Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Am J Physiol Cell Physiol ; 305(3): C355-9, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23720021

RESUMEN

Evidence from the literature suggests that testosterone plays an important role in visceral fat accumulation since both men and women with hyperandrogenism accumulate more adipose tissue in the abdominal cavity than healthy women. However, the underlying mechanisms remain to be elucidated. To shed light on this issue, we have used an in vitro approach to examine the effect of testosterone on human visceral preadipocyte proliferation. Our results showed that testosterone treatment significantly increased proliferation of human visceral preadipocytes in proliferation assays using flow cytometric analysis. We next performed a microarray gene expression analysis of human visceral preadipocytes treated with testosterone or vehicle to identify which genes were involved in the testosterone-induced increase in preadipocyte proliferation. The results showed a total of 140 genes differentially expressed between testosterone vs. vehicle. Among the top 10 upregulated genes, 5 were involved in cellular cycle and proliferation, and 3 (APOBEC3b, CCNA2, and PRC1) were significantly overexpressed by testosterone treatment when analyzed by real-time PCR. We conclude that testosterone exerts a proliferative effect on preadipocytes that may participate in the sex differences in fat distribution and that it may explain visceral fat accumulation in women with hyperandrogenism.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Andrógenos/farmacología , Testosterona/farmacología , Tejido Adiposo/crecimiento & desarrollo , Ciclo Celular/genética , Proliferación Celular , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Humanos , Hiperandrogenismo , Grasa Intraabdominal/citología , Grasa Intraabdominal/metabolismo , Masculino
2.
Exp Cell Res ; 315(17): 3004-13, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19447103

RESUMEN

Cancer stem cells are a distinct cellular population that is believed to be responsible for tumor initiation and maintenance. Recent data suggest that solid tumors also contain another type of stem cells, the mesenchymal stem cells or multipotent mesenchymal stromal cells (MSCs), which contribute to the formation of tumor-associated stroma. The Hoechst 33342 efflux assay has proved useful to identify a rare cellular fraction, named Side Population (SP), enriched in cells with stem-like properties. Using this assay, we identified SP cells in a prostate cancer xenograft containing human prostate cancer cells and mouse stromal cells. The SP isolation, subculture and sequential sorting allowed the generation of single-cell-derived clones of murine origin that were recognized as MSC by their morphology, plastic adherence, proliferative potential, adipogenic and osteogenic differentiation ability and immunophenotype (CD45(-), CD81(+) and Sca-1(+)). We also demonstrated that SP clonal cells secrete transforming growth factor beta1 (TGF-beta1) and that their inhibition reduces proliferation and accelerates differentiation. These results reveal the existence of SP cells in the stroma of a cancer xenograft, and provide evidence supporting their MSC nature and the role of TGF-beta1 in maintaining their proliferation and undifferentiated status. Our data also reveal the usefulness of the SP assay to identify and isolate MSC cells from carcinomas.


Asunto(s)
Células Madre Pluripotentes/patología , Neoplasias de la Próstata/patología , Células del Estroma/patología , Animales , Cartilla de ADN , Citometría de Flujo , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo , Microglobulina beta-2/genética
3.
BMC Mol Biol ; 10: 59, 2009 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-19534810

RESUMEN

BACKGROUND: The human Sex Hormone Binding Globulin (SHBG) gene, located at 17p13.1, comprises, at least, two different transcription units regulated by two different promoters. The first transcription unit begins with the exon 1 sequence and is responsible for the production of plasma SHBG by the hepatocytes, while the second begins with an alternative exon 1 sequence, which replaces the exon 1 present in liver transcripts. Alternative exon 1 transcription and translation has only been demonstrated in the testis of transgenic mice containing an 11-kb human SHBG transgene and in the human testis. Our goal has been to further characterize the 5' end of the SHBG gene and analyze the presence of the SHBG alternative transcripts in human prostate tissue and derived cell lines. RESULTS: Using a combination of in silico and in vitro studies, we have demonstrated that the SHBG gene, along with exon 1 and alternative exon 1 (renamed here exon 1A), contains four additional alternative first exons: the novel exons 1B, 1C, and 1E, and a previously identified exon 1N, which has been further characterized and renamed as exon 1D. We have shown that these four alternative first exons are all spliced to the same 3' splice site of SHBG exon 2, and that exon 1A and the novel exon 1B can be spliced to exon 1. We have also demonstrated the presence of SHBG transcripts beginning with exons 1B, 1C and 1D in prostate tissues and cell lines, as well as in several non-prostatic cell lines. Finally, the alignment of the SHBG mammalian sequences revealed that, while exons 1C, 1D and 1E are very well conserved phylogenetically through non-primate mammal species, exon 1B probably aroused in apes due to a single nucleotide change that generated a new 5' splice site in exon 1B. CONCLUSION: The identification of multiple transcription start sites (TSS) upstream of the annotated first exon of human SHBG, and the detection of the alternative transcripts in human prostate, concur with the prediction of the ENCODE (ENCyclopedia of DNA Elements) project, and suggest that the regulation of SHBG is much more complex than previously reported.


Asunto(s)
Empalme Alternativo , Neoplasias de la Próstata/genética , Globulina de Unión a Hormona Sexual/genética , Animales , Secuencia de Bases , Línea Celular Tumoral , Exones , Humanos , Masculino , Datos de Secuencia Molecular , Filogenia , Alineación de Secuencia , Sitio de Iniciación de la Transcripción
4.
Cell Oncol ; 30(4): 349-65, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18607069

RESUMEN

BACKGROUND: It is well known that estrogens regulate cell cycle progression, but the specific contributions and mechanisms of action of the estrogen receptor beta (ERbeta) remain elusive. METHODS: We have analyzed the levels of ERbeta1 and ERbeta2 throughout the cell cycle, as well as the mechanisms of action and the consequences of the over-expression of ERbeta1 in the human prostate cancer LNCaP cell line. RESULTS: Both ERbeta1 mRNA and protein expression increased from the G1 to the S phase and decreased before entering the G2/M phase, whereas ERbeta2 levels decreased during the S phase and increased in the G2/M phase. ERbeta1 protein was detected in both the nuclear and non-nuclear fractions, and ERbeta2 was found exclusively in the nucleus. Regarding the mechanisms of action, endogenous ERbeta was able to activate transcription via ERE during the S phase in a ligand-dependent manner, whereas no changes in AP1 and NFkappaB transactivation were observed after exposure to estradiol or the specific inhibitor ICI 182,780. Over-expression of either wild type ERbeta1 or ERbeta1 mutated in the DNA-binding domain caused an arrest in early G1. This arrest was accompanied by the interaction of over-expressed ERbeta1 with c-Jun N-terminal protein kinase 1 (JNK1) and a decrease in c-Jun phosphorylation and cyclin D1 expression. The administration of ICI impeded the JNK1-ERbeta1 interaction, increased c-Jun phosphorylation and cyclin D1 expression and allowed the cells to progress to late G1, where they became arrested. CONCLUSIONS: Our results demonstrate that, in LNCaP prostate cancer cells, both ERbeta isoforms are differentially expressed during the cell cycle and that ERbeta regulates the G1 phase by a non-genomic mechanism.


Asunto(s)
Receptor beta de Estrógeno , Fase G1/genética , Neoplasias de la Próstata/patología , Fase S/genética , Activación Transcripcional , Sitios de Unión/genética , Línea Celular , Proteínas de Unión al ADN/genética , Estradiol/análogos & derivados , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Receptor beta de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Fulvestrant , Fase G1/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Unión Proteica/efectos de los fármacos , Activación Transcripcional/genética , Transgenes/genética
5.
Endocrinology ; 158(3): 545-559, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28359088

RESUMEN

Low plasma sex hormone-binding globulin (SHBG) levels are a hallmark in chronic metabolic diseases, including nonalcoholic fatty liver disease (NAFLD), which represents a spectrum of disease ranging from hepatocellular steatosis through steatohepatitis to fibrosis and irreversible cirrhosis. The functional link between altered SHBG production and NAFLD development and progression remains unclear. We investigated the effects of overexpressing human SHBG in 2 mouse models of NAFLD: a genetically induced double transgenic mouse and a diet-induced model. Remarkably, SHBG overexpression in both NAFLD models significantly reduced liver fat accumulation by reducing key lipogenic enzymes. These findings were corroborated by modulating SHBG expression and by adding exogenous SHBG in HepG2 cells, suggesting the cell autonomous nature of the mechanism. Mechanistically, exogenous SHBG treatment downregulated key lipogenic enzymes by reducing PPARγ messenger RNA and protein levels through activation of extracellular signal-regulated kinase-1/2 mitogen-activated protein kinase pathway. Taking together, we found that SHBG modulates hepatic lipogenesis. This is of importance because reduction of SHBG plasma levels in obese and type 2 diabetic subjects could be directly associated with NAFLD development through an increase in hepatic lipogenesis. Our results point to SHBG as a therapeutic target for preventing or arresting NAFLD development.


Asunto(s)
Lipogénesis , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , PPAR gamma/metabolismo , Globulina de Unión a Hormona Sexual/metabolismo , Animales , Regulación hacia Abajo , Femenino , Fructosa/efectos adversos , Células Hep G2 , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico/metabolismo
6.
Trends Endocrinol Metab ; 26(7): 376-83, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26044465

RESUMEN

Sex hormone-binding globulin (SHBG) is produced and secreted by the liver into the bloodstream where it binds sex steroids and regulates their bioavailability. Traditionally, body mass index (BMI) was thought to be the major determinant of SHBG concentrations and hyperinsulinemia the main cause for low SHBG levels found in obesity. However, no mechanisms have ever been described. Emerging evidence now shows that liver fat content rather than BMI is a strong determinant of circulating SHBG. In this review we discuss evidence demonstrating that insulin might not regulate SHBG production, describe putative molecular mechanisms by which proinflammatory cytokines downregulate SHBG, and comment on recent findings suggesting dietary SHBG regulation. Finally, clinical implications of all of these findings and future perspectives are discussed.


Asunto(s)
Globulina de Unión a Hormona Sexual/metabolismo , Biomarcadores/metabolismo , Humanos , Insulina/metabolismo , Interleucina-1beta/metabolismo , Hígado/metabolismo , Monosacáridos/metabolismo , Obesidad/metabolismo , Palmitatos/metabolismo
7.
PLoS One ; 8(9): e74179, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24058525

RESUMEN

Type 2 diabetes has been associated with decreased risk of prostate cancer in observational studies, and this inverse association has been recently confirmed in several large cohort studies. However the mechanisms involved in this protective effect remain to be elucidated. The aim of the present study was to explore whether different features of type 2 diabetes (hyperinsulinemia, hyperglycemia and tumor necrosis factor alpha [TNF-α]) protect against the development of prostate cancer. For this purpose LNCaP cells were used for in vitro experiments and nude mice in which PAC120 (hormone-dependent human prostate cancer) xenografts had been implanted were used for in vivo examinations. We provide evidence that increasing glucose concentrations downregulate androgen receptor (AR) mRNA and protein levels through NF-κB activation in LNCaP cells. Moreover, there was a synergic effect of glucose and TNFα in downregulating the AR in LNCaP cells. By contrast, insulin had no effect on AR regulation. In vivo experiments showed that streptozotocin-induced diabetes (STZ-DM) produces tumor growth retardation and a significant reduction in AR expression in PAC120 prostate cancer mice. In conclusion, our results suggest that hyperglycemia and TNF-α play an important role in protecting against prostate cancer by reducing androgen receptor levels via NF-κB.


Asunto(s)
Adenocarcinoma/genética , Diabetes Mellitus Experimental/genética , Glucosa/farmacología , Hiperglucemia/genética , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Factor de Necrosis Tumoral alfa/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma/prevención & control , Animales , Línea Celular Tumoral , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Regulación de la Expresión Génica , Humanos , Hiperglucemia/metabolismo , Hiperglucemia/patología , Insulina/farmacología , Masculino , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Trasplante de Neoplasias , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/prevención & control , Receptores Androgénicos/metabolismo , Transducción de Señal , Estreptozocina , Trasplante Heterólogo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
8.
Mol Endocrinol ; 26(11): 1917-27, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22902540

RESUMEN

Patients suffering from low-grade chronic inflammatory diseases, such as rheumatoid arthritis, osteoarthritis, diabetes, and obesity, have low plasma sex hormone-binding globulin (SHBG) levels. These diseases are characterized among other features by high plasma IL1ß levels. The aim of the present study is to explore whether IL1ß could regulate hepatic SHBG production to account for low SHBG levels in these diseases. We provide evidence that daily IL1ß treatment reduces SHBG production in HepG2 cells by the down-regulation of HNF-4A via the MAPK kinase (MEK)-1/2 and c-Jun N-terminal kinase (JNK) MAPK signaling pathways through the activation c-Jun transcription factors. The human SHBG promoter sequence contains two putative activator protein 1 (AP1) binding sites recognized by c-Jun transcription factors, but they are not necessary for the IL1ß-induced down-regulation of SHBG promoter activity in luciferase reporter gene assays. Daily treatment with IL1ß reduces hepatic nuclear factor (HNF)-4α mRNA and protein levels via the MEK-1/2 and JNK MAPK signaling pathways. Moreover, IL1ß rapidly decreased HNF-4α mRNA and protein levels while increased phospho-c-Jun protein levels after the treatment. Finally, daily IL1ß treatment of human SHBG transgenic mice reduced plasma SHBG and SHBG mRNA levels. Moreover, IL1ß treatment also reduced HNF-4α mRNA and protein levels while increased hepatic phospho-c-Jun protein levels. Our results show that IL1ß reduces hepatic SHBG production by decreasing HNF-4α via MEK-1/2 and JNK MAPK pathways. In addition, our findings suggest that IL1ß could be involved the low plasma SHBG levels reported in chronic low-grade inflammatory diseases.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Factor Nuclear 4 del Hepatocito/metabolismo , Interleucina-1beta/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Globulina de Unión a Hormona Sexual/biosíntesis , Animales , Sitios de Unión/genética , Regulación hacia Abajo/genética , Activación Enzimática/efectos de los fármacos , Genes Reporteros , Células Hep G2 , Factor Nuclear 4 del Hepatocito/genética , Humanos , Luciferasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Globulina de Unión a Hormona Sexual/genética , Testosterona/sangre , Factor de Transcripción AP-1/metabolismo
9.
Diabetes ; 61(2): 372-82, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22210320

RESUMEN

Low plasma sex hormone-binding globulin (SHBG) levels are associated with obesity and predict the development of type 2 diabetes. The reason why obese individuals have low circulating SHBG has been attributed to hyperinsulinemia, but no mechanistic evidence has been described. The aim of the current study is to explore whether tumor necrosis factor-α (TNF-α) rather than insulin could be the main factor accounting for low SHBG levels in obesity. We performed in vitro and in vivo studies using human HepG2 cells and human SHBG transgenic mice. In addition, a cross-sectional study to explore the relationship between TNF-α and SHBG in obese patients and an interventional study to examine the effect of insulin administration on circulating SHBG in type 2 diabetic patients were performed. We provide evidence that TNF-α, but not insulin, is the main factor by which SHBG is reduced in obesity. Plasma SHBG was significantly increased rather than decreased after insulin treatment in diabetic patients. TNF-α-induced reduction of SHBG expression was mediated by downregulating HNF4A. Finally, a negative and independent correlation was found between plasma TNF-α receptor 1 and SHBG levels in obese patients. Our results suggest that TNF-α plays an important role downregulating SHBG in chronic low-grade inflammatory diseases such as obesity and type 2 diabetes.


Asunto(s)
Globulina de Unión a Hormona Sexual/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Regulación hacia Abajo , Células Hep G2 , Factor Nuclear 4 del Hepatocito/análisis , Humanos , Insulina/farmacología , Lipogénesis , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/fisiología , Globulina de Unión a Hormona Sexual/análisis , Globulina de Unión a Hormona Sexual/antagonistas & inhibidores
10.
Mol Endocrinol ; 26(3): 438-46, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22301786

RESUMEN

The reason why obesity (a chronic low-grade inflammatory disease) is associated with low levels of sex hormone-binding globulin (SHBG) remains to be elucidated. The present study provides evidence that TNFα (a proinflammatory cytokine increased in obesity) reduces SHBG production by human HepG2 hepatoblastoma cells. Although the human SHBG promoter contains one nuclear factor-κB (NF-κB) binding site, the human SHBG promoter activity did not change after TNFα treatment or transfection with either small interfering RNA against p65 or a p65 expression vector in luciferase reporter gene assays. The effect of TNFα on human SHBG expression was indirect, and it was mediated by NF-κB through the down-regulation of hepatocyte nuclear factor (HNF)-4A: a key SHBG transcriptional regulator. Furthermore, the HNF-4A proximal promoter contains three putative NF-κB binding sites. The HNF-4A promoter activity was decreased by the treatment with TNFα or the transfection of a p65 expression vector, and it was increased by the treatment with small interfering RNA against NF-κB in luciferase reporter gene assays. Finally, the TNFα treatment promotes the NF-κB binding to the HNF-4A promoter in chromatin immunoprecipitation assays. We conclude that sustained exposition to elevated levels of TNFα decreases SHBG production by reducing hepatic HNF-4α levels via NF-κB activation in HepG2 cells.


Asunto(s)
Regulación hacia Abajo , Regulación de la Expresión Génica , Globulina de Unión a Hormona Sexual/genética , Factor de Necrosis Tumoral alfa/fisiología , Secuencia de Bases , Sitios de Unión , Genes Reporteros , Células Hep G2 , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , Regiones Promotoras Genéticas , Unión Proteica , Globulina de Unión a Hormona Sexual/metabolismo , Factor de Transcripción ReIA/metabolismo , Transcripción Genética
11.
PLoS One ; 5(11): e13844, 2010 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-21079794

RESUMEN

BACKGROUND: The human sex hormone-binding globulin (SHBG) gene comprises at least 6 different transcription units (TU-1, -1A, -1B, -1C, -1D and -1E), and is regulated by no less than 6 different promoters. The best characterized are TU-1 and TU-1A: TU-1 is responsible for producing plasma SHBG, while TU-1A is transcribed and translated in the testis. Transcription of the recently described TU-1B, -1C, and -1D has been demonstrated in human prostate tissue and prostate cancer cell lines, as well as in other human cell lines such as HeLa, HepG2, HeK 293, CW 9019 and imr 32. However, there are no reported data demonstrating their translation. In the present study, we aimed to determine whether TU-1A and TU-1B are indeed translated in the human prostate and whether 5' UTR exons 1A and 1B differently regulate SHBG translation. RESULTS: Cis-regulatory elements that could potentially regulate translation were identified within the 5'UTRs of SHBG TU-1A and TU-1B. Although full-length SHBG TU-1A and TU-1B mRNAs were present in prostate cancer cell lines, the endogenous SHBG protein was not detected by western blot in any of them. LNCaP prostate cancer cells transfected with several SHBG constructs containing exons 2 to 8 but lacking the 5'UTR sequence did show SHBG translation, whereas inclusion of the 5'UTR sequences of either exon 1A or 1B caused a dramatic decrease in SHBG protein levels. The molecular weight of SHBG did not vary between cells transfected with constructs with or without the 5'UTR sequence, thus confirming that the first in-frame ATG of exon 2 is the translation start site of TU-1A and TU-1B. CONCLUSIONS: The use of alternative SHBG first exons 1A and 1B differentially inhibits translation from the ATG situated in exon 2, which codes for methionine 30 of transcripts that begin with the exon 1 sequence.


Asunto(s)
Empalme Alternativo , Exones/genética , Biosíntesis de Proteínas/genética , ARN Mensajero/genética , Globulina de Unión a Hormona Sexual/genética , Regiones no Traducidas 5'/genética , Western Blotting , Línea Celular Tumoral , Codón Iniciador/genética , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Masculino , Metionina/genética , Conformación de Ácido Nucleico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Mensajero/química , Secuencias Reguladoras de Ácidos Nucleicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Globulina de Unión a Hormona Sexual/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA