Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Syst Biol ; 20(4): 374-402, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38459198

RESUMO

Sex-based differences in obesity-related hepatic malignancies suggest the protective roles of estrogen. Using a preclinical model, we dissected estrogen receptor (ER) isoform-driven molecular responses in high-fat diet (HFD)-induced liver diseases of male and female mice treated with or without an estrogen agonist by integrating liver multi-omics data. We found that selective ER activation recovers HFD-induced molecular and physiological liver phenotypes. HFD and systemic ER activation altered core liver pathways, beyond lipid metabolism, that are consistent between mice and primates. By including patient cohort data, we uncovered that ER-regulated enhancers govern central regulatory and metabolic genes with clinical significance in metabolic dysfunction-associated steatotic liver disease (MASLD) patients, including the transcription factor TEAD1. TEAD1 expression increased in MASLD patients, and its downregulation by short interfering RNA reduced intracellular lipid content. Subsequent TEAD small molecule inhibition improved steatosis in primary human hepatocyte spheroids by suppressing lipogenic pathways. Thus, TEAD1 emerged as a new therapeutic candidate whose inhibition ameliorates hepatic steatosis.


Assuntos
Fígado Gorduroso , Hepatopatia Gordurosa não Alcoólica , Animais , Feminino , Humanos , Masculino , Camundongos , Dieta Hiperlipídica/efeitos adversos , Estrogênios , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Expressão Gênica , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores de Estrogênio/uso terapêutico , Fatores de Transcrição de Domínio TEA
2.
Commun Biol ; 5(1): 1057, 2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36195702

RESUMO

Male and female offspring of obese mothers are known to differ extensively in their metabolic adaptation and later development of complications. We investigate the sex-dependent responses in obese offspring mice with maternal obesity, focusing on changes in liver glucose and lipid metabolism. Here we show that maternal obesity prior to and during gestation leads to hepatic steatosis and inflammation in male offspring, while female offspring are protected. Females from obese mothers display important changes in hepatic transcriptional activity and triglycerides profile which may prevent the damaging effects of maternal obesity compared to males. These differences are sustained later in life, resulting in a better metabolic balance in female offspring. In conclusion, sex and maternal obesity drive differently transcriptional and posttranscriptional regulation of major metabolic processes in offspring liver, explaining the sexual dimorphism in obesity-associated metabolic risk.


Assuntos
Obesidade Materna , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Glucose/metabolismo , Humanos , Metabolismo dos Lipídeos , Fígado/metabolismo , Masculino , Camundongos , Mães , Obesidade/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Triglicerídeos/metabolismo
3.
Int J Obes (Lond) ; 46(4): 831-842, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34997206

RESUMO

OBJECTIVE: The prevalence of overweight and obesity among children has drastically increased during the last decades and maternal obesity has been demonstrated as one of the ultimate factors. Nutrition-stimulated transgenerational regulation of key metabolic genes is fundamental to the developmental origins of the metabolic syndrome. Fetal nutrition may differently influence female and male offspring. METHODS: Mice dam were fed either a control diet or a high-fat diet (HFD) for 6-week prior mating and continued their respective diet during gestation and lactation. At weaning, female and male offspring were fed the HFD until sacrifice. White (WAT) and brown (BAT) adipose tissues were investigated in vivo by nuclear magnetic resonance at two different timepoints in life (midterm and endterm) and tissues were collected at endterm for lipidomic analysis and RNA sequencing. We explored the sex-dependent metabolic adaptation and gene programming changes by maternal HFD in visceral AT (VAT), subcutaneous AT (SAT) and BAT of offspring. RESULTS: We show that the triglyceride profile varies between adipose depots, sexes and maternal diet. In female offspring, maternal HFD remodels the triglycerides profile in SAT and BAT, and increases thermogenesis and cell differentiation in BAT, which may prevent metabolic complication later in life. Male offspring exhibit whitening of BAT and hyperplasia in VAT when born from high-fat mothers, with impaired metabolic profile. Maternal HFD differentially programs gene expression in WAT and BAT of female and male offspring. CONCLUSION: Maternal HFD modulates metabolic profile in offspring in a sex-dependent manner. A sex- and maternal diet-dependent gene programming exists in VAT, SAT, and BAT which may be key player in the sexual dimorphism in the metabolic adaptation later in life.


Assuntos
Tecido Adiposo Marrom , Lipidômica , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Dieta Hiperlipídica , Feminino , Humanos , Masculino , Camundongos , Gravidez , Termogênese , Transcriptoma/genética
4.
Commun Biol ; 4(1): 14, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33398027

RESUMO

With the increasing prevalence of obesity in women of reproductive age, there is an urgent need to understand the metabolic impact on the fetus. Sex-related susceptibility to liver diseases has been demonstrated but the underlying mechanism remains unclear. Here we report that maternal obesity impacts lipid metabolism differently in female and male offspring. Males, but not females, gained more weight and had impaired insulin sensitivity when born from obese mothers compared to control. Although lipid mass was similar in the livers of female and male offspring, sex-specific modifications in the composition of fatty acids, triglycerides and phospholipids was observed. These overall changes could be linked to sex-specific regulation of genes controlling metabolic pathways. Our findings revised the current assumption that sex-dependent susceptibility to metabolic disorders is caused by sex-specific postnatal regulation and instead we provide molecular evidence supporting in utero metabolic adaptations in the offspring of obese mothers.


Assuntos
Metabolismo dos Lipídeos , Fígado/metabolismo , Obesidade , Complicações na Gravidez , Efeitos Tardios da Exposição Pré-Natal , Adaptação Fisiológica , Animais , Feminino , Lipidômica , Fígado/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino , Camundongos Endogâmicos C57BL , Gravidez , Caracteres Sexuais
5.
Methods Mol Biol ; 2164: 109-119, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32607888

RESUMO

Fat distribution, on top of general obesity, contributes to the severity of histologic features in patients with nonalcoholic fatty liver diseases (NAFLD); and visceral obesity has been correlated to fatty liver diseases. Therefore, investigation of fat distribution in vivo could be a good predictor of fatty liver risks in obesity. Fatty acids composition is a key player in hepatic dysfunctions and cardiovascular risk in obesity. Because fatty acids can damage biological membranes, fatty acid accumulation in the liver may be partially responsible for the functional and morphological changes that are observed in NAFLD. Fatty acids stored into triglycerides are lipid species that act as signaling molecules and therefore are key regulators of posttranslational regulation of biological functions such as lipid homeostasis and lipotoxicity. Here, we describe magnetic resonance methods to investigate in vivo whole-body fat distribution and hepatic liver fatty acid composition in order to directly assess the liver metabolic status and may allow to anticipate liver diseases.


Assuntos
Distribuição da Gordura Corporal/métodos , Metabolismo dos Lipídeos/fisiologia , Lipídeos/química , Fígado/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Animais , Dieta Hiperlipídica , Ácidos Graxos/metabolismo , Feminino , Lipogênese/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Triglicerídeos/metabolismo
6.
Metabolites ; 10(3)2020 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-32244965

RESUMO

In this communication, we aim to summarize the role of estrogen receptor beta (ERß) in lipid metabolism in the main metabolic organs with a special focus on sex differences. The action of ERß is tissue-specific and acts in a sex-dependent manner, emphasizing the necessity of developing sex- and tissue-selective targeting drugs in the future.

7.
Mol Cell Endocrinol ; 502: 110672, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31811898

RESUMO

Estrogen exerts its action through the binding to two major receptors, estrogen receptor (ER)α and ß. Recently, the beneficial role of selective ERß activation in the regulation of metabolic homeostasis in obesity has been demonstrated, but its importance is still controversial. However, no data are available regarding possible gender differences in response to pharmaceutical activation of ERß. Male mice were fed a control diet (CD) or a high fat diet (HFD) before being treated with the ERß selective ligand, 4-(2-(3-5-dimethylisoxazol-4-yl)-1H-indol-3yl)phenol (DIP) in the same conditions as in our recently published paper in female mice. Magnetic resonance imaging and spectroscopy were performed repeatedly in vivo after 6 weeks of diet and after 2 weeks of DIP. Adipose tissue distribution and hepatic triglycerides composition were quantified. HFD-treated males showed a feminization of their fat distribution towards more subcutaneous fat depots and increase total fat content and visceral adipose tissue showed clear browning sites after DIP. Hepatic lipid composition was modified by DIP, with less saturated and more unsaturated lipids and an improved insulin sensitivity. Finally, brown adipose tissue size expended after DIP, due to an increase of the size of the lipid droplets. Our data demonstrate that selective activation of ERß exerts a tissue-specific and sex-dependent response to metabolic adaptation to overfeeding. Most importantly, together with our previously published results in females, the current findings support the concept that sex should be considered in the future development of obesity-moderating drugs.


Assuntos
Receptor beta de Estrogênio/metabolismo , Obesidade/metabolismo , Fenóis/farmacologia , Triglicerídeos/metabolismo , Gordura Abdominal/metabolismo , Animais , Estudos de Casos e Controles , Dieta Hiperlipídica , Feminino , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Obesos , Obesidade/induzido quimicamente , Caracteres Sexuais , Gordura Subcutânea/metabolismo , Distribuição Tecidual
8.
Biol Sex Differ ; 10(1): 11, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30808418

RESUMO

The response to overfeeding is sex dependent, and metabolic syndrome is more likely associated to obesity in men or postmenopausal women than in young fertile women. We hypothesized that obesity-induced metabolic syndrome is sex dependent due to a sex-specific regulation of the fatty acid (FA) synthesis pathways in liver and white adipose depots. We aimed to identify distinctive molecular signatures between sexes using a lipidomics approach to characterize lipid species in liver, perigonadal adipose tissue, and inguinal adipose tissue and correlate them to the physiopathological responses observed. Males had less total fat but lower subcutaneous on visceral fat ratio together with higher liver weight and higher liver and serum triglyceride (TG) levels. Males were insulin resistant compared to females. Fatty acid (FA) and TG profiles differed between sexes in both fat pads, with longer chain FAs and TGs in males compared to that in females. Remarkably, hepatic phospholipid composition was sex dependent with more abundant lipotoxic FAs in males than in females. This may contribute to the sexual dimorphism in response to obesity towards more metaflammation in males. Our work presents an exhaustive novel description of a sex-specific lipid signature in the pathophysiology of metabolic disorders associated with obesity in ob/ob mice. These data could settle the basis for future pharmacological treatment in obesity.


Assuntos
Tecido Adiposo/metabolismo , Fígado/metabolismo , Obesidade/metabolismo , Caracteres Sexuais , Animais , Feminino , Metabolismo dos Lipídeos , Lipidômica , Masculino , Camundongos Endogâmicos C57BL , Camundongos Obesos
9.
Mol Cell Endocrinol ; 479: 147-158, 2019 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-30342056

RESUMO

OBJECTIVE: Estrogens play a key role in the distribution of adipose tissue and have their action by binding to both estrogen receptors (ER), α and ß. Although ERß has a role in the energy metabolism, limited data of the physiological mechanism and metabolic response involved in the pharmacological activation of ERß is available. METHODS: For clinical relevance, non-ovariectomized female mice were subjected to high fat diet together with pharmacological (DIP - 4-(2-(3,5-dimethylisoxazol-4-yl)-1H-indol-3-yl)phenol) interventions to ERß selective activation. The physiological mechanism was assessed in vivo by magnetic resonance imaging and spectroscopy, and oral glucose and intraperitoneal insulin tolerance test before and after DIP treatment. Liver and adipose tissue metabolic response was measured in HFD + vehicle and HFD + DIP by stable isotope, RNA sequencing and protein content. RESULTS: HFD-fed females treated with DIP had a tissue-specific response towards ERß selective activation. The metabolic profile showed an improved fasting glucose level, insulin sensitivity and reduced liver steatosis. CONCLUSIONS: Our data demonstrate that selective activation of ERß exerts a tissue-specific activity which promotes a beneficial effect on whole body metabolic response to obesity.


Assuntos
Tecido Adiposo/metabolismo , Metabolismo Energético , Receptor beta de Estrogênio/metabolismo , Mitocôndrias/metabolismo , Obesidade/metabolismo , Biogênese de Organelas , Animais , Feminino , Perfilação da Expressão Gênica , Isoxazóis , Ligantes , Lipogênese , Lipólise , Fígado/metabolismo , Camundongos Endogâmicos C57BL
10.
Horm Mol Biol Clin Investig ; 29(1): 13-26, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27831918

RESUMO

Both functional ovaries and estrogen replacement therapy (ERT) reduce the risk of type 2 diabetes (T2D). Understanding the mechanisms underlying the antidiabetic effects of 17ß-estradiol (E2) may permit the development of a molecular targeting strategy for the treatment of metabolic disease. This study examines how the promotion of insulin sensitivity and weight loss by E2 treatment in high-fat-diet (HFD)-fed mice involve several anti-adipogenic processes in the visceral adipose tissue. Magnetic resonance imaging (MRI) revealed specific reductions in visceral adipose tissue volume in HFD+E2 mice, compared with HFD mice. This loss of adiposity was associated with diminished visceral adipocyte size and reductions in expression of lipogenic genes, adipokines and of the nuclear receptor nr2c2/tr4. Meanwhile, expression levels of adipose triglyceride lipase/pnpla2 and leptin receptor were increased. As mRNA levels of stat3, a transcription factor involved in brown adipose tissue differentiation, were also increased in visceral adipose, the expression of other brown adipose-specific markers was assessed. Both expression and immunohistochemical staining of ucp-1 were increased, and mRNA levels of dio-2, and of adrß3, a regulator of ucp-1 expression during the thermogenic response, were increased. Furthermore, expression of cpt-1b, a brown adipose-specific gene involved in fatty acid utilization, was also increased. Methylation studies demonstrated that the methylation status of both dio-2 and adrß3 was significantly reduced. These results show that improved glycemic control and weight loss due to E2 involve anti-adipogenic mechanisms which include suppressed lipogenesis and augmented fatty acid utilization, and in addition, the activation of brown adipose tissue-specific gene expression in association with E2-dependent epigenetic modifications in these genes.


Assuntos
Adipogenia/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Biomarcadores/metabolismo , Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Gordura Intra-Abdominal/metabolismo , Obesidade/metabolismo , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/patologia , Animais , Western Blotting , Metilação de DNA , Dieta Hiperlipídica/efeitos adversos , Feminino , Perfilação da Expressão Gênica , Resistência à Insulina , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/patologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Biomol Concepts ; 6(3): 177-90, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25945723

RESUMO

The liver X receptors (LXR) are crucial regulators of metabolism. After ligand binding, they regulate gene transcription and thereby mediate changes in metabolic pathways. Modulation of LXR and their downstream targets has appeared to be a promising treatment for metabolic diseases especially atherosclerosis and cholesterol metabolism. However, the complexity of LXR action in various metabolic tissues and the liver side effect of LXR activation have slowed down the interest for LXR drugs. In this review, we summarized the role of LXR in the main metabolically active tissues with a special focus on obesity and associated diseases in mammals. We will also discuss the dual interplay between the two LXR isoforms suggesting that they may collaborate to establish a fine and efficient system for the maintenance of metabolism homeostasis.


Assuntos
Metabolismo dos Lipídeos , Fígado/metabolismo , Receptores Nucleares Órfãos/metabolismo , Tecido Adiposo Marrom/metabolismo , Animais , Humanos , Receptores X do Fígado , Músculo Esquelético/metabolismo , Fatores Sexuais
12.
Am J Physiol Endocrinol Metab ; 306(5): E494-502, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24368671

RESUMO

The liver X receptors (LXR)α and LXRß are transcription factors belonging to the nuclear receptor family, which play a central role in metabolic homeostasis, being master regulators of key target genes in the glucose and lipid pathways. Wild-type (WT), LXRα(-/-), and LXRß(-/-) mice were fed a chow diet with (treated) or without (control) the synthetic dual LXR agonist GW3965 for 5 wk. GW3965 raised intrahepatic triglyceride (TG) level but, surprisingly, reduced serum TG level through the activation of serum lipase activity. The serum TG reduction was associated with a repression of both catecholamine-stimulated lipolysis and relative glucose incorporation into lipid in isolated adipocytes through activation of LXRß. We also demonstrated that LXRα is required for basal (nonstimulated) adipocyte metabolism, whereas LXRß acts as a repressor of lipolysis. On the contrary, in skeletal muscle (SM), the lipogenic and cholesterol transporter LXR target genes were markedly induced in WT and LXRα(-/-) mice and to a lesser extent in LXRß(-/-) mice following treatment with GW3965. Moreover, TG content was reduced in SM of LXRß(-/-) mice, associated with increased expression of the main TG-lipase genes Hsl and Atgl. Energy expenditure was increased, and a switch from glucose to lipid oxidation was observed. In conclusion, we provide evidence that LXR might be an essential regulator of the lipid balance between tissues to ensure appropriate control of the flux of fuel. Importantly, we show that, after chronic treatment with GW3965, SM becomes the target tissue for LXR activation, as opposed to liver, in acute treatment.


Assuntos
Adipócitos/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Receptores Nucleares Órfãos/agonistas , Adipócitos/metabolismo , Animais , Benzoatos/farmacologia , Benzilaminas/farmacologia , Colesterol/metabolismo , Feminino , Homeostase/fisiologia , Metabolismo dos Lipídeos/fisiologia , Lipólise/efeitos dos fármacos , Lipólise/fisiologia , Receptores X do Fígado , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , Triglicerídeos/sangue
13.
J Lipid Res ; 54(5): 1300-11, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23446231

RESUMO

To investigate the role of liver X receptor (LXR) in adipose tissue metabolism during obesity, ob/ob mice were treated for 5 weeks with the synthetic LXR agonist GW3965. MRI analysis revealed that pharmacological activation of LXR modified fat distribution by decreasing visceral (VS) fat and inversely increasing subcutaneous (SC) fat storage without affecting whole body fat content. This was concordant with opposite regulation by GW3965 of the lipolytic markers hormone-sensitive lipase (HSL) and adipose triglyceride lipase (ATGL) in the two fat depots; moreover, the expression of genes involved in lipogenesis was significantly induced in SC fat. Lipidomic analysis suggested that changes in lipid composition in response to GW3965 also varied between VS and SC fat. In both depots, the observed alteration in lipid composition indicated an overall change toward less lipotoxic lipids. Flow cytometry analysis showed decreased immune cell infiltration in adipose tissue of ob/ob mice in response to GW3965 treatment, which in VS fat mainly affected the macrophage population and in SC fat the lymphocyte population. In line with this, the expression and secretion of proinflammatory markers was decreased in both fat deposits with GW3965 treatment.


Assuntos
Tecido Adiposo/metabolismo , Benzoatos/administração & dosagem , Benzilaminas/administração & dosagem , Obesidade/metabolismo , Receptores Nucleares Órfãos/metabolismo , Adipogenia , Animais , Distribuição da Gordura Corporal , Feminino , Inflamação/metabolismo , Inflamação/patologia , Lipólise , Receptores X do Fígado , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Obesidade/genética , Obesidade/patologia , Receptores Nucleares Órfãos/agonistas , Receptores Nucleares Órfãos/genética
14.
Mol Endocrinol ; 26(12): 1980-90, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23073827

RESUMO

The liver plays a pivotal role in the physiological adaptation to fasting and a better understanding of the metabolic adaptive responses may give hints on new therapeutic strategies to control the metabolic diseases. The liver X receptors (LXRs) are well-established regulators of lipid and glucose metabolism. More recently fibroblast growth factor 21 (FGF21) has emerged as an important regulator of energy homeostasis. We hypothesized that the LXR transcription factors could influence Fgf21 expression, which is induced in response to fasting. Wild-type, LXRα(-/-), and LXRß(-/-) mice were treated for 3 d with vehicle or the LXR agonist GW3965 and fasted for 12 h prior to the killing of the animals. Interestingly, serum FGF21 levels were induced after fasting, but this increase was blunted when the mice were treated with GW3965 independently of genotypes. Compared with wild-type mice, GW3965-treated LXRα(-/-) and LXRß(-/-) mice showed improved insulin sensitivity and enhanced ketogenic response at fasting. Of note is that during fasting, GW3965 treatment tended to reduce liver triglycerides as opposed to the effect of the agonist in the fed state. The LXR-dependent repression of Fgf21 seems to be mainly mediated by the recruitment of LXRß onto the Fgf21 promoter upon GW3965 treatment. This repression by LXRß occurs through the recruitment and stabilization of the repressor complex composed of retinoid-related orphan receptor-α/Rev-Erbα/histone deacetylase 3 onto the Fgf21 promoter. Our data clearly demonstrate that there is a cross talk between the LXR and FGF21 signaling pathways in the adaptive response to fasting.


Assuntos
Jejum/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Histona Desacetilases/metabolismo , Receptores Nucleares Órfãos/metabolismo , Animais , Benzoatos/farmacologia , Benzilaminas/farmacologia , Fatores de Crescimento de Fibroblastos/sangue , Glucose/metabolismo , Homeostase , Resistência à Insulina , Metabolismo dos Lipídeos , Fígado , Receptores X do Fígado , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Receptores Nucleares Órfãos/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Transdução de Sinais , Triglicerídeos/metabolismo
15.
Am J Physiol Endocrinol Metab ; 301(1): E210-22, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21521718

RESUMO

The liver X receptors (LXRs) play a key role in cholesterol and bile acid metabolism but are also important regulators of glucose metabolism. Recently, LXRs have been proposed as a glucose sensor affecting LXR-dependent gene expression. We challenged wild-type (WT) and LXRαß(-/-) mice with a normal diet (ND) or a high-carbohydrate diet (HCD). Magnetic resonance imaging showed different fat distribution between WT and LXRαß(-/-) mice. Surprisingly, gonadal (GL) adipocyte volume decreased on HCD compared with ND in WT mice, whereas it slightly increased in LXRαß(-/-) mice. Interestingly, insulin-stimulated lipogenesis of isolated GL fat cells was reduced on HCD compared with ND in LXRαß(-/-) mice, whereas no changes were observed in WT mice. Net de novo lipogenesis (DNL) calculated from Vo(2) and Vco(2) was significantly higher in LXRαß(-/-) than in WT mice on HCD. Histology of HCD-fed livers showed hepatic steatosis in WT mice but not in LXRαß(-/-) mice. Glucose tolerance was not different between groups, but insulin sensitivity was decreased by the HCD in WT but not in LXRαß(-/-) mice. Finally, gene expression analysis of adipose tissue showed induced expression of genes involved in DNL in LXRαß(-/-) mice compared with WT animals as opposed to the liver, where expression of DNL genes was repressed in LXRαß(-/-) mice. We thus conclude that absence of LXRs stimulates DNL in adipose tissue, but suppresses DNL in the liver, demonstrating opposite roles of LXR in DNL regulation in these two tissues. These results show tissue-specific regulation of LXR activity, a crucial finding for drug development.


Assuntos
Lipogênese/genética , Receptores Nucleares Órfãos/fisiologia , Adipócitos/citologia , Adipócitos/metabolismo , Adipócitos/fisiologia , Tecido Adiposo/metabolismo , Adiposidade/genética , Animais , Distribuição da Gordura Corporal , Células Cultivadas , Feminino , Lipólise/genética , Lipólise/fisiologia , Receptores X do Fígado , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade de Órgãos/genética , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo
16.
Proc Natl Acad Sci U S A ; 108(1): 403-8, 2011 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-21173252

RESUMO

Brown adipocytes are multilocular lipid storage cells that play a crucial role in nonshivering thermogenesis. Uncoupling protein 1 (UCP1) is a unique feature of brown fat cells that allows heat generation on sympathetic nervous system stimulation. As conventional transcriptional factors that are activated in various signaling pathways, liver-X receptors (LXRs) play important roles in many physiological processes. The role of LXRs in the regulation of energy homeostasis remains unclear, however. Female WT, LXRαß(-/-), LXRα(-/-), and LXRß(-/-) mice were fed with either a normal diet (ND) or a high-carbohydrate diet (HCD) supplemented with or without GW3965-LXR agonist. LXRαß(-/-) mice exhibited higher energy expenditure (EE) as well as higher UCP1 expression in brown adipose tissue (BAT) compared with WT mice on the HCD. In addition, long-term treatment of WT mice with GW3965 showed lower EE at thermoneutrality (30 °C) and lower Ucp1 expression level in BAT. Furthermore, H&E staining of the BAT of LXRαß(-/-) mice exhibited decreased lipid droplet size compared with WT mice on the HCD associated with a more intense UCP1-positive reaction. Quantification of triglyceride (TG) content in BAT showed lower TG accumulation in LXRß(-/-) mice compared with WT mice. Surprisingly, GW3965 treatment increased TG content (twofold) in the BAT of WT and LXRα(-/-) mice but not in LXRß(-/-) mice. Furthermore, glucose transporter (GLUT4) in the BAT of LXRα(-/-) and LXRß(-/-) mice was sixfold and fourfold increased, respectively, compared with WT mice on the ND. These findings suggest that LXRα as well as LXRß could play a crucial role in the regulation of energy homeostasis in female mice and may be a potential target for the treatment of obesity and energy regulation.


Assuntos
Tecido Adiposo Marrom/metabolismo , Metabolismo Energético/fisiologia , Canais Iônicos/metabolismo , Proteínas Mitocondriais/metabolismo , Receptores Nucleares Órfãos/metabolismo , Termogênese/fisiologia , Tecido Adiposo Marrom/fisiologia , Análise de Variância , Animais , Benzoatos/farmacologia , Benzilaminas/farmacologia , Glicemia , Western Blotting , Calorimetria , Feminino , Transportador de Glucose Tipo 4/metabolismo , Imuno-Histoquímica , Receptores X do Fígado , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Nucleares Órfãos/agonistas , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/fisiologia , Reação em Cadeia da Polimerase , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Triglicerídeos/metabolismo , Proteína Desacopladora 1
17.
PLoS One ; 5(9)2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20927337

RESUMO

BACKGROUND: Intervention strategies for obesity are global issues that require immediate attention. One approach is to exploit the growing consensus that beneficial gut microbiota could be of use in intervention regimes. Our objective was to determine the mechanism by which the probiotic bacteria Lactobacillus paracasei ssp paracasei F19 (F19) could alter fat storage. Angiopoietin-like 4 (ANGPTL4) is a circulating lipoprotein lipase (LPL) inhibitor that controls triglyceride deposition into adipocytes and has been reported to be regulated by gut microbes. METHODOLOGY/PRINCIPAL FINDINGS: A diet intervention study of mice fed high-fat chow supplemented with F19 was carried out to study potential mechanistic effects on fat storage. Mice given F19 displayed significantly less body fat, as assessed by magnetic resonance imaging, and a changed lipoprotein profile. Given that previous studies on fat storage have identified ANGPTL4 as an effector, we also investigated circulating levels of ANGPTL4, which proved to be higher in the F19-treated group. This increase, together with total body fat and triglyceride levels told a story of inhibited LPL action through ANGPTL4 leading to decreased fat storage. Co-culture experiments of colonic cell lines and F19 were set up in order to monitor any ensuing alterations in ANGPTL4 expression by qPCR. We observed that potentially secreted factors from F19 can induce ANGPTL4 gene expression, acting in part through the peroxisome proliferator activated receptors alpha and gamma. To prove validity of in vitro findings, germ-free mice were monocolonized with F19. Here we again found changes in serum triglycerides as well as ANGPTL4 in response to F19. CONCLUSIONS/SIGNIFICANCE: Our results provide an interesting mechanism whereby modifying ANGPTL4, a central player in fat storage regulation, through manipulating gut flora could be an important gateway upon which intervention trials of weight management can be based.


Assuntos
Angiopoietinas/metabolismo , Gorduras/metabolismo , Lactobacillus/fisiologia , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/genética , Animais , Linhagem Celular Tumoral , Expressão Gênica , Humanos , Lipase Lipoproteica/antagonistas & inibidores , Lipase Lipoproteica/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/tratamento farmacológico , Obesidade/enzimologia , Probióticos/uso terapêutico
18.
Proc Natl Acad Sci U S A ; 107(33): 14763-8, 2010 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-20679224

RESUMO

Gallbladder cancer is a highly aggressive disease with poor prognosis that is two to six times more frequent in women than men. The development of gallbladder cancer occurs over a long time (more than 15 y) and evolves from chronic inflammation to dysplasia/metaplasia, carcinoma in situ, and invasive carcinoma. In the present study we found that, in female mice in which the oxysterol receptor liver X receptor-beta (LXRbeta) has been inactivated, preneoplastic lesions of the gallbladder developed and evolved to cancer in old animals. LXRbeta is a nuclear receptor involved in the control of lipid homeostasis, glucose metabolism, inflammation, proliferation, and CNS development. LXRbeta(-/-) female gallbladders were severely inflamed, with regions of dysplasia and high cell density, hyperchromasia, metaplasia, and adenomas. No abnormalities were evident in male mice, nor in LXRalpha(-/-) or LXRalpha(-/-)beta(-/-) animals of either sex. Interestingly, the elimination of estrogens with ovariectomy prevented development of preneoplastic lesions in LXRbeta(-/-) mice. The etiopathological mechanism seems to involve TGF-beta signaling, as the precancerous lesions were characterized by strong nuclear reactivity of phospho-SMAD-2 and SMAD-4 and loss of E-cadherin expression. Upon ovariectomy, E-cadherin was reexpressed on the cell membranes and immunoreactivity of pSMAD-2 in the nuclei was reduced. These findings suggest that LXRbeta in a complex interplay with estrogens and TGF-beta could play a crucial role in the malignant transformation of the gallbladder epithelium.


Assuntos
Estrogênios/metabolismo , Neoplasias da Vesícula Biliar/metabolismo , Vesícula Biliar/metabolismo , Receptores Nucleares Órfãos/metabolismo , Fatores Etários , Animais , Apoptose , Caderinas/metabolismo , Proliferação de Células , Feminino , Vesícula Biliar/patologia , Neoplasias da Vesícula Biliar/genética , Humanos , Imuno-Histoquímica , Receptores X do Fígado , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Nucleares Órfãos/genética , Ovariectomia , Fosforilação , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/metabolismo , Proteína Smad2/metabolismo , Proteína Smad4/metabolismo , Fator de Crescimento Transformador beta/sangue , Fator de Crescimento Transformador beta/metabolismo
19.
Am J Physiol Endocrinol Metab ; 298(2): E167-78, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19690071

RESUMO

The two liver X receptors (LXRs), LXRalpha and LXRbeta, are transcriptional regulators of cholesterol, lipid, and glucose metabolism and are both activated by oxysterols. Impaired metabolism is linked with obesity, insulin resistance, and type 2-diabetes (T2D). In the present study, we aimed to delineate the specific roles of LXRalpha and -beta in metabolic processes. C57Bl/6 female mice were fed a normal or a high-fat diet (HFD) and metabolic responses in wild-type, LXRalpha(-/-), LXRbeta(-/-), and LXRalphabeta(-/-) mice were analyzed. Whole body fat and intramyocellular lipid contents were measured by nuclear magnetic resonance. Energy expenditure was measured in individual metabolic cages. Glucose, insulin, and pyruvate tolerance tests were performed and gene expression profiles analyzed by qPCR. We found that both LXRbeta(-/-) and LXRalphabeta(-/-) mice are resistant to HFD-induced obesity independently of the presence of high cholesterol. Using tolerance tests, we found that, on an HFD, LXRbeta(-/-) mice enhanced their endogenous glucose production and became highly insulin resistant, whereas LXRalpha(-/-) and LXRalphabeta(-/-) mice remained glucose tolerant and insulin sensitive. Gene expression profiling confirmed that LXRbeta is the regulator of lipogenic genes in visceral white adipose tissue (WAT) and muscle tissue and, surprisingly, that Ucp1 and Dio2 are not responsible for the protection against diet-induced obesity observed in LXRbeta(-/-) and LXRalphabeta(-/-) mice. LXRalpha is required for the control of cholesterol metabolism in the liver, while LXRbeta appears to be a major regulator of glucose homeostasis and energy utilization and of fat storage in muscle and WAT. We conclude that selective LXRbeta agonists would be novel pharmaceuticals in the treatment of T2D.


Assuntos
Glicemia/metabolismo , Colesterol/metabolismo , Gorduras na Dieta/metabolismo , Metabolismo Energético/fisiologia , Receptores Nucleares Órfãos/metabolismo , Tecido Adiposo Branco/metabolismo , Ração Animal , Animais , Metabolismo Energético/genética , Feminino , Perfilação da Expressão Gênica , Teste de Tolerância a Glucose , Resistência à Insulina/fisiologia , Fígado/metabolismo , Receptores X do Fígado , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Obesidade/genética , Obesidade/metabolismo , Receptores Nucleares Órfãos/classificação , Isoformas de Proteínas/metabolismo
20.
Proc Natl Acad Sci U S A ; 105(20): 7252-6, 2008 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-18480259

RESUMO

Growth/differentiation factor 3 (GDF3) is highly expressed in adipose tissue, and previous overexpression experiments in mice have suggested that it may act as an adipogenic factor under conditions of high lipid load. GDF3 has been shown to signal via the activin receptor ALK4 during embryogenesis, but functional receptors in adipose tissue are unknown. In this study, we show that Gdf3(-/-) mutant mice accumulate less adipose tissue than WT animals and show partial resistance to high-fat diet-induced obesity despite similar food intake. We also demonstrate that GDF3 can signal via the ALK4-homolog ALK7 and the coreceptor Cripto, both of which are expressed in adipose tissue. In agreement with a role for ALK7 in GDF3 signaling in vivo, mutant mice lacking ALK7 also showed reduced fat accumulation and partial resistance to diet-induced obesity. We propose that GDF3 regulates adipose-tissue homeostasis and energy balance under nutrient overload in part by signaling through the ALK7 receptor.


Assuntos
Receptores de Ativinas Tipo I/fisiologia , Tecido Adiposo/metabolismo , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Obesidade/genética , Ração Animal , Animais , Dieta , Fator 3 de Diferenciação de Crescimento , Humanos , Insulina/metabolismo , Camundongos , Camundongos Transgênicos , Obesidade/metabolismo , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...