Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Immunity ; 44(4): 913-923, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27096320

RESUMO

The participation of a specific subset of B cells and how they are regulated in cancer is unclear. Here, we demonstrate that the proportion of CD5(+) relative to interleukin-6 receptor α (IL-6Rα)-expressing B cells was greatly increased in tumors. CD5(+) B cells responded to IL-6 in the absence of IL-6Rα. IL-6 directly bound to CD5, leading to activation of the transcription factor STAT3 via gp130 and its downstream kinase JAK2. STAT3 upregulated CD5 expression, thereby forming a feed-forward loop in the B cells. In mouse tumor models, CD5(+) but not CD5(-) B cells promoted tumor growth. CD5(+) B cells also showed activation of STAT3 in multiple types of human tumor tissues. Thus, our findings demonstrate a critical role of CD5(+) B cells in promoting cancer.


Assuntos
Linfócitos B/imunologia , Antígenos CD5/metabolismo , Interleucina-6/metabolismo , Melanoma Experimental/patologia , Fator de Transcrição STAT3/imunologia , Animais , Antígenos CD5/biossíntese , Linhagem Celular Tumoral , Receptor gp130 de Citocina/metabolismo , Humanos , Interleucina-6/imunologia , Janus Quinase 2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3 , Ligação Proteica , Receptores de Interleucina-6/biossíntese , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/imunologia , Ativação Transcricional/imunologia
3.
Proc Natl Acad Sci U S A ; 110(32): 13079-84, 2013 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-23878227

RESUMO

Dysregulated inflammation in adipose tissue, marked by increased proinflammatory T-cell accumulation and reduced regulatory T cells (Tregs), contributes to obesity-associated insulin resistance. The molecular mechanisms underlying T-cell-mediated inflammation in adipose tissue remain largely unknown, however. Here we show a crucial role for signal transducer and activator of transcription 3 (Stat3) in T cells in skewing adaptive immunity in visceral adipose tissue (VAT), thereby contributing to diet-induced obesity (DIO) and insulin resistance. Stat3 activity is elevated in obese VAT and in VAT-resident T cells. Functional ablation of Stat3 in T cells reduces DIO, improves insulin sensitivity and glucose tolerance, and suppresses VAT inflammation. Importantly, Stat3 ablation reverses the high Th1/Treg ratio in VAT of DIO mice that is likely secondary to elevated IL-6 production, leading in turn to suppression of Tregs. In addition, Stat3 in T cells in DIO mice affects adipose tissue macrophage accumulation and M2 phenotype. Our study identifies Stat3 in VAT-resident T cells as an important mediator and direct target for regulating adipose tissue inflammation, DIO, and its associated metabolic dysfunctions.


Assuntos
Resistência à Insulina/imunologia , Gordura Intra-Abdominal/imunologia , Obesidade/imunologia , Fator de Transcrição STAT3/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Glicemia/metabolismo , Western Blotting , Dieta Hiperlipídica/efeitos adversos , Jejum/sangue , Feminino , Citometria de Fluxo , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Insulina/sangue , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/sangue , Obesidade/etiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/imunologia , Células Th1/metabolismo
4.
Nucleic Acids Res ; 41(11): e117, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23598999

RESUMO

COHCAP (City of Hope CpG Island Analysis Pipeline) is an algorithm to analyze single-nucleotide resolution DNA methylation data produced by either an Illumina methylation array or targeted bisulfite sequencing. The goal of the COHCAP algorithm is to identify CpG islands that show a consistent pattern of methylation among CpG sites. COHCAP is currently the only DNA methylation package that provides integration with gene expression data to identify a subset of CpG islands that are most likely to regulate downstream gene expression, and it can generate lists of differentially methylated CpG islands with ∼50% concordance with gene expression from both cell line data and heterogeneous patient data. For example, this article describes known breast cancer biomarkers (such as estrogen receptor) with a negative correlation between DNA methylation and gene expression. COHCAP also provides visualization for quality control metrics, regions of differential methylation and correlation between methylation and gene expression. This software is freely available at https://sourceforge.net/projects/cohcap/.


Assuntos
Algoritmos , Ilhas de CpG , Metilação de DNA , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Feminino , Expressão Gênica , Genômica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Análise de Sequência de DNA , Sulfitos
5.
Proc Natl Acad Sci U S A ; 109(20): 7765-9, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22547799

RESUMO

The mechanisms underlying hypermethylation of tumor-suppressor gene promoters in cancer is not well understood. Here, we report that lysine acetylation of the oncogenic transcription factor STAT3 is elevated in tumors. We also show that genetically altering STAT3 at Lys685 reduces tumor growth, which is accompanied by demethylation and reactivation of several tumor-suppressor genes. Moreover, mutating STAT3 at Lys685 disrupts DNA methyltransferase 1-STAT3 interactions in cultured tumor cells and in tumors. These observations are confirmed by treatment with an acetylation inhibitor, resveratrol. Furthermore, reduction of acetylated STAT3 in triple-negative breast cancer cells leads to demethylation and activation of the estrogen receptor-α gene, sensitizing the tumor cells to antiestrogens. Our results also demonstrate a correlation between STAT3 acetylation and methylation of estrogen receptor-α in melanoma, which predicts melanoma progression. Taken together, these results suggest a role of STAT3 acetylation in regulating CpG island methylation, which may partially explain aberrant gene silencing in cancer. These findings also provide a rationale for targeting acetylated STAT3 for chemoprevention and cancer therapy.


Assuntos
Metilação de DNA/genética , Genes Supressores de Tumor , Regiões Promotoras Genéticas/genética , Fator de Transcrição STAT3/metabolismo , Estilbenos/farmacologia , Acetilação , Análise de Variância , Animais , Western Blotting , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Metilação de DNA/efeitos dos fármacos , Humanos , Imunoprecipitação , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Reação em Cadeia da Polimerase em Tempo Real , Resveratrol , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética
6.
J Biol Chem ; 288(19): 13842-9, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23548897

RESUMO

BACKGROUND: Signaling pathways underlying BV8-mediated oncogenesis remain unknown. RESULTS: BV8-STAT3 forms a feed-forward loop in both normal and malignant myeloid cells and promotes tumor growth. CONCLUSION: JAK2/STAT3 signaling plays critical roles in BV8-mediated myeloid cell-dependent oncogenesis. SIGNIFICANCE: This study identifies a novel role of BV8-STAT3 signaling in mediating cross-talk between tumor microenvironment and tumor cells. An important role of BV8 in mobilization of myeloid cells and myeloid cell-dependent angiogenesis has been established. Recently, it has also been shown that granulocyte colony-stimulating factor (G-CSF)-induced BV8 expression is STAT3 dependent in CD11b(+)Gr1(+) myeloid cells. However, the BV8 downstream signaling pathway(s) intrinsic to myeloid cells crucial for angiogenesis, and potentially also for development of cancers of myeloid origin, remains largely unknown. Here we show that BV8 activates STAT3, which is critical for regulating genes important for both tumor cell proliferation/survival and tumor angiogenesis, in both normal and malignant myeloid cells. Further, BV8-induced STAT3 activation requires Janus-activated kinase 2 (JAK2) activity as shown by both genetic and pharmacologic inhibition. Knocking down BV8 in human myeloid leukemia cells inhibits STAT3 activity and expression of STAT3 downstream angiogenic and pro-proliferation/survival genes, leading to a decrease in tumor cell viability. BV8 shRNA expressing leukemia cells exhibit reduced STAT3 activity and tumor growth in vivo. Taken together, we have delineated a signaling pathway downstream of BV8 that plays critical roles in both the tumor microenvironment and malignant myeloid cells for angiogenesis and tumor cell proliferation/survival.


Assuntos
Hormônios Gastrointestinais/genética , Leucemia Mieloide Aguda/metabolismo , Células Mieloides/metabolismo , Neuropeptídeos/genética , Fator de Transcrição STAT3/metabolismo , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Retroalimentação Fisiológica , Hormônios Gastrointestinais/metabolismo , Hormônios Gastrointestinais/fisiologia , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Janus Quinase 2/metabolismo , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Transplante de Neoplasias , Neuropeptídeos/metabolismo , Neuropeptídeos/fisiologia , RNA Interferente Pequeno/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Carga Tumoral
7.
Blood ; 120(7): 1458-65, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22745305

RESUMO

STAT3 plays a crucial role in promoting progression of human cancers, including several types of B-cell lymphoma. However, as a transcription factor lacking its own enzymatic activity, STAT3 remains difficult to target with small-molecule drugs in the clinic. Here we demonstrate that persistent activated STAT3 colocalizes with elevated expression of S1PR1, a G-protein-coupled receptor for sphingosine-1-phosphate (S1P), in the tumor cells of the activated B cell-like subtype of diffuse large B-cell lymphoma patient specimens. Inhibition of S1PR1 expression by shRNA in the lymphoma cells validates that blocking S1PR1 affects expression of STAT3 downstream genes critically involved in tumor cell survival, proliferation, tumor invasion, and/or immunosuppression. Using S1PR1 shRNA, or FTY720, an antagonist of S1P that is in the clinic for other indications, we show that inhibiting S1PR1 expression down-regulates STAT3 activity and causes growth inhibition of the lymphoma tumor cells in vitro and in vivo. Our results suggest that targeting S1P/S1PR1 using a clinically relevant and available drug or other approaches is potentially an effective new therapeutic modality for treating the activated B cell-like subtype of diffuse large B-cell lymphoma, a subset of lymphoma that is less responsive to current available therapies.


Assuntos
Linfócitos B/imunologia , Ativação Linfocitária/imunologia , Linfoma Difuso de Grandes Células B/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Linfócitos B/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Cloridrato de Fingolimode , Inativação Gênica/efeitos dos fármacos , Humanos , Ativação Linfocitária/efeitos dos fármacos , Linfoma Difuso de Grandes Células B/imunologia , Linfoma Difuso de Grandes Células B/patologia , Camundongos , Invasividade Neoplásica , Fosforilação/efeitos dos fármacos , Propilenoglicóis/farmacologia , RNA Interferente Pequeno/metabolismo , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato
8.
Curr Top Microbiol Immunol ; 344: 41-59, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20517723

RESUMO

Signal transducer and activator of transcription 3 (Stat3) has emerged as a critical regulator for tumor-associated inflammation. Activation of Stat3 negatively regulates the Th1-type immune response and promotes expansion of myeloid-derived suppressor cells (MDSCs) and regulatory T-cell functions in the tumor microenvironment. Mounting evidence suggests that Stat3 and related pathways may serve as a target for changing the tumor immunologic microenvironment to benefit cancer immunotherapies. Many recent studies support the use of certain tyrosine kinase inhibitors, through inhibition of Stat3, in decreasing immunosuppression in the tumor microenvironment. Other potential therapeutic avenues include the use of targeted delivery of Stat3 siRNA into immune cells. Here, we describe the role of Stat3 in regulating the immunologic properties of tumors as a background for Stat3-based therapeutic interventions.


Assuntos
Neoplasias/imunologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/fisiologia , Animais , Humanos , Tolerância Imunológica , Neoplasias/terapia , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais , Células Th1/imunologia
9.
J Clin Invest ; 118(10): 3367-77, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18776941

RESUMO

The underlying molecular mechanisms that cause immune cells, mediators of our defense system, to promote tumor invasion and angiogenesis remain incompletely understood. Constitutively activated Stat3 in tumor cells has been shown to promote tumor invasion and angiogenesis. Therefore, we sought to determine whether Stat3 activation in tumor-associated inflammatory cells has a similar function. We found that Stat3 signaling mediates multidirectional crosstalk among tumor cells, myeloid cells in the tumor stroma, and ECs that contributes to tumor angiogenesis in mice. Myeloid-derived suppressor cells and macrophages isolated from mouse tumors displayed activated Stat3 and induced angiogenesis in an in vitro tube formation assay via Stat3 induction of angiogenic factors, including VEGF and bFGF. Stat3-regulated factors produced by both tumor cells and tumor-derived myeloid cells also induced constitutive activation of Stat3 in tumor endothelium, and inhibiting Stat3 in ECs substantially reduced in vitro tumor factor-induced endothelial migration and tube formation. In vivo assays demonstrated the requirement for Stat3 signaling in tumor-associated myeloid cells for tumor angiogenesis. Our results indicate that, by virtue of the ability of Stat3 in tumor cells and tumor-derived myeloid cells to upregulate expression of factors that activate Stat3 in ECs, Stat3 mediates multidirectional crosstalk among tumor cells, tumor-associated myeloid cells, and ECs that contributes to tumor angiogenesis.


Assuntos
Células Endoteliais/metabolismo , Células Mieloides/metabolismo , Neoplasias/irrigação sanguínea , Neovascularização Patológica/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Linhagem Celular Tumoral , Colágeno/metabolismo , Regulação da Expressão Gênica , Humanos , Análise dos Mínimos Quadrados , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL
10.
Oncogene ; 39(10): 2156-2169, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31819166

RESUMO

Both the extracellular matrix (ECM) and DNA epigenetic regulation are critical for maintaining stem cell phenotype and cancer progression. Whether and how ECM regulates epigenetic alterations to influence cancer stem cells (CSCs) remain to be explored. Here we report that ECM through laminin-integrin α6 upregulates ten-eleven translocation enzyme 3 (TET3) dioxygenase. TET3 in turn mediates DNA cytosine 5'-hydroxymethylation (5hmC) and upregulates genes critical for maintenance of glioma stem cells (GSCs). Activating integrin α6-FAK pathway increases STAT3 activity, TET3 expression and 5hmC levels in GSCs. Moreover, targeting STAT3 disrupts integrin α6-FAK signaling and inhibits TET3+ GSC maturation in vivo. STAT3 directly regulates TET3 expression and the two proteins are co-localized with 5hmC in GSC clusters. 5hmC is upregulated by STAT3 at the promoters of several tumorigenic genes, including c-Myc, known to be critical for GSCs. In vivo silencing of TET3 in GSC-enriched tumors reduces 5hmC accumulation and expression of the GSC critical genes, leading to tumor growth inhibition. TET3 expression and 5hmC accumulation also co-segregate with integrin α6 in patient malignant glioma. Thus, ECM- integrin α6-STAT3-TET3 axis regulates hydroxymethylation of genes important for GSCs, thereby increasing GSC tumorigenicity and resistance to therapies.


Assuntos
Metilação de DNA , Dioxigenases/metabolismo , Epigênese Genética , Glioma/genética , Integrina alfa6/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fator de Transcrição STAT3/metabolismo , 5-Metilcitosina , Animais , Linhagem Celular , Dioxigenases/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Glioma/enzimologia , Glioma/metabolismo , Humanos , Masculino , Camundongos Nus , Células-Tronco Neoplásicas/enzimologia , Transdução de Sinais
11.
Cell Metab ; 31(1): 148-161.e5, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31761565

RESUMO

Although obesity is known to be critical for cancer development, how obesity negatively impacts antitumor immune responses remains largely unknown. Here, we show that increased fatty acid oxidation (FAO) driven by activated STAT3 in CD8+ T effector cells is critical for obesity-associated breast tumor progression. Ablating T cell Stat3 or treatment with an FAO inhibitor in obese mice spontaneously developing breast tumor reduces FAO, increases glycolysis and CD8+ T effector cell functions, leading to inhibition of breast tumor development. Moreover, PD-1 ligation in CD8+ T cells activates STAT3 to increase FAO, inhibiting CD8+ T effector cell glycolysis and functions. Finally, leptin enriched in mammary adipocytes and fat tissues downregulates CD8+ T cell effector functions through activating STAT3-FAO and inhibiting glycolysis. We identify a critical role of increased oxidation of fatty acids driven by leptin and PD-1 through STAT3 in inhibiting CD8+ T effector cell glycolysis and in promoting obesity-associated breast tumorigenesis.


Assuntos
Neoplasias da Mama/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Carcinogênese/imunologia , Ácidos Graxos/metabolismo , Obesidade/metabolismo , Fator de Transcrição STAT3/metabolismo , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Animais , Neoplasias da Mama/metabolismo , Carcinogênese/genética , Carcinogênese/metabolismo , Linhagem Celular , Proliferação de Células/genética , Imunoprecipitação da Cromatina , Feminino , Glicólise/genética , Glicólise/fisiologia , Humanos , Interferon gama/metabolismo , Leptina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Obesidade/imunologia , Oxirredução/efeitos dos fármacos , Receptor de Morte Celular Programada 1/metabolismo , Fator de Transcrição STAT3/genética
12.
Mol Cancer Res ; 6(7): 1099-105, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18644974

RESUMO

Hypoxia-inducible factor 1 (HIF-1) is a potent tumorigenic factor. Its alpha subunit (HIF-1alpha), which is tightly regulated in normal tissues, is elevated in tumors due to hypoxia and overactive growth signaling pathways. Although much is known about HIF-1alpha regulation in cancer cells, crucial molecular targets that affect HIF-1alpha levels modulated by both hypoxia and oncogenic signaling pathways remain to be identified. Additionally, whether and how the tumor microenvironment contributes to HIF-1alpha accumulation is unclear. This study shows a novel mechanism by which HIF-1alpha availability is regulated in both cancer cells and in myeloid cells in the tumor microenvironment. We show a requirement of signal transducer and activator of transcription 3 (Stat3) for HIF-1alpha RNA expression under both hypoxia and growth signaling conditions. Furthermore, tumor-derived myeloid cells express elevated levels of HIF-1alpha mRNA relative to their counterparts from normal tissues in a Stat3-dependent manner. Additionally, Stat3 activity in the nontransformed cells in the tumor milieu affects HIF-1alpha RNA expression of the entire growing tumor. Consistent with a role of Stat3 in regulating HIF-1alpha RNA transcription, elevated Stat3 activity increases HIF-1alpha promoter activity, and Stat3 protein binds to the HIF-1alpha promoter in both transformed cells and in growing tumors. Taken together, these findings show a novel mode by which HIF-1alpha is regulated not only in cancer cells but also in the tumor-associated inflammatory cells, suggesting Stat3 as an important molecular target for inhibiting the oncogenic potential of HIF-1 induced by both hypoxia and overactive growth signaling pathways prevalent in cancer.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Células Mieloides/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Hipóxia Celular , Linhagem Celular Transformada , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Células Estromais/metabolismo
13.
Mol Cell Biol ; 26(14): 5259-69, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16809764

RESUMO

Histone deacetylases (HDACs) are enzymes that regulate the functions of histones as well as nonhistones by catalyzing the removal of acetyl groups from lysine residues. HDACs regulate many biological processes, including the cell division cycle and tumorigenesis. Although recent studies have implicated HDAC8 in tumor cell proliferation, the molecular mechanisms linking HDAC8 to cell growth remain unknown. Here, we report that the human ortholog of the yeast ever-shorter telomeres 1B (EST1B) binds HDAC8. This interaction is regulated by protein kinase A-mediated HDAC8 phosphorylation and protects human EST1B (hEST1B) from ubiquitin-mediated degradation. Phosphorylated HDAC8 preferentially recruits Hsp70 to a complex that inhibits the CHIP (C-terminal heat shock protein interacting protein) E3 ligase-mediated degradation of hEST1B. Importantly, HDAC8 regulation of hEST1B protein stability modulates total telomerase enzymatic activity. Our findings reveal a novel mechanism by which HDAC8 contributes to tumorigenesis by regulating telomerase activity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Histona Desacetilases/metabolismo , Proteínas Repressoras/metabolismo , Telomerase/metabolismo , Ubiquitina/metabolismo , Acetilação , Sequência de Bases , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , DNA Complementar/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Inativação Gênica , Proteínas de Choque Térmico HSP70/química , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP90/química , Proteínas de Choque Térmico HSP90/metabolismo , Células HeLa , Histona Desacetilases/química , Histona Desacetilases/genética , Humanos , Técnicas In Vitro , Complexos Multiproteicos , Fosforilação , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Telomerase/química , Telomerase/genética , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
14.
Cell Metab ; 27(1): 136-150.e5, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29249690

RESUMO

Cancer stem cells (CSCs) are critical for cancer progression and chemoresistance. How lipid metabolism regulates CSCs and chemoresistance remains elusive. Here, we demonstrate that JAK/STAT3 regulates lipid metabolism, which promotes breast CSCs (BCSCs) and cancer chemoresistance. Inhibiting JAK/STAT3 blocks BCSC self-renewal and expression of diverse lipid metabolic genes, including carnitine palmitoyltransferase 1B (CPT1B), which encodes the critical enzyme for fatty acid ß-oxidation (FAO). Moreover, mammary-adipocyte-derived leptin upregulates STAT3-induced CPT1B expression and FAO activity in BCSCs. Human breast-cancer-derived data suggest that the STAT3-CPT1B-FAO pathway promotes cancer cell stemness and chemoresistance. Blocking FAO and/or leptin re-sensitizes them to chemotherapy and inhibits BCSCs in mouse breast tumors in vivo. We identify a critical pathway for BCSC maintenance and breast cancer chemoresistance.


Assuntos
Neoplasias da Mama/patologia , Autorrenovação Celular , Resistencia a Medicamentos Antineoplásicos , Janus Quinases/metabolismo , Células-Tronco Neoplásicas/patologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Adipócitos/metabolismo , Idoso , Animais , Neoplasias da Mama/genética , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Linhagem Celular Tumoral , Ácidos Graxos/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Leptina/metabolismo , Metabolismo dos Lipídeos/genética , Metabolômica , Camundongos , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/metabolismo , Oxirredução , Transcrição Gênica
15.
Mol Cell Biol ; 22(16): 5835-45, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12138194

RESUMO

Estrogen receptors are phosphoproteins which can be activated by ligands, kinase activators, or phosphatase inhibitors. Our previous study showed that p38 mitogen-activated protein kinase was involved in estrogen receptor activation by estrogens and MEKK1. Here, we report estrogen receptor-dependent p38 activation by estrogens in endometrial adenocarcinoma cells and in vitro and in vivo phosphorylation of the estrogen receptor alpha mediated through p38. The phosphorylation site was identified as threonine-311 (Thr(311)), located in helix 1 of the hormone-binding domain. The mutation of threonine-311 to alanine did not affect estrogen binding of the receptor but compromised its interaction with coactivators. Suppression of p38 activity or mutation of the site inhibited the estrogen-induced receptor nuclear localization as well as its transcriptional activation by estrogens and MEKK1. The inhibition of the p38 signal pathway by a specific chemical inhibitor blocked the biological activities of estrogens in regulating endogenous gene expression as well as endometrial cancer cell growth. Our studies demonstrate the role of estrogen receptor phosphorylation induced by the natural ligand in estrogen receptor's cellular distribution and its significant contribution to the growth-stimulating activity of estrogens in endometrial cancer cells.


Assuntos
Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Estradiol/farmacologia , MAP Quinase Quinase Quinase 1 , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores de Estrogênio/metabolismo , Tamoxifeno/análogos & derivados , Transporte Ativo do Núcleo Celular/fisiologia , Adenocarcinoma/fisiopatologia , Sequência de Aminoácidos , Animais , Neoplasias do Endométrio/fisiopatologia , Ativação Enzimática , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio , Ácidos Graxos Insaturados/farmacologia , Feminino , Humanos , Ligantes , Dados de Sequência Molecular , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Sinais Direcionadores de Proteínas , Receptores de Estrogênio/genética , Transdução de Sinais/fisiologia , Tamoxifeno/farmacologia , Treonina/metabolismo , Transcrição Gênica , Proteínas Quinases p38 Ativadas por Mitógeno
16.
Mol Cell Biol ; 24(2): 765-73, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14701748

RESUMO

Histone deacetylases (HDACs) are enzymes that catalyze the removal of acetyl groups from lysine residues of histone and nonhistone proteins. Recent studies suggest that they are key regulators of many cellular events, including cell proliferation and cancer development. Human class I HDACs possess homology to the yeast RPD3 protein and include HDAC1, HDAC2, HDAC3, and HDAC8. While HDAC1, HDAC2, and HDAC3 have been characterized extensively, almost nothing is known about HDAC8. Here we report that HDAC8 is phosphorylated by cyclic AMP-dependent protein kinase A (PKA) in vitro and in vivo. The PKA phosphoacceptor site of HDAC8 is Ser(39), a nonconserved residue among class I HDACs. Mutation of Ser(39) to Ala enhances the deacetylase activity of HDAC8. In contrast, mutation of Ser(39) to Glu or induction of HDAC8 phosphorylation by forskolin, a potent activator of adenyl cyclase, decreases HDAC8's enzymatic activity. Remarkably, inhibition of HDAC8 activity by hyperphosphorylation leads to hyperacetylation of histones H3 and H4, suggesting that PKA-mediated phosphorylation of HDAC8 plays a central role in the overall acetylation status of histones.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Histona Desacetilases/metabolismo , Proteínas Repressoras/metabolismo , Acetilação , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação/genética , Domínio Catalítico/genética , DNA Complementar/genética , Células HeLa , Histona Desacetilases/química , Histona Desacetilases/genética , Histonas/química , Histonas/metabolismo , Humanos , Técnicas In Vitro , Dados de Sequência Molecular , Fosforilação , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Serina/química
17.
Mol Cell Biol ; 23(1): 104-18, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12482965

RESUMO

Recent studies suggested that the protection of cell apoptosis by AKT involves phosphorylation and inhibition of FKHR and related FOXO forkhead transcription factors and that androgens provide an AKT-independent cell survival signal in prostate cancer cells. Here, we report receptor-dependent repression of FKHR function by androgens in prostate cancer cells. Transcriptional analysis demonstrated that activation of the androgen receptor caused an inhibition of both wild-type FKHR and a mutant in which all three known AKT sites were mutated to alanines, showing that the repression is AKT independent. In vivo and in vitro coprecipitation studies demonstrated that the repression is mediated through protein-protein interaction between FKHR and the androgen receptor. Mapping analysis localized the interacting domains to the carboxyl terminus between amino acids 350 and 655 of FKHR and to the amino-terminal A/B region and the ligand binding domain of the receptor. Further analysis demonstrated that the activated androgen receptor blocked FKHR's DNA binding activity and impaired its ability to induce Fas ligand expression and prostate cancer cell apoptosis and cell cycle arrest. These studies identify a new mechanism for androgen-mediated prostate cancer cell survival that appears to be independent of the activity of the receptor on androgen response element-mediated transcription and establish FKHR and related FOXO forkhead proteins as important nuclear targets for both AKT-dependent and -independent survival signals in prostate cancer cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Receptores Androgênicos/metabolismo , Fatores de Transcrição/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Sítios de Ligação , Ciclo Celular/fisiologia , Proteínas de Ligação a DNA/genética , Receptor beta de Estrogênio , Proteína Ligante Fas , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead , Humanos , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Metribolona/farmacologia , Mutação , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Receptores Androgênicos/efeitos dos fármacos , Receptores Androgênicos/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Elementos de Resposta , Congêneres da Testosterona/farmacologia , Fatores de Transcrição/genética , Transcrição Gênica , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
18.
Mol Cancer Ther ; 16(11): 2516-2527, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28716816

RESUMO

Drug resistance is a major barrier for the development of effective and durable cancer therapies. Overcoming this challenge requires further defining the cellular and molecular mechanisms underlying drug resistance, both acquired and environment-mediated drug resistance (EMDR). Here, using neuroblastoma (NB), a childhood cancer with high incidence of recurrence due to resistance to chemotherapy, as a model we show that human bone marrow-mesenchymal stromal cells induce tumor expression of sphingosine-1-phosphate receptor-1 (S1PR1), leading to their resistance to chemotherapy. Targeting S1PR1 by shRNA markedly enhances etoposide-induced apoptosis in NB cells and abrogates EMDR, while overexpression of S1PR1 significantly protects NB cells from multidrug-induced apoptosis via activating JAK-STAT3 signaling. Elevated S1PR1 expression and STAT3 activation are also observed in human NB cells with acquired resistance to etoposide. We show in vitro and in human NB xenograft models that treatment with FTY720, an FDA-approved drug and antagonist of S1PR1, dramatically sensitizes drug-resistant cells to etoposide. In summary, we identify S1PR1 as a critical target for reducing both EMDR and acquired chemoresistance in NB. Mol Cancer Ther; 16(11); 2516-27. ©2017 AACR.


Assuntos
Etoposídeo/administração & dosagem , Interação Gene-Ambiente , Neuroblastoma/tratamento farmacológico , Receptores de Lisoesfingolipídeo/genética , Fator de Transcrição STAT3/genética , Animais , Apoptose/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Cloridrato de Fingolimode/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Neuroblastoma/genética , Neuroblastoma/patologia , RNA Interferente Pequeno , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Esfingosina-1-Fosfato , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Cancer Res ; 77(18): 5118-5128, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28716895

RESUMO

CTL-associated antigen 4 (CTLA4) is a well-established immune checkpoint for antitumor immune responses. The protumorigenic function of CTLA4 is believed to be limited to T-cell inhibition by countering the activity of the T-cell costimulating receptor CD28. However, as we demonstrate here, there are two additional roles for CTLA4 in cancer, including via CTLA4 overexpression in diverse B-cell lymphomas and in melanoma-associated B cells. CTLA4-CD86 ligation recruited and activated the JAK family member Tyk2, resulting in STAT3 activation and expression of genes critical for cancer immunosuppression and tumor growth and survival. CTLA4 activation resulted in lymphoma cell proliferation and tumor growth, whereas silencing or antibody-blockade of CTLA4 in B-cell lymphoma tumor cells in the absence of T cells inhibits tumor growth. This inhibition was accompanied by reduction of Tyk2/STAT3 activity, tumor cell proliferation, and induction of tumor cell apoptosis. The CTLA4-Tyk2-STAT3 signal pathway was also active in tumor-associated nonmalignant B cells in mouse models of melanoma and lymphoma. Overall, our results show how CTLA4-induced immune suppression occurs primarily via an intrinsic STAT3 pathway and that CTLA4 is critical for B-cell lymphoma proliferation and survival. Cancer Res; 77(18); 5118-28. ©2017 AACR.


Assuntos
Linfócitos B/patologia , Biomarcadores Tumorais/metabolismo , Antígeno CTLA-4/metabolismo , Linfoma de Células B/patologia , Fator de Transcrição STAT3/metabolismo , TYK2 Quinase/metabolismo , Adulto , Idoso , Animais , Apoptose , Linfócitos B/imunologia , Linfócitos B/metabolismo , Antígenos CD28/metabolismo , Proliferação de Células , Feminino , Humanos , Ativação Linfocitária , Linfoma de Células B/imunologia , Linfoma de Células B/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Transdução de Sinais , Linfócitos T/imunologia , Células Tumorais Cultivadas
20.
Nat Rev Cancer ; 14(11): 736-46, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25342631

RESUMO

The Janus kinases (JAKs) and signal transducer and activator of transcription (STAT) proteins, particularly STAT3, are among the most promising new targets for cancer therapy. In addition to interleukin-6 (IL-6) and its family members, multiple pathways, including G-protein-coupled receptors (GPCRs), Toll-like receptors (TLRs) and microRNAs were recently identified to regulate JAK-STAT signalling in cancer. Well known for its role in tumour cell proliferation, survival, invasion and immunosuppression, JAK-STAT3 signalling also promotes cancer through inflammation, obesity, stem cells and the pre-metastatic niche. In addition to its established role as a transcription factor in cancer, STAT3 regulates mitochondrion functions, as well as gene expression through epigenetic mechanisms. Newly identified regulators and functions of JAK-STAT3 in tumours are important targets for potential therapeutic strategies in the treatment of cancer.


Assuntos
Neoplasias/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Animais , Antineoplásicos/uso terapêutico , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa