Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell ; 167(1): 171-186.e15, 2016 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-27641501

RESUMO

While acute myeloid leukemia (AML) comprises many disparate genetic subtypes, one shared hallmark is the arrest of leukemic myeloblasts at an immature and self-renewing stage of development. Therapies that overcome differentiation arrest represent a powerful treatment strategy. We leveraged the observation that the majority of AML, despite their genetically heterogeneity, share in the expression of HoxA9, a gene normally downregulated during myeloid differentiation. Using a conditional HoxA9 model system, we performed a high-throughput phenotypic screen and defined compounds that overcame differentiation blockade. Target identification led to the unanticipated discovery that inhibition of the enzyme dihydroorotate dehydrogenase (DHODH) enables myeloid differentiation in human and mouse AML models. In vivo, DHODH inhibitors reduced leukemic cell burden, decreased levels of leukemia-initiating cells, and improved survival. These data demonstrate the role of DHODH as a metabolic regulator of differentiation and point to its inhibition as a strategy for overcoming differentiation blockade in AML.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Terapia de Alvo Molecular , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/antagonistas & inibidores , Animais , Antineoplásicos/química , Antineoplásicos/isolamento & purificação , Diferenciação Celular , Di-Hidro-Orotato Desidrogenase , Inibidores Enzimáticos/química , Inibidores Enzimáticos/isolamento & purificação , Ensaios de Triagem em Larga Escala , Proteínas de Homeodomínio/genética , Humanos , Leucemia Mieloide Aguda/genética , Camundongos , Células Mieloides/patologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Pirimidinas/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/isolamento & purificação , Bibliotecas de Moléculas Pequenas/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Proc Natl Acad Sci U S A ; 112(18): 5708-13, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25902495

RESUMO

Colony formation in soft agar is the gold-standard assay for cellular transformation in vitro, but it is unsuited for high-throughput screening. Here, we describe an assay for cellular transformation that involves growth in low attachment (GILA) conditions and is strongly correlated with the soft-agar assay. Using GILA, we describe high-throughput screens for drugs and genes that selectively inhibit or increase transformation, but not proliferation. Such molecules are unlikely to be found through conventional drug screening, and they include kinase inhibitors and drugs for noncancer diseases. In addition to known oncogenes, the genetic screen identifies genes that contribute to cellular transformation. Lastly, we demonstrate the ability of Food and Drug Administration-approved noncancer drugs to selectively kill ovarian cancer cells derived from patients with chemotherapy-resistant disease, suggesting this approach may provide useful information for personalized cancer treatment.


Assuntos
Ágar/química , Antineoplásicos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Neoplasias Ovarianas/tratamento farmacológico , Trifosfato de Adenosina/química , Antineoplásicos/química , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Transformação Celular Neoplásica , Química Farmacêutica , Desenho de Fármacos , Feminino , Fibroblastos/patologia , Citometria de Fluxo , Ensaios de Triagem em Larga Escala , Humanos , Fases de Leitura Aberta , Neoplasias Ovarianas/patologia
3.
Exp Cell Res ; 348(2): 123-131, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27646113

RESUMO

Cell migration is a central process in the development and maintenance of multicellular organisms. Tissue formation during embryonic development, wound healing, immune responses and invasive tumors all require the orchestrated movement of cells to specific locations. Histone demethylase proteins alter transcription by regulating the chromatin state at specific gene loci. FBXL10 is a conserved and ubiquitously expressed member of the JmjC domain-containing histone demethylase family and is implicated in the demethylation of H3K4me3 and H3K36me2 and thereby removing active chromatin marks. However, the physiological role of FBXL10 in vivo remains largely unknown. Therefore, we established an inducible gain of function model to analyze the role of Fbxl10 and compared wild-type with Fbxl10 overexpressing mouse embryonic fibroblasts (MEFs). Our study shows that overexpression of Fbxl10 in MEFs doesn't influence the proliferation capability but leads to an enhanced migration capacity in comparison to wild-type MEFs. Transcriptome and ChIP-seq experiments demonstrated that Fbxl10 binds to genes involved in migration like Areg, Mdk, Lmnb1, Thbs1, Mgp and Cxcl12. Taken together, our results strongly suggest that Fbxl10 plays a critical role in migration by binding to the promoter region of migration-associated genes and thereby might influences cell behaviour to a possibly more aggressive phenotype.


Assuntos
Movimento Celular , Embrião de Mamíferos/citologia , Proteínas F-Box/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Doxiciclina/farmacologia , Proteínas F-Box/genética , Feminino , Fibroblastos/efeitos dos fármacos , Perfilação da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Interferente Pequeno/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética , Transgenes
4.
Proc Natl Acad Sci U S A ; 111(29): 10574-9, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-25002509

RESUMO

Metformin, a first-line diabetes drug linked to cancer prevention in retrospective clinical analyses, inhibits cellular transformation and selectively kills breast cancer stem cells (CSCs). Although a few metabolic effects of metformin and the related biguanide phenformin have been investigated in established cancer cell lines, the global metabolic impact of biguanides during the process of neoplastic transformation and in CSCs is unknown. Here, we use LC/MS/MS metabolomics (>200 metabolites) to assess metabolic changes induced by metformin and phenformin in an Src-inducible model of cellular transformation and in mammosphere-derived breast CSCs. Although phenformin is the more potent biguanide in both systems, the metabolic profiles of these drugs are remarkably similar, although not identical. During the process of cellular transformation, biguanide treatment prevents the boost in glycolytic intermediates at a specific stage of the pathway and coordinately decreases tricarboxylic acid (TCA) cycle intermediates. In contrast, in breast CSCs, biguanides have a modest effect on glycolytic and TCA cycle intermediates, but they strongly deplete nucleotide triphosphates and may impede nucleotide synthesis. These metabolic profiles are consistent with the idea that biguanides inhibit mitochondrial complex 1, but they indicate that their metabolic effects differ depending on the stage of cellular transformation.


Assuntos
Ciclo do Ácido Cítrico/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Metformina/farmacologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Nucleotídeos/metabolismo , Fenformin/farmacologia , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/metabolismo , Biguanidas/farmacologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Ácido Fólico/metabolismo , Glicerofosfatos/metabolismo , Humanos , Lactatos/metabolismo , Metaboloma/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Ribonucleotídeos/metabolismo , Tamoxifeno/farmacologia , Quinases da Família src/metabolismo
5.
J Biol Chem ; 287(37): 30984-92, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22825849

RESUMO

Fbxl10 (Jhdm1b/Kdm2b) is a conserved and ubiquitously expressed member of the JHDM (JmjC domain-containing histone demethylase) family. Fbxl10 was implicated in the demethylation of H3K4me3 or H3K36me2 thereby removing active chromatin marks and inhibiting gene transcription. Apart from the JmjC domain, Fbxl10 consists of a CxxC domain, a PHD domain, and an Fbox domain. By purifying the JmjC and the PHD domain of Fbxl10 and using different approaches we were able to characterize the properties of these domains in vitro. Our results suggest that Fbxl10 is rather a H3K4me3 than a H3K36me2 histone demethylase. The PHD domain exerts a dual function in binding H3K4me3 and H3K36me2 and exhibiting E3 ubiquitin ligase activity. We generated mouse embryonic fibroblasts stably overexpressing Fbxl10. These cells reveal an increase in cell size but no changes in proliferation, mitosis, or apoptosis. Using a microarray approach we were able to identify potentially new target genes for Fbxl10 including chemokines, the noncoding RNA Xist, and proteins involved in metabolic processes. Additionally, we found that Fbxl10 is recruited to the promoters of Ccl7, Xist, Crabp2, and RipK3. Promoter occupancy by Fbxl10 was accompanied by reduced levels of H3K4me3 but unchanged levels of H3K36me2. Furthermore, knockdown of Fbxl10 using small interfering RNA approaches showed inverse regulation of Fbxl10 target genes. In summary, our data reveal a regulatory role of Fbxl10 in cell morphology, chemokine expression, and the metabolic control of fibroblasts.


Assuntos
Quimiocinas/biossíntese , Proteínas F-Box/metabolismo , Fibroblastos/metabolismo , Regulação da Expressão Gênica/fisiologia , Histona Desmetilases com o Domínio Jumonji/metabolismo , Metaboloma/fisiologia , Oxirredutases N-Desmetilantes/metabolismo , Animais , Células Cultivadas , Quimiocinas/genética , Proteínas F-Box/genética , Fibroblastos/citologia , Histonas/genética , Histonas/metabolismo , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Camundongos , Oxirredutases N-Desmetilantes/genética , Regiões Promotoras Genéticas/fisiologia , Estrutura Terciária de Proteína , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Transcrição Gênica/fisiologia
6.
Clin Exp Med ; 23(8): 5445-5461, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37935952

RESUMO

The PI3K pathway is one of the most frequently altered signaling pathways in human cancer. In addition to its function in cancer cells, PI3K plays a complex role in modulating anti-tumor immune responses upon immune checkpoint inhibition (ICI). Here, we evaluated the effects of the pan-Class I PI3K inhibitor copanlisib on different immune cell types in vitro and on tumor growth and immune cell infiltration in syngeneic murine cancer models. Intermittent treatment with copanlisib resulted in a strong in vivo anti-tumor efficacy, increased tumor infiltration of activated T cells and macrophages, and increased CD8+ T cell/regulatory T cell and M1/M2 macrophage ratios. The strong in vivo efficacy was at least partially due to immunomodulatory activity of copanlisib, as in vitro these murine cancer cells were resistant to PI3K inhibition. Furthermore, the combination of copanlisib with the ICI antibody anti-PD-1 demonstrated enhanced anti-tumor efficacy in both ICI-sensitive and insensitive syngeneic mouse tumor models. Importantly, in an ICI-sensitive model, combination therapy resulted in complete remission and prevention of tumor recurrence. Thus, the combination of ICIs with PI3K inhibition by intermittently dosed copanlisib represents a promising new strategy to increase sensitivity to ICI therapies and to treat human solid cancers.


Assuntos
Neoplasias , Fosfatidilinositol 3-Quinases , Humanos , Animais , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Linfócitos T Reguladores/metabolismo , Neoplasias/tratamento farmacológico , Imunidade , Microambiente Tumoral
7.
Int J Cancer ; 131(11): 2704-9, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22447389

RESUMO

Post-translational modifications of histones by chromatin modifying enzymes regulate chromatin structure and gene expression. As deregulation of histone modifications contributes to cancer progression, inhibition of chromatin modifying enzymes such as histone demethylases is an attractive therapeutic strategy to impair cancer growth. Lysine-specific demethylase 1 (LSD1) removes mono- and dimethyl marks from lysine 4 or 9 of histone H3. LSD1 in association with the androgen receptor (AR) controls androgen-dependent gene expression and prostate tumor cell proliferation, thus highlighting LSD1 as a drug target. By combining protein structure similarity clustering and in vitro screening, we identified Namoline, a γ-pyrone, as a novel, selective and reversible LSD1 inhibitor. Namoline blocks LSD1 demethylase activity in vitro and in vivo. Inhibition of LSD1 by Namoline leads to silencing of AR-regulated gene expression and severely impairs androgen-dependent proliferation in vitro and in vivo. Thus, Namoline is a novel promising starting compound for the development of therapeutics to treat androgen-dependent prostate cancer.


Assuntos
Inibidores Enzimáticos/farmacologia , Histona Desmetilases/antagonistas & inibidores , Neoplasias da Próstata/tratamento farmacológico , Pironas/farmacologia , Androgênios/metabolismo , Animais , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Metilação/efeitos dos fármacos , Camundongos , Camundongos Nus , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptores Androgênicos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Biochem Biophys Res Commun ; 421(4): 665-70, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22542627

RESUMO

Histone modifying enzymes confer epigenetic marks, directing the changes in gene expression required for diverse cellular processes. Lysine-specific demethylase 1 (LSD1) functions as a transcriptional coregulator by demethylating histone H3 on lysine 4 and lysine 9. Analyzing transcriptomes on microarrays, we identified genes which represent inflammatory-related targets of LSD1. We demonstrate a repressive role of LSD1 in proinflammatory cytokine expression such as IL1α, IL1ß, IL6 and IL8 and classical complement components. Consistently, LSD1 occupies and regulates the promoter of these genes. In addition, we demonstrate that HDAC1 and LSD1 synergistically regulate these inflammatory-related genes. Our data reveal a novel role for LSD1 in suppressing immune responses.


Assuntos
Via Clássica do Complemento/genética , Citocinas/genética , Epigênese Genética , Histona Desacetilase 1/metabolismo , Histona Desmetilases/metabolismo , Imunidade/genética , Inflamação/genética , Linhagem Celular Tumoral , Citocinas/antagonistas & inibidores , Regulação para Baixo , Técnicas de Silenciamento de Genes , Células Hep G2 , Histona Desacetilase 1/genética , Histona Desmetilases/genética , Humanos
9.
Cancer Cell ; 40(9): 939-956.e16, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-35985343

RESUMO

Mutations affecting isocitrate dehydrogenase (IDH) enzymes are prevalent in glioma, leukemia, and other cancers. Although mutant IDH inhibitors are effective against leukemia, they seem to be less active in aggressive glioma, underscoring the need for alternative treatment strategies. Through a chemical synthetic lethality screen, we discovered that IDH1-mutant glioma cells are hypersensitive to drugs targeting enzymes in the de novo pyrimidine nucleotide synthesis pathway, including dihydroorotate dehydrogenase (DHODH). We developed a genetically engineered mouse model of mutant IDH1-driven astrocytoma and used it and multiple patient-derived models to show that the brain-penetrant DHODH inhibitor BAY 2402234 displays monotherapy efficacy against IDH-mutant gliomas. Mechanistically, this reflects an obligate dependence of glioma cells on the de novo pyrimidine synthesis pathway and mutant IDH's ability to sensitize to DNA damage upon nucleotide pool imbalance. Our work outlines a tumor-selective, biomarker-guided therapeutic strategy that is poised for clinical translation.


Assuntos
Neoplasias Encefálicas , Glioma , Leucemia , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Inibidores Enzimáticos/uso terapêutico , Glioma/tratamento farmacológico , Glioma/genética , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Camundongos , Mutação , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Salicilanilidas , Triazóis
10.
Biochem J ; 430(1): 87-95, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20518742

RESUMO

Matriptase-2 is a member of the TTSPs (type II transmembrane serine proteases), an emerging class of cell surface proteases involved in tissue homoeostasis and several human disorders. Matriptase-2 exhibits a domain organization similar to other TTSPs, with a cytoplasmic N-terminus, a transmembrane domain and an extracellular C-terminus containing the non-catalytic stem region and the protease domain. To gain further insight into the biochemical functions of matriptase-2, we characterized the subcellular localization of the monomeric and multimeric form and identified cell surface shedding as a defining point in its proteolytic processing. Using HEK (human embryonic kidney)-293 cells, stably transfected with cDNA encoding human matriptase-2, we demonstrate a cell membrane localization for the inactive single-chain zymogen. Membrane-associated matriptase-2 is highly N-glycosylated and occurs in monomeric, as well as multimeric, forms covalently linked by disulfide bonds. Furthermore, matriptase-2 undergoes shedding into the conditioned medium as an activated two-chain form containing the catalytic domain, which is cleaved at the canonical activation motif, but is linked to a released portion of the stem region via a conserved disulfide bond. Cleavage sites were identified by MS, sequencing and mutational analysis. Interestingly, cell surface shedding and activation of a matriptase-2 variant bearing a mutation at the active-site serine residue is dependent on the catalytic activity of co-expressed or co-incubated wild-type matriptase-2, indicating a transactivation and trans-shedding mechanism.


Assuntos
Precursores Enzimáticos/metabolismo , Proteínas de Membrana/metabolismo , Serina Endopeptidases/metabolismo , Catálise , Domínio Catalítico , Linhagem Celular , Membrana Celular/metabolismo , Meios de Cultivo Condicionados , Ativação Enzimática , Precursores Enzimáticos/genética , Espaço Extracelular/enzimologia , Humanos , Proteínas de Membrana/genética , Mutação , Ligação Proteica , Serina Endopeptidases/genética , Transfecção
11.
Carcinogenesis ; 31(3): 512-20, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20042638

RESUMO

Breast carcinogenesis is a multistep process involving both genetic and epigenetic changes. Since epigenetic changes like histone modifications are potentially reversible processes, much effort has been directed toward understanding this mechanism with the goal of finding novel therapies as well as more refined diagnostic and prognostic tools in breast cancer. Lysine-specific demethylase 1 (LSD1) plays a key role in the regulation of gene expression by removing the methyl groups from methylated lysine 4 of histone H3 and lysine 9 of histone H3. LSD1 is essential for mammalian development and involved in many biological processes. Considering recent evidence that LSD1 is involved in carcinogenesis, we investigated the role of LSD1 in breast cancer. Therefore, we developed an enzyme-linked immunosorbent assay to determine LSD1 protein levels in tissue specimens of breast cancer and measured very high LSD1 levels in estrogen receptor (ER)-negative tumors. Pharmacological LSD1 inhibition resulted in growth inhibition of breast cancer cells. Knockdown of LSD1 using small interfering RNA approach induced regulation of several proliferation-associated genes like p21, ERBB2 and CCNA2. Additionally, we found that LSD1 is recruited to the promoters of these genes. In summary, our data indicate that LSD1 may provide a predictive marker for aggressive biology and a novel attractive therapeutic target for treatment of ER-negative breast cancers.


Assuntos
Adenocarcinoma/enzimologia , Neoplasias da Mama/enzimologia , Histona Desmetilases/fisiologia , Invasividade Neoplásica/genética , Proteínas de Neoplasias/fisiologia , Adenocarcinoma/química , Adenocarcinoma/genética , Adenocarcinoma/patologia , Biomarcadores Tumorais , Neoplasias da Mama/química , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral/enzimologia , Imunoprecipitação da Cromatina , Ciclina A2/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Genes erbB-2 , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/biossíntese , Histona Desmetilases/genética , Histonas/análise , Humanos , Metilação , Proteínas de Neoplasias/análise , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Prognóstico , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Receptores de Estrogênio/análise , Receptores de Progesterona/análise
12.
Leukemia ; 33(10): 2403-2415, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30940908

RESUMO

Acute myeloid leukemia (AML) is a devastating disease, with the majority of patients dying within a year of diagnosis. For patients with relapsed/refractory AML, the prognosis is particularly poor with currently available treatments. Although genetically heterogeneous, AML subtypes share a common differentiation arrest at hematopoietic progenitor stages. Overcoming this differentiation arrest has the potential to improve the long-term survival of patients, as is the case in acute promyelocytic leukemia (APL), which is characterized by a chromosomal translocation involving the retinoic acid receptor alpha gene. Treatment of APL with all-trans retinoic acid (ATRA) induces terminal differentiation and apoptosis of leukemic promyelocytes, resulting in cure rates of over 80%. Unfortunately, similarly efficacious differentiation therapies have, to date, been lacking outside of APL. Inhibition of dihydroorotate dehydrogenase (DHODH), a key enzyme in the de novo pyrimidine synthesis pathway, was recently reported to induce differentiation of diverse AML subtypes. In this report we describe the discovery and characterization of BAY 2402234 - a novel, potent, selective and orally bioavailable DHODH inhibitor that shows monotherapy efficacy and differentiation induction across multiple AML subtypes. Herein, we present the preclinical data that led to initiation of a phase I evaluation of this inhibitor in myeloid malignancies.


Assuntos
Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Di-Hidro-Orotato Desidrogenase , Feminino , Células HL-60 , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pirimidinas/metabolismo , Células THP-1 , Translocação Genética/efeitos dos fármacos
13.
Nat Commun ; 9(1): 2068, 2018 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-29802342

RESUMO

Transient activation of Src oncoprotein in non-transformed, breast epithelial cells can initiate an epigenetic switch to the stably transformed state via a positive feedback loop that involves the inflammatory transcription factors STAT3 and NF-κB. Here, we develop an experimental and computational pipeline that includes 1) a Bayesian network model (AccessTF) that accurately predicts protein-bound DNA sequence motifs based on chromatin accessibility, and 2) a scoring system (TFScore) that rank-orders transcription factors as candidates for being important for a biological process. Genetic experiments validate TFScore and suggest that more than 40 transcription factors contribute to the oncogenic state in this model. Interestingly, individual depletion of several of these factors results in similar transcriptional profiles, indicating that a complex and interconnected transcriptional network promotes a stable oncogenic state. The combined experimental and computational pipeline represents a general approach to comprehensively identify transcriptional regulators important for a biological process.


Assuntos
Neoplasias da Mama/genética , Transformação Celular Neoplásica/genética , Epigênese Genética , Regulação Neoplásica da Expressão Gênica/genética , Fatores de Transcrição/metabolismo , Mama/citologia , Mama/patologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Redes Reguladoras de Genes/genética , Genoma Humano/genética , Humanos , Modelos Genéticos , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA