Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Cancer Res ; 74(18): 5218-28, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25047528

RESUMEN

Understanding supports for cancer stem-like cells in malignant glioma may suggest therapeutic strategies for their elimination. Here, we show that the Toll-like receptor TLR9 is elevated in glioma stem-like cells (GSC) in which it contributes to glioma growth. TLR9 overexpression is regulated by STAT3, which is required for GSC maintenance. Stimulation of TLR9 with a CpG ligand (CpG ODN) promoted GSC growth, whereas silencing TLR9 expression abrogated GSC development. CpG-ODN treatment induced Frizzled4-dependent activation of JAK2, thereby activating STAT3. Targeted delivery of siRNA into GSC was achieved via TLR9 using CpG-siRNA conjugates. Through local or systemic treatment, administration of CpG-Stat3 siRNA to silence STAT3 in vivo reduced GSC along with glioma growth. Our findings identify TLR9 as a functional marker for GSC and a target for the delivery of efficacious therapeutics for glioma treatment. Cancer Res; 74(18); 5218-28. ©2014 AACR.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular , Glioma/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID
11.
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
12.
PLoS One ; 8(5): e64159, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23734190

RESUMEN

The role of B cells in cancer and the underlying mechanisms remain to be further explored. Here, we show that tumor-associated B cells with activated STAT3 contribute to tumor development by promoting tumor angiogenesis. B cells with or without Stat3 have opposite effects on tumor growth and tumor angiogenesis in both B16 melanoma and Lewis Lung Cancer mouse models. Ex vivo angiogenesis assays show that B cell-mediated tumor angiogenesis is mainly dependent on the induction of pro-angiogenic gene expression, which requires Stat3 signaling in B cells. Furthermore, B cells with activated STAT3 are mainly found in or near tumor vasculature and correlate significantly with overall STAT3 activity in human tumors. Moreover, the density of B cells in human tumor tissues correlates significantly with expression levels of several STAT3-downstream pro-angiogenic genes, as well as the degree of tumor angiogenesis. Together, these findings define a novel role of B cells in promoting tumor progression through angiogenesis and identify STAT3 in B cells as potential therapeutic target for anti-angiogenesis therapy.


Asunto(s)
Linfocitos B/inmunología , Neoplasias Experimentales/inmunología , Neovascularización Patológica/inmunología , Factor de Transcripción STAT3/inmunología , Animales , Linfocitos B/metabolismo , Western Blotting , Línea Celular Tumoral , Progresión de la Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Carga Tumoral/genética , Carga Tumoral/inmunología
13.
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
14.
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
15.
PLoS One ; 8(1): e54029, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23326565

RESUMEN

BACKGROUND: Several previous studies have identified a strong association between T-cell infiltration and clinical outcome in ovarian cancer. The role of B-cells remains controversial, however. METHODS: Forty-nine paraffin-embedded omental specimens derived from patients with high grade epithelial ovarian cancer were assessed. Immunohistochemical analyses were performed to characterize expression of CD19(+) B-cells and pSTAT3 as high (>50% positively staining cells [PSCs]) or low (<50% PSCs). The Kaplan-Meier method with log-rank test was used to determine the association between clinicopathologic parameters and overall survival (OS). A multi-variate Cox proportional hazards regression analysis including nature of debulking (primary vs secondary), histology, tumor grade, receipt of prior chemotherapy, B-cell infiltration and pSTAT3 expression was performed. RESULTS: Median OS was 160.6 months in those patients with low B-cell expression vs 47.3 months in those with high B-cell expression (P = 0.0015). Similarly, median OS was improved in those patients with low pSTAT3 expression (160.6 vs 47.9 months, P = 0.02). In a multivariate model to predict survival, only the degree of B-cell infiltration and clinical stage were retained. pSTAT3 expression did not enter the final model, possibly be due to a high positive correlation with B-cell infiltration (r = 0.82, P<0.0001). CONCLUSIONS: Increased B-cell infiltration and pSTAT3 expression in omental tissue are associated with poorer survival.


Asunto(s)
Linfocitos B , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Factor de Transcripción STAT3/genética , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos B/metabolismo , Linfocitos B/patología , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Ováricas/metabolismo , Pronóstico , Modelos de Riesgos Proporcionales , Factor de Transcripción STAT3/metabolismo
16.
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
17.
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
18.
Cancer Cell ; 21(5): 642-654, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22624714

RESUMEN

Recent studies underscore the importance of myeloid cells in rendering distant organs hospitable for disseminating tumor cells to colonize. However, what enables myeloid cells to have an apparently superior capacity to colonize distant organs is unclear. Here, we show that S1PR1-STAT3 upregulation in tumor cells induces factors that activate S1PR1-STAT3 in various cells in premetastatic sites, leading to premetastatic niche formation. Targeting either S1PR1 or STAT3 in myeloid cells disrupts existing premetastatic niches. S1PR1-STAT3 pathway enables myeloid cells to intravasate, prime the distant organ microenvironment and mediate sustained proliferation and survival of their own and other stromal cells at future metastatic sites. Analyzing tumor-free lymph nodes from cancer patients shows elevated myeloid infiltrates, STAT3 activity, and increased survival signal.


Asunto(s)
Células Mieloides/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias de la Próstata/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Microambiente Tumoral , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Islas de CpG , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Metástasis Linfática , Masculino , Melanoma/metabolismo , Melanoma/secundario , Ratones , Ratones Noqueados , Células Mieloides/patología , Invasividad Neoplásica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Interferencia de ARN , Receptores de Lisoesfingolípidos/deficiencia , Receptores de Lisoesfingolípidos/genética , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Receptores de Esfingosina-1-Fosfato , Factores de Tiempo , Transducción Genética , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
19.
Cancer Res ; 71(11): 3772-80, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21502401

RESUMEN

Both STAT3 and NF-κB are persistently activated in diverse cancers and promote tumor cell proliferation, survival, angiogenesis, and metastasis through transcriptional activation of multiple common genes. Paradoxically, STAT3 also suppresses many NF-κB-inducible genes involved in innate and adaptive antitumor immunity in spite of elevated levels of NF-κB in tumors. In this study, we show that expression of many NF-κB downstream target genes in tumors depends on STAT3 DNA binding. When STAT3 is elevated in tumor cells and tumor-infiltrating immune cells, persistently activated NF-κB interacts with STAT3 and preferentially binds to genes with STAT3-binding site(s) in promoters. A large number of NF-κB downstream genes associated with oncogenesis and chronic inflammation contain STAT3 DNA-binding site(s). However, in contrast, many genes frequently associated with antitumor immunity lack STAT3 DNA-binding site(s) and can only be activated by NF-κB when STAT3 is inhibited in tumors. The introduction of STAT3 DNA-binding sequences by site-specific mutagenesis in an immunostimulatory gene promoter allows its transcriptional activation by NF-κB in tumor cells. Furthermore, STAT3 facilitates NF-κB binding to genes that are important for tumor growth while inhibiting its binding to Th-1 immunostimulatory genes in growing tumors, including in tumor-infiltrating immune cells. The results of this study provide insight into how some of the oncogenic/inflammatory and Th-1 immunostimulatory genes are differentially regulated in cancer.


Asunto(s)
ADN de Neoplasias/inmunología , Melanoma/inmunología , FN-kappa B/inmunología , Factor de Transcripción STAT3/inmunología , Células TH1/fisiología , Animales , Sitios de Unión , Procesos de Crecimiento Celular/genética , Procesos de Crecimiento Celular/inmunología , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Células TH1/inmunología , Transcripción Genética , Activación Transcripcional
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...