Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Nutr Food Res ; : e2400020, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38934352

RESUMO

Sex hormone-binding globulin (SHBG) is a homodimeric glycoprotein produced by the human liver and secreted into the systemic circulation where it binds with high affinity sex steroids regulating their availability in blood and accessibility to target tissues. Plasma SHBG levels are altered in metabolic disorders such as obesity, anorexia, and insulin resistance. Several reports have shown that diets in terms of total calories or fat, fiber, or protein content can alter plasma SHBG levels. However, there are many components in a diet that can affect SHBG gene expression in the liver. In order to unravel the molecular mechanisms by which diets regulate SHBG production, it would be necessary to analyze single diet components and/or nutritional factors. This review summarizes the recent advances in identifying different nutritional factors regulating SHBG production and the related molecular mechanism, as well as the clinical implications.

2.
J Nutr Biochem ; 85: 108480, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32795655

RESUMO

Obese subjects of all ages and sex have reduced plasma SHBG levels. Whether these low plasma SHBG levels play a role in obesity development is unknown. In the present work we wanted to explore if SHBG overexpression could prevent obesity development induced by high fat diet (HFD). To do so, we fed humanized SHBG transgenic male mice and their wild-type littermates with control diet (CD) or HFD over the course of 8 weeks. The results showed that SHBG overexpression protected against body weight gain and fat accumulation induced by HFD. In addition, SHBG overexpression also abrogated the increase in insulin, leptin and resistin levels, as well as the reduction in adiponectin, induced by HFD. Mechanistically, the SHBG protection against HFD-induced obesity was achieved by stimulating lipolysis in white adipose tissue. Furthermore, we have demonstrated the SHBG cell-autonomous effect using human primary visceral adipocytes. Taking together, our results demonstrate that SHBG overexpression protects against diet-induced obesity and improves the metabolic profile of male mice fed a HFD diet.


Assuntos
Obesidade/genética , Globulina de Ligação a Hormônio Sexual/genética , Regulação para Cima , Animais , Linhagem Celular , Dieta Hiperlipídica , Humanos , Lipólise , Masculino , Camundongos Transgênicos , Obesidade/etiologia , Obesidade/metabolismo , Fatores de Proteção , Globulina de Ligação a Hormônio Sexual/metabolismo
3.
J Clin Endocrinol Metab ; 104(5): 1500-1507, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30496542

RESUMO

CONTEXT: There is emerging evidence that SHBG is substantially reduced in chronic metabolic diseases, including obesity and nonalcoholic fatty liver disease (NAFLD). We have recently reported, through use of in vitro (HepG2 cells) and in vivo (SHBG-C57BL/ksJ-db/db mice) models, that SHBG could play a role in arresting the progression of NAFLD by downregulating lipogenesis. OBJECTIVE: The main aim of this study was to investigate the mechanisms by which SHBG prevents hepatic lipogenesis by examining the relationship between SHBG and a key lipogenic enzyme, such as acetyl-coenzyme A carboxylase (ACC) in the liver of obese persons. PARTICIPANTS AND METHODS: SHBG and ACC mRNA levels, as well as triglyceride content, were analyzed in 41 liver samples from nondiabetic obese patients with NAFLD who had undergone bariatric surgery. We also studied the effect of SHBG overexpression in HepG2 cells cultured under high-glucose conditions. RESULTS: SHBG mRNA and protein levels were lower in patients with metabolic syndrome than in those without metabolic syndrome; however, these differences were significant only for mRNA level. SHBG mRNA levels correlated positively with SHBG protein levels and hepatic triglyceride content. In addition, SHBG mRNA and protein levels correlated negatively with ACC mRNA levels and triglyceride content. Furthermore, SHBG overexpression abrogated the increase in ACC expression induced by high-glucose treatment in HepG2 cells. CONCLUSIONS: Our findings suggest that SHBG plays a role in regulating hepatic lipogenesis by reducing ACC levels. These results suggest a strategy for the treatment of NAFLD.


Assuntos
Acetil-CoA Carboxilase/metabolismo , Fígado Gorduroso/fisiopatologia , Síndrome Metabólica/diagnóstico , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Obesidade/complicações , Globulina de Ligação a Hormônio Sexual/metabolismo , Triglicerídeos/sangue , Acetil-CoA Carboxilase/genética , Adulto , Biomarcadores/análise , Estudos de Casos e Controles , Feminino , Seguimentos , Células Hep G2 , Humanos , Lipogênese , Masculino , Síndrome Metabólica/etiologia , Síndrome Metabólica/metabolismo , Pessoa de Meia-Idade , Prognóstico , Globulina de Ligação a Hormônio Sexual/genética
4.
Sci Rep ; 8(1): 5046, 2018 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-29555933

RESUMO

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

5.
Sci Rep ; 7(1): 12284, 2017 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-28947831

RESUMO

Sex hormone-binding globulin (SHBG) carries sex steroids in blood regulating their bioavailability. Red wine consumption increases plasma SHBG levels, and we have discovered that resveratrol, a polyphenol enriched in red wine, acts specifically through the human constitutive androstane receptor (CAR), a drug/xenobiotic detoxification gene regulator, to increase hepatic SHBG production. Chromatin immunoprecipitation and luciferase reporter gene assays show that human CAR binds to a typical direct repeat 1 nuclear hormone receptor-binding element in the human SHBG proximal promoter. Resveratrol also increased hepatic SHBG production in humanized SHBG/CAR transgenic mice. Moreover, SHBG expression correlated significantly with CAR mRNA levels in human liver biopsies. We conclude that the beneficial effects of red wine on the metabolic syndrome and it associated co-morbidities, including cardiovascular disease and type 2 diabetes, may be mediated in part by resveratrol acting via CAR to increase plasma SHBG levels.


Assuntos
Consumo de Bebidas Alcoólicas/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Resveratrol/administração & dosagem , Globulina de Ligação a Hormônio Sexual/metabolismo , Vinho , Consumo de Bebidas Alcoólicas/sangue , Animais , Biópsia , Doenças Cardiovasculares/prevenção & controle , Receptor Constitutivo de Androstano , Meios de Cultura/química , Diabetes Mellitus Tipo 2/prevenção & controle , Feminino , Genes Reporter , Células Hep G2 , Humanos , Fígado/patologia , Luciferases , Masculino , Síndrome Metabólica/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Citoplasmáticos e Nucleares/sangue , Receptores Citoplasmáticos e Nucleares/genética , Globulina de Ligação a Hormônio Sexual/análise , Globulina de Ligação a Hormônio Sexual/genética
6.
Endocrinology ; 158(3): 545-559, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28359088

RESUMO

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.


Assuntos
Lipogênese , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/etiologia , PPAR gama/metabolismo , Globulina de Ligação a Hormônio Sexual/metabolismo , Animais , Regulação para Baixo , Feminino , Frutose/efeitos adversos , Células Hep G2 , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Hepatopatia Gordurosa não Alcoólica/metabolismo
7.
Endocrinology ; 156(12): 4571-81, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26441241

RESUMO

Low plasma sex hormone-binding globulin (SHBG) levels in overweight individuals are a biomarker for the metabolic syndrome and are predictive of type 2 diabetes and cardiovascular disease risk. There are no in vivo models to study SHBG expression and regulation during obesity development. The main reason for this is that the obesity-prone rodent models cannot be used to study this issue, because rodents, unlike humans, do not express the SHBG gene in their livers. We have developed a unique mouse model that expresses the human SHBG, and it develops obesity, by crossing the human SHBG transgenic mice with the C57BL/ksJ-db/db mice. The results obtained with the SHBG-C57BL/ksJ-db/db mouse model have allowed us to determine that the SHBG overexpression in the C57BL/ksJ-db/db reduced the body weight gain but did not change the metabolic profile of these mice. Moreover, we elucidated the molecular mechanisms and transcription factors causing the SHBG down-regulation during obesity development, which involved changes in liver hepatocyte nuclear factor 4α and peroxisome proliferator-activated receptor-γ mRNA and protein levels. Furthermore, these results were confirmed using human liver biopsies. Importantly, we also showed that this model resembles what occurs in human obese subjects, because plasma SHBG and total testosterone levels where reduced in obese mice when compared with lean mice. Future research using this unique mouse model will determine the role of SHBG in the development and progression of obesity, type 2 diabetes, or fatty liver disease.


Assuntos
Fator 4 Nuclear de Hepatócito/genética , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/genética , Obesidade/genética , PPAR gama/genética , Globulina de Ligação a Hormônio Sexual/genética , Adulto , Animais , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/metabolismo , Obesidade/metabolismo , PPAR gama/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Globulina de Ligação a Hormônio Sexual/metabolismo
8.
Trends Endocrinol Metab ; 26(7): 376-83, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26044465

RESUMO

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.


Assuntos
Globulina de Ligação a Hormônio Sexual/metabolismo , Biomarcadores/metabolismo , Humanos , Insulina/metabolismo , Interleucina-1beta/metabolismo , Fígado/metabolismo , Monossacarídeos/metabolismo , Obesidade/metabolismo , Palmitatos/metabolismo
9.
Endocrinology ; 155(8): 2820-30, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24828613

RESUMO

Epidemiological studies have shown that plasma SHBG levels correlate with plasma adiponectin levels, both in men and women. There are no reports describing any molecular mechanism by which adiponectin regulates hepatic SHBG production. The aim of the present study is to explore whether adiponectin regulates SHBG production by increasing HNF-4α levels through reducing hepatic lipid content. For this purpose, in vitro studies using human HepG2 cells, as well as human liver biopsies, were performed. Our results show that adiponectin treatment increased SHBG production via AMPK activation in HepG2 cells. Adiponectin treatment decreased the mRNA and protein levels of enzymes related to hepatic lipogenesis (ACC) and increased those related to fatty acid oxidation (ACOX and CPTI). These adiponectin-induced changes in hepatic enzymes resulted in a reduction of total TG and FFA and an increase of HNF-4α. When HNF-4α expression was silenced by using siRNA, adiponectin-induced SHBG overexpression was blocked. Furthermore, adiponectin-induced upregulation of SHBG production via HNF-4α overexpression was abrogated by the inhibition of fatty acid oxidation or by the induction of lipogenesis with a 30mM glucose treatment in HepG2 cells. Finally, adiponectin levels correlated positively and significantly with both HNF-4α and SHBG mRNA levels in human liver biopsies. Our results suggest for the first time that adiponectin increases SHBG production by activating AMPK, which reduces hepatic lipid content and increases HNF-4α levels.


Assuntos
Adiponectina/química , Adiponectina/fisiologia , Globulina de Ligação a Hormônio Sexual/biossíntese , Globulina de Ligação a Hormônio Sexual/química , Regulação para Cima/fisiologia , Proteínas Quinases Ativadas por AMP/metabolismo , Ácidos Graxos/metabolismo , Células Hep G2 , Fator 4 Nuclear de Hepatócito/química , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Metabolismo dos Lipídeos , Fígado/química , Fígado/metabolismo , Masculino , Obesidade/metabolismo , Oxirredução , Globulina de Ligação a Hormônio Sexual/metabolismo
10.
PLoS One ; 9(1): e85753, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465683

RESUMO

Overproduction of zinc-α2-glycoprotein by adipose tissue is crucial in accounting for the lipolysis occurring in cancer cachexia of certain malignant tumors. The main aim of this study was to explore whether thyroid hormone could enhance zinc-α2-glycoprotein production in adipose tissue. In addition, the regulation of zinc-α2-glycoprotein by thyroid hormone in the liver was investigated. We performed in vitro (HepG2 cells and primary human adipocytes) and in vivo (C57BL6/mice) experiments addressed to examine the effect of thyroid hormone on zinc-α2-glycoprotein production (mRNA and protein levels) in liver and visceral adipose tissue. We also measured the zinc-α2-glycoprotein serum levels in a cohort of patients before and after controlling their hyperthyroidism. Our results showed that thyroid hormone up-regulates zinc-α2-glycoprotein production in HepG2 cells in a dose-dependent manner. In addition, the zinc-α2-glycoprotein proximal promoter contains functional thyroid hormone receptor binding sites that respond to thyroid hormone treatment in luciferase reporter gene assays in HepG2 cells. Furthermore, zinc-α2-glycoprotein induced lipolysis in HepG2 in a dose-dependent manner. Our in vivo experiments in mice confirmed the up-regulation of zinc-α2-glycoprotein induced by thyroid hormone in the liver, thus leading to a significant increase in zinc-α2-glycoprotein circulating levels. However, thyroid hormone did not regulate zinc-α2-glycoprotein production in either human or mouse adipocytes. Finally, in patients with hyperthyroidism a significant reduction of zinc-α2-glycoprotein serum levels was detected after treatment but was unrelated to body weight changes. We conclude that thyroid hormone up-regulates the production of zinc-α2-glycoprotein in the liver but not in the adipose tissue. The neutral effect of thyroid hormones on zinc-α2-glycoprotein expression in adipose tissue could be the reason why zinc-α2-glycoprotein is not related to weight loss in hyperthyroidism.


Assuntos
Tecido Adiposo/metabolismo , Fígado/metabolismo , Proteínas de Plasma Seminal/biossíntese , Tri-Iodotironina/farmacologia , Regulação para Cima/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Animais , Sequência de Bases , Peso Corporal/efeitos dos fármacos , Feminino , Células Hep G2 , Humanos , Hipertireoidismo/sangue , Hipertireoidismo/fisiopatologia , Lipólise/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Proteínas de Plasma Seminal/sangue , Proteínas de Plasma Seminal/genética , Proteínas de Plasma Seminal/metabolismo , Testes de Função Tireóidea , Glicoproteína Zn-alfa-2
11.
Acta Diabetol ; 51(4): 543-52, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24458975

RESUMO

Liver and muscle glycogen content is reduced in diabetic patients but there is no information on the effect of diabetes on the glycogen content in the retinal pigment epithelium (RPE). The main aim of the study was to compare the glycogen content in the RPE between diabetic and non-diabetic human donors. Glycogen synthase (GS) and glycogen phosphorylase (GP), the key enzymes of glycogen metabolism, as well as their isoforms, were also assessed. For this purpose, 44 human postmortem eye cups were included (22 from 11 type 2 diabetic and 22 from 11 non-diabetic donors matched by age). Human RPE cells cultured in normoglycemic and hyperglycemic conditions were also analyzed. Glycogen content as well as the mRNA, protein content and enzyme activity of GS and GP were determined. In addition, GS and GP isoforms were characterized. In the RPE from diabetic donors, as well as in RPE cells grown in hyperglycemic conditions, the glycogen content was increased. The increase in glycogen content was associated with an increase in GS without changes in GP levels. In RPE form human donors, the muscle GS isoform but not the liver GS isoform was detected. Regarding GP, the muscle and brain isoform of GP but not the liver GP isoform were detected. We conclude that glycogen storage is increased in the RPE of diabetic patients, and it is associated with an increase in GS activity. Further studies aimed at determining the role of glycogen deposits in the pathogenesis of diabetic retinopathy are warranted.


Assuntos
Diabetes Mellitus/metabolismo , Glicogênio/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Idoso , Idoso de 80 Anos ou mais , Diabetes Mellitus/enzimologia , Diabetes Mellitus/genética , Feminino , Glicogênio Fosforilase/genética , Glicogênio Fosforilase/metabolismo , Glicogênio Sintase/genética , Glicogênio Sintase/metabolismo , Humanos , Fígado/enzimologia , Fígado/metabolismo , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/enzimologia , Músculo Esquelético/metabolismo , Epitélio Pigmentado da Retina/enzimologia , Doadores de Tecidos
12.
Mol Nutr Food Res ; 58(4): 760-7, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24142580

RESUMO

SCOPE: Low circulating sex hormone-binding globulin (SHBG) is an independent risk factor for cardiovascular disease. Mediterranean diet has been associated with a decreased risk of cardiovascular disease. We aimed to test the hypothesis that the increase of circulating MUFA associated with olive oil consumption (primary fat source in Mediterranean diet) increases SHBG serum levels. METHODS AND RESULTS: A total of 315 men were included. In these patients, nutrition data and plasma samples for SHBG assessment were obtained. In vitro studies to examine the effects of oleic and linoleic acid on SHBG production using HepG2 cells were performed. We provided evidence that SHBG serum levels were significantly higher in subjects using olive oil for cooking in comparison with subjects using sunflower oil. The SHBG levels correlated positively with MUFA (p < 0.001) and negatively with saturated fatty acids (p = 0.003). In the multiple regression analysis, MUFA were independently associated with SHBG levels and accounted for the 20.4% of SHBG variance. In vitro studies revealed that oleoyl-CoA increases SHBG production by downregulating PPAR-γ levels in HepG2 cells. CONCLUSION: Olive oil consumption is associated with elevated SHBG serum levels. PPAR-γ downregulation induced by oleoyl-CoA is an important underlying mechanism of such regulation.


Assuntos
Dieta Mediterrânea , Ácido Oleico/farmacologia , Globulina de Ligação a Hormônio Sexual/análise , Acil Coenzima A/farmacologia , Adulto , Culinária , Ácidos Graxos Monoinsaturados/sangue , Ácidos Graxos Monoinsaturados/farmacologia , Células Hep G2/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Azeite de Oliva , PPAR gama/metabolismo , Óleos de Plantas , Análise de Regressão , Óleo de Girassol
13.
Mol Endocrinol ; 26(3): 438-46, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22301786

RESUMO

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.


Assuntos
Regulação para Baixo , Regulação da Expressão Gênica , Globulina de Ligação a Hormônio Sexual/genética , Fator de Necrose Tumoral alfa/fisiologia , Sequência de Bases , Sítios de Ligação , Genes Reporter , Células Hep G2 , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Regiões Promotoras Genéticas , Ligação Proteica , Globulina de Ligação a Hormônio Sexual/metabolismo , Fator de Transcrição RelA/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA