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

RESUMEN

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.


Asunto(s)
Linfocitos B/inmunología , Antígenos CD5/metabolismo , Interleucina-6/metabolismo , Melanoma Experimental/patología , Factor de Transcripción STAT3/inmunología , Animales , Antígenos CD5/biosíntesis , Línea Celular Tumoral , Receptor gp130 de Citocinas/metabolismo , Humanos , Interleucina-6/inmunología , Janus Quinasa 2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Unión Proteica , Receptores de Interleucina-6/biosíntesis , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/inmunología , Activación Transcripcional/inmunología
3.
Proc Natl Acad Sci U S A ; 110(32): 13079-84, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23878227

RESUMEN

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.


Asunto(s)
Resistencia a la Insulina/inmunología , Grasa Intraabdominal/inmunología , Obesidad/inmunología , Factor de Transcripción STAT3/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Glucemia/metabolismo , Western Blotting , Dieta Alta en Grasa/efectos adversos , Ayuno/sangre , Femenino , Citometría de Flujo , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Insulina/sangre , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-6/inmunología , Interleucina-6/metabolismo , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/sangre , Obesidad/etiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células TH1/inmunología , Células TH1/metabolismo
4.
Nucleic Acids Res ; 41(11): e117, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23598999

RESUMEN

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/.


Asunto(s)
Algoritmos , Islas de CpG , Metilación de ADN , Neoplasias de la Mama/genética , Línea Celular Tumoral , Femenino , Expresión Génica , Genómica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Secuencia de ADN , Sulfitos
5.
Proc Natl Acad Sci U S A ; 109(20): 7765-9, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22547799

RESUMEN

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.


Asunto(s)
Metilación de ADN/genética , Genes Supresores de Tumor , Regiones Promotoras Genéticas/genética , Factor de Transcripción STAT3/metabolismo , Estilbenos/farmacología , Acetilación , Análisis de Varianza , Animales , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Metilación de ADN/efectos de los fármacos , Humanos , Inmunoprecipitación , Ratones , Microscopía Confocal , Microscopía Fluorescente , Reacción en Cadena en Tiempo Real de la Polimerasa , Resveratrol , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética
6.
J Biol Chem ; 288(19): 13842-9, 2013 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-23548897

RESUMEN

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.


Asunto(s)
Hormonas Gastrointestinales/genética , Leucemia Mieloide Aguda/metabolismo , Células Mieloides/metabolismo , Neuropéptidos/genética , Factor de Transcripción STAT3/metabolismo , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Retroalimentación Fisiológica , Hormonas Gastrointestinales/metabolismo , Hormonas Gastrointestinales/fisiología , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Janus Quinasa 2/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Trasplante de Neoplasias , Neuropéptidos/metabolismo , Neuropéptidos/fisiología , ARN Interferente Pequeño/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Carga Tumoral
7.
Blood ; 120(7): 1458-65, 2012 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-22745305

RESUMEN

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.


Asunto(s)
Linfocitos B/inmunología , Activación de Linfocitos/inmunología , Linfoma de Células B Grandes Difuso/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Apoptosis/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod , Silenciador del Gen/efectos de los fármacos , Humanos , Activación de Linfocitos/efectos de los fármacos , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/patología , Ratones , Invasividad Neoplásica , Fosforilación/efectos de los fármacos , Glicoles de Propileno/farmacología , ARN Interferente Pequeño/metabolismo , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato
8.
Curr Top Microbiol Immunol ; 344: 41-59, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20517723

RESUMEN

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.


Asunto(s)
Neoplasias/inmunología , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/fisiología , Animales , Humanos , Tolerancia Inmunológica , Neoplasias/terapia , Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal , Células TH1/inmunología
9.
J Clin Invest ; 118(10): 3367-77, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18776941

RESUMEN

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.


Asunto(s)
Células Endoteliales/metabolismo , Células Mieloides/metabolismo , Neoplasias/irrigación sanguínea , Neovascularización Patológica/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Línea Celular Tumoral , Colágeno/metabolismo , Regulación de la Expresión Génica , Humanos , Análisis de los Mínimos Cuadrados , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/fisiopatología , Ratones , Ratones Endogámicos C57BL
10.
Oncogene ; 39(10): 2156-2169, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31819166

RESUMEN

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.


Asunto(s)
Metilación de ADN , Dioxigenasas/metabolismo , Epigénesis Genética , Glioma/genética , Integrina alfa6/metabolismo , Células Madre Neoplásicas/metabolismo , Factor de Transcripción STAT3/metabolismo , 5-Metilcitosina , Animales , Línea Celular , Dioxigenasas/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/enzimología , Glioma/metabolismo , Humanos , Masculino , Ratones Desnudos , Células Madre Neoplásicas/enzimología , Transducción de Señal
11.
Cell Metab ; 31(1): 148-161.e5, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31761565

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Carcinogénesis/inmunología , Ácidos Grasos/metabolismo , Obesidad/metabolismo , Factor de Transcripción STAT3/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular , Proliferación Celular/genética , Inmunoprecipitación de Cromatina , Femenino , Glucólisis/genética , Glucólisis/fisiología , Humanos , Interferón gamma/metabolismo , Leptina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Obesidad/inmunología , Oxidación-Reducción/efectos de los fármacos , Receptor de Muerte Celular Programada 1/metabolismo , Factor de Transcripción STAT3/genética
12.
Mol Cancer Res ; 6(7): 1099-105, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18644974

RESUMEN

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.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células Mieloides/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Hipoxia de la Célula , Línea Celular Transformada , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Células del Estroma/metabolismo
13.
Mol Cell Biol ; 26(14): 5259-69, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16809764

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Represoras/metabolismo , Telomerasa/metabolismo , Ubiquitina/metabolismo , Acetilación , Secuencia de Bases , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , ADN Complementario/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Silenciador del Gen , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/metabolismo , Células HeLa , Histona Desacetilasas/química , Histona Desacetilasas/genética , Humanos , Técnicas In Vitro , Complejos Multiproteicos , Fosforilación , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/química , Proteínas Represoras/genética , Telomerasa/química , Telomerasa/genética , Técnicas del Sistema de Dos Híbridos , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
14.
Cell Metab ; 27(1): 136-150.e5, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29249690

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/patología , Autorrenovación de las Células , Resistencia a Antineoplásicos , Quinasas Janus/metabolismo , Células Madre Neoplásicas/patología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Adipocitos/metabolismo , Anciano , Animales , Neoplasias de la Mama/genética , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Línea Celular Tumoral , Ácidos Grasos/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Leptina/metabolismo , Metabolismo de los Lípidos/genética , Metabolómica , Ratones , Persona de Mediana Edad , Células Madre Neoplásicas/metabolismo , Oxidación-Reducción , Transcripción Genética
15.
Mol Cell Biol ; 22(16): 5835-45, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12138194

RESUMEN

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.


Asunto(s)
Transporte Activo de Núcleo Celular/efectos de los fármacos , Estradiol/farmacología , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores de Estrógenos/metabolismo , Tamoxifeno/análogos & derivados , Transporte Activo de Núcleo Celular/fisiología , Adenocarcinoma/fisiopatología , Secuencia de Aminoácidos , Animales , Neoplasias Endometriales/fisiopatología , Activación Enzimática , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno , Ácidos Grasos Insaturados/farmacología , Femenino , Humanos , Ligandos , Datos de Secuencia Molecular , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Señales de Clasificación de Proteína , Receptores de Estrógenos/genética , Transducción de Señal/fisiología , Tamoxifeno/farmacología , Treonina/metabolismo , Transcripción Genética , Proteínas Quinasas p38 Activadas por Mitógenos
16.
Mol Cell Biol ; 24(2): 765-73, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14701748

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Represoras/metabolismo , Acetilación , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión/genética , Dominio Catalítico/genética , ADN Complementario/genética , Células HeLa , Histona Desacetilasas/química , Histona Desacetilasas/genética , Histonas/química , Histonas/metabolismo , Humanos , Técnicas In Vitro , Datos de Secuencia Molecular , Fosforilación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/química , Proteínas Represoras/genética , Serina/química
17.
Mol Cell Biol ; 23(1): 104-18, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12482965

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Receptores Androgénicos/metabolismo , Factores de Transcripción/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Sitios de Unión , Ciclo Celular/fisiología , Proteínas de Unión al ADN/genética , Receptor beta de Estrógeno , Proteína Ligando Fas , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead , Humanos , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Metribolona/farmacología , Mutación , Fosfohidrolasa PTEN , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt , Receptores Androgénicos/efectos de los fármacos , Receptores Androgénicos/genética , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Elementos de Respuesta , Congéneres de la Testosterona/farmacología , Factores de Transcripción/genética , Transcripción Genética , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
18.
Mol Cancer Ther ; 16(11): 2516-2527, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28716816

RESUMEN

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.


Asunto(s)
Etopósido/administración & dosificación , Interacción Gen-Ambiente , Neuroblastoma/tratamiento farmacológico , Receptores de Lisoesfingolípidos/genética , Factor de Transcripción STAT3/genética , Animales , Apoptosis/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Clorhidrato de Fingolimod/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Neuroblastoma/genética , Neuroblastoma/patología , ARN Interferente Pequeño , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Esfingosina-1-Fosfato , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cancer Res ; 77(18): 5118-5128, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28716895

RESUMEN

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.


Asunto(s)
Linfocitos B/patología , Biomarcadores de Tumor/metabolismo , Antígeno CTLA-4/metabolismo , Linfoma de Células B/patología , Factor de Transcripción STAT3/metabolismo , TYK2 Quinasa/metabolismo , Adulto , Anciano , Animales , Apoptosis , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígenos CD28/metabolismo , Proliferación Celular , Femenino , Humanos , Activación de Linfocitos , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Transducción de Señal , Linfocitos T/inmunología , Células Tumorales Cultivadas
20.
Nat Rev Cancer ; 14(11): 736-46, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25342631

RESUMEN

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.


Asunto(s)
Neoplasias/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Antineoplásicos/uso terapéutico , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda