefit from treatment. OBJECTIVES.­: To establish an immunofluorescent scanning and digital image analysis workflow that provides an objective approach for staining intensity assessment of the immunofluorescence dystrophin labeling and determination of the percentage of biomarker-positive fibers in muscle cryosections. DESIGN.­: Optimal and repeatable digital image capture was achieved by a rigorously qualified fluorescent scanning process. After scanning qualification, the MuscleMap (Flagship Biosciences, Westminster, Colorado) algorithm was validated by comparing high-power microscopic field total and dystrophin-positive fiber counts obtained by trained pathologists to data derived by MuscleMap. Next, the algorithm was tested on whole-slide images of immunofluorescent-labeled muscle sections from Duchenne muscular dystrophy, Becker muscular dystrophy, and control patients. RESULTS.­: When used under the guidance of a trained pathologist, the digital image analysis tool met predefined validation criteria and demonstrated functional and statistical equivalence with manual assessment. This work is the first, to our knowledge, to qualify and validate immunofluorescent scanning and digital tissue image-analysis workflow, respectively, with the rigor required to support the clinical trial environments. CONCLUSIONS.­: MuscleMap enables analysis of all fibers within an entire muscle biopsy section and provides data on a fiber-by-fiber basis. This will allow future clinical trials to objectively investigate myofibers' dystrophin expression at a greater level of consistency and detail.


Assuntos
Distrofina/análise , Interpretação de Imagem Assistida por Computador/métodos , Distrofia Muscular de Duchenne/diagnóstico , Adolescente , Biópsia , Criança , Pré-Escolar , Feminino , Secções Congeladas , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/patologia
12.
Cell ; 174(4): 803-817.e16, 2018 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-30057114

RESUMO

Acquired chromosomal DNA amplifications are features of many tumors. Although overexpression and stabilization of the histone H3 lysine 9/36 (H3K9/36) tri-demethylase KDM4A generates transient site-specific copy number gains (TSSGs), additional mechanisms directly controlling site-specific DNA copy gains are not well defined. In this study, we uncover a collection of H3K4-modifying chromatin regulators that function with H3K9 and H3K36 regulators to orchestrate TSSGs. Specifically, the H3K4 tri-demethylase KDM5A and specific COMPASS/KMT2 H3K4 methyltransferases modulate different TSSG loci through H3K4 methylation states and KDM4A recruitment. Furthermore, a distinct chromatin modifier network, MLL1-KDM4B-KDM5B, controls copy number regulation at a specific genomic locus in a KDM4A-independent manner. These pathways comprise an epigenetic addressing system for defining site-specific DNA rereplication and amplifications.


Assuntos
Cromatina/metabolismo , Variações do Número de Cópias de DNA , Metilação de DNA , Histonas/metabolismo , Lisina/metabolismo , Proteína 2 de Ligação ao Retinoblastoma/metabolismo , Ciclo Celular , Células HEK293 , Humanos , Proteína 2 de Ligação ao Retinoblastoma/genética
13.
Lab Invest ; 96(12): 1327-1336, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27775692

RESUMO

Tissue image analysis (tIA) is emerging as a powerful tool for quantifying biomarker expression and distribution in complex diseases and tissues. Pancreatic ductal adenocarcinoma (PDAC) develops in a highly complex and heterogeneous tissue environment and, generally, has a very poor prognosis. Early detection of PDAC is confounded by limited knowledge of the pre-neoplastic disease stages and limited methods to quantitatively assess disease heterogeneity. We sought to develop a tIA approach to assess the most common PDAC precursor lesions, pancreatic intraepithelial neoplasia (PanIN), in tissues from KrasLSL-G12D/+; Trp53LSL-R172H/+; Pdx-Cre (KPC) mice, a validated model of PDAC development. tIA profiling of training regions of PanIN and tumor microenvironment (TME) cells was utilized to guide identification of PanIN/TME tissue compartment stratification criteria. A custom CellMap algorithm implementing these criteria was applied to whole-slide images of KPC mice pancreata sections to quantify p53 and Ki-67 biomarker staining in each tissue compartment as a proof-of-concept for the algorithm platform. The algorithm robustly identified a higher percentage of p53-positive cells in PanIN lesions relative to the TME, whereas no difference was observed for Ki-67. Ki-67 expression was also quantified in a human pancreatic tissue sample available to demonstrate the translatability of the CellMap algorithm to human samples. Together, our data demonstrated the utility of CellMap to enable objective and quantitative assessments, across entire tissue sections, of PDAC precursor lesions in preclinical and clinical models of this disease to support efforts leading to novel insights into disease progression, diagnostic markers, and potential therapeutic targets.


Assuntos
Adenocarcinoma in Situ/diagnóstico , Carcinoma Ductal Pancreático/diagnóstico , Pâncreas/patologia , Neoplasias Pancreáticas/diagnóstico , Lesões Pré-Cancerosas/diagnóstico , Proteína Supressora de Tumor p53/metabolismo , Adenocarcinoma in Situ/diagnóstico por imagem , Adenocarcinoma in Situ/metabolismo , Adenocarcinoma in Situ/patologia , Algoritmos , Animais , Automação Laboratorial , Carcinoma Ductal Pancreático/diagnóstico por imagem , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Cruzamentos Genéticos , Modelos Animais de Doenças , Detecção Precoce de Câncer/métodos , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Camundongos Mutantes , Camundongos Transgênicos , Pâncreas/metabolismo , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Lesões Pré-Cancerosas/diagnóstico por imagem , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Software , Organismos Livres de Patógenos Específicos , Bancos de Tecidos , Ultrassonografia
14.
J Biol Chem ; 291(10): 4862-71, 2016 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-26755726

RESUMO

Intra-tumor copy number heterogeneity is commonly observed in cancer; however, the molecular mechanisms that contribute to heterogeneity remain poorly understood. Up-regulation of the histone demethylase KDM4A promotes transient site-specific copy gain (TSSG) in cells; therefore, uncovering how KDM4A levels are controlled is important for understanding the regulation of copy number heterogeneity. Here, we demonstrate that KDM4A is regulated by hsa-mir-23a-3p, hsa-mir-23b-3p, and hsa-mir-137. Altering expression of these microRNAs (miRNAs) regulates KDM4A-dependent TSSG. miRNA inhibition promoted copy gains and increased expression of the drug-resistant oncogene CKS1B, which was further substantiated in primary breast tumors. Consistent with increased CKS1B expression, miRNA inhibition reduced breast cancer cell sensitivity to cisplatin. Our data identify these miRNAs as regulators of TSSG and copy gains of a drug resistance gene.


Assuntos
Neoplasias da Mama/genética , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/genética , MicroRNAs/genética , Quinases relacionadas a CDC2 e CDC28/genética , Quinases relacionadas a CDC2 e CDC28/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos
15.
Cancer Discov ; 5(12): 1241-3, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26637658

RESUMO

RNF2/RING1B is amplified and overexpressed in numerous tumors and contributes to tumorigenicity; however, the biologic importance is poorly understood. Surprisingly, the role of RNF2 in tumorigenesis and invasion can be separated into catalytically independent and catalytically dependent processes.


Assuntos
Complexo Repressor Polycomb 1 , Ubiquitina-Proteína Ligases , Humanos , Melanoma
16.
Nat Commun ; 6: 8257, 2015 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-26394836

RESUMO

Expression of the p53-inducible antiproliferative gene BTG2 is suppressed in many cancers in the absence of inactivating gene mutations, suggesting alternative mechanisms of silencing. Using a shRNA screen targeting 43 histone lysine methyltransferases (KMTs), we show that SETD1A suppresses BTG2 expression through its induction of several BTG2-targeting miRNAs. This indirect but highly specific mechanism, by which a chromatin regulator that mediates transcriptional activating marks can lead to the downregulation of a critical effector gene, is shared with multiple genes in the p53 pathway. Through such miRNA-dependent effects, SETD1A regulates cell cycle progression in vitro and modulates tumorigenesis in mouse xenograft models. Together, these observations help explain the remarkably specific genetic consequences associated with alterations in generic chromatin modulators in cancer.


Assuntos
Ciclo Celular , Regulação Neoplásica da Expressão Gênica , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas Imediatamente Precoces/metabolismo , MicroRNAs/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Carcinogênese , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Camundongos Nus , Neoplasias Experimentais , Regiões Promotoras Genéticas , Proteína Supressora de Tumor p53/metabolismo
17.
Genes Dev ; 29(10): 1018-31, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25995187

RESUMO

Copy number heterogeneity is a prominent feature within tumors. The molecular basis for this heterogeneity remains poorly characterized. Here, we demonstrate that hypoxia induces transient site-specific copy gains (TSSGs) in primary, nontransformed, and transformed human cells. Hypoxia-driven copy gains are not dependent on HIF1α or HIF2α; however, they are dependent on the KDM4A histone demethylase and are blocked by inhibition of KDM4A with a small molecule or the natural metabolite succinate. Furthermore, this response is conserved at a syntenic region in zebrafish cells. Regions with site-specific copy gain are also enriched for amplifications in hypoxic primary tumors. These tumors exhibited amplification and overexpression of the drug resistance gene CKS1B, which we recapitulated in hypoxic breast cancer cells. Our results demonstrate that hypoxia provides a biological stimulus to create transient site-specific copy alterations that could result in heterogeneity within tumors and cell populations. These findings have major implications in our understanding of copy number heterogeneity and the emergence of drug resistance genes in cancer.


Assuntos
Hipóxia Celular/fisiologia , Variações do Número de Cópias de DNA/genética , Regulação da Expressão Gênica , Animais , Quinases relacionadas a CDC2 e CDC28/genética , Hipóxia Celular/genética , Linhagem Celular , Proliferação de Células , Células Cultivadas , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Peixe-Zebra
18.
Cancer Discov ; 5(3): 255-63, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25564516

RESUMO

UNLABELLED: Chromatin-modifying enzymes are predominantly nuclear; however, these factors are also localized to the cytoplasm, and very little is known about their role in this compartment. In this report, we reveal a non-chromatin-linked role for the lysine-specific demethylase KDM4A. We demonstrate that KDM4A interacts with the translation initiation complex and affects the distribution of translation initiation factors within polysome fractions. Furthermore, KDM4A depletion reduced protein synthesis and enhanced the protein synthesis suppression observed with mTOR inhibitors, which paralleled an increased sensitivity to these drugs. Finally, we demonstrate that JIB-04, a JmjC demethylase inhibitor, suppresses translation initiation and enhances mTOR inhibitor sensitivity. These data highlight an unexpected cytoplasmic role for KDM4A in regulating protein synthesis and suggest novel potential therapeutic applications for this class of enzyme. SIGNIFICANCE: This report documents an unexpected cytoplasmic role for the lysine demethylase KDM4A. We demonstrate that KDM4A interacts with the translation initiation machinery, regulates protein synthesis and, upon coinhibition with mTOR inhibitors, enhances the translation suppression and cell sensitivity to these therapeutics.


Assuntos
Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Lisina/metabolismo , Biossíntese de Proteínas , Aminopiridinas/farmacologia , Resistência a Medicamentos/genética , Humanos , Hidrazonas/farmacologia , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Metilação , Iniciação Traducional da Cadeia Peptídica , Fatores de Iniciação de Peptídeos/metabolismo , Ligação Proteica , Biossíntese de Proteínas/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Ribossômicas/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores
19.
Cancer Discov ; 5(3): 245-54, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25564517

RESUMO

UNLABELLED: SNPs occur within chromatin-modulating factors; however, little is known about how these variants within the coding sequence affect cancer progression or treatment. Therefore, there is a need to establish their biochemical and/or molecular contribution, their use in subclassifying patients, and their impact on therapeutic response. In this report, we demonstrate that coding SNP-A482 within the lysine tridemethylase gene KDM4A/JMJD2A has different allelic frequencies across ethnic populations, associates with differential outcome in patients with non-small cell lung cancer (NSCLC), and promotes KDM4A protein turnover. Using an unbiased drug screen against 87 preclinical and clinical compounds, we demonstrate that homozygous SNP-A482 cells have increased mTOR inhibitor sensitivity. mTOR inhibitors significantly reduce SNP-A482 protein levels, which parallels the increased drug sensitivity observed with KDM4A depletion. Our data emphasize the importance of using variant status as candidate biomarkers and highlight the importance of studying SNPs in chromatin modifiers to achieve better targeted therapy. SIGNIFICANCE: This report documents the first coding SNP within a lysine demethylase that associates with worse outcome in patients with NSCLC. We demonstrate that this coding SNP alters the protein turnover and associates with increased mTOR inhibitor sensitivity, which identifies a candidate biomarker for mTOR inhibitor therapy and a therapeutic target for combination therapy.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Histona Desmetilases com o Domínio Jumonji/genética , Lisina/genética , Fases de Leitura Aberta , Polimorfismo de Nucleotídeo Único , Inibidores de Proteínas Quinases/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Alelos , Linhagem Celular , Linhagem Celular Tumoral , Análise Mutacional de DNA , Frequência do Gene , Humanos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/mortalidade , Prognóstico , Ubiquitinação
20.
Sci Transl Med ; 5(212): 212ra162, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-24259050

RESUMO

MicroRNAs (miRNAs) regulate many aspects of human biology. They target mRNAs for translational repression or degradation through base pairing with 3' untranslated regions, primarily via seed sequences (nucleotides 2 to 8 in the mature miRNA sequence). A number of individual miRNAs and miRNA families share seed sequences and targets, but differ in the sequences outside of the seed. miRNAs have been implicated in the etiology of a wide variety of human diseases and therefore represent promising therapeutic targets. However, potential redundancy of different miRNAs sharing the same seed sequence and the challenge of simultaneously targeting miRNAs that differ significantly in nonseed sequences complicate therapeutic targeting approaches. We recently demonstrated effective inhibition of entire miRNA families using seed-targeting 8-mer locked nucleic acid (LNA)-modified antimiRs in short-term experiments in mammalian cells and in mice. However, the long-term efficacy and safety of this approach in higher organisms, such as humans and nonhuman primates, have not been determined. We show that pharmacological inhibition of the miR-33 family, key regulators of cholesterol/lipid homeostasis, by a subcutaneously delivered 8-mer LNA-modified antimiR in obese and insulin-resistant nonhuman primates results in derepression of miR-33 targets, such as ABCA1, increases circulating high-density lipoprotein cholesterol, and is well tolerated over 108 days of treatment. These findings demonstrate the efficacy and safety of an 8-mer LNA-antimiR against an miRNA family in a nonhuman primate metabolic disease model, suggesting that this could be a feasible approach for therapeutic targeting of miRNA families sharing the same seed sequence in human diseases.


Assuntos
Inativação Gênica , MicroRNAs/antagonistas & inibidores , Animais , HDL-Colesterol/sangue , Feminino , Células Hep G2 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Primatas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA