Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(3): e1012060, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38442126

RESUMO

The recent discovery of Hepatitis D (HDV)-like viruses across a wide range of taxa led to the establishment of the Kolmioviridae family. Recent studies suggest that kolmiovirids can be satellites of viruses other than Hepatitis B virus (HBV), challenging the strict HBV/HDV-association dogma. Studying whether kolmiovirids are able to replicate in any animal cell they enter is essential to assess their zoonotic potential. Here, we compared replication of three kolmiovirids: HDV, rodent (RDeV) and snake (SDeV) deltavirus in vitro and in vivo. We show that SDeV has the narrowest and RDeV the broadest host cell range. High resolution imaging of cells persistently replicating these viruses revealed nuclear viral hubs with a peculiar RNA-protein organization. Finally, in vivo hydrodynamic delivery of viral replicons showed that both HDV and RDeV, but not SDeV, efficiently replicate in mouse liver, forming massive nuclear viral hubs. Our comparative analysis lays the foundation for the discovery of specific host factors controlling Kolmioviridae host-shifting.


Assuntos
Hepatite D , Vírus Delta da Hepatite , Camundongos , Animais , Humanos , Roedores , Vírus da Hepatite B/genética , Serpentes , Replicação Viral , RNA Viral/genética
2.
EMBO Mol Med ; 16(2): 238-250, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38228803

RESUMO

FGF19 hormone has pleiotropic metabolic functions, including the modulation of insulin sensitivity, glucose/lipid metabolism and energy homeostasis. On top of its physiological metabolic role, FGF19 has been identified as a potentially targetable oncogenic driver, notably in hepatocellular carcinoma (HCC). Nevertheless, FGF19 remained an attractive candidate for treatment of metabolic disease, prompting the development of analogs uncoupling its metabolic and tumor-promoting activities. Using pre-clinical mice models of somatic mutation driven HCC, we assessed the oncogenicity of FGF19 in combination with frequent HCC tumorigenic alterations: p53 inactivation, CTNNB1 mutation, CCND1 or MYC overexpression. Our data revealed a strong oncogenic cooperation between FGF19 and MYC. Most importantly, we show that this oncogenic synergy is conserved with a FGF19-analog Aldafermin (NGM282), designed to solely mimic the hormone's metabolic functions. In particular, even a short systemic treatment with recombinant proteins triggered rapid appearance of proliferative foci of MYC-expressing hepatocytes. The fact that FGF19 analog Aldafermin is not fully devoid of the hormone's oncogenic properties raises concerns in the context of its potential use for patients with damaged, mutation-prone liver.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Camundongos , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Hormônios
4.
Elife ; 122023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36656749

RESUMO

Quantitative differences in signal transduction are to date an understudied feature of tumour heterogeneity. The MAPK Erk pathway, which is activated in a large proportion of human tumours, is a prototypic example of distinct cell fates being driven by signal intensity. We have used primary hepatocyte precursors transformed with different dosages of an oncogenic form of Ras to model subclonal variations in MAPK signalling. Orthotopic allografts of Ras-transformed cells in immunocompromised mice gave rise to fast-growing aggressive tumours, both at the primary location and in the peritoneal cavity. Fluorescent labelling of cells expressing different oncogene levels, and consequently varying levels of MAPK Erk activation, highlighted the selection processes operating at the two sites of tumour growth. Indeed, significantly higher Ras expression was observed in primary as compared to secondary, metastatic sites, despite the apparent evolutionary trade-off of increased apoptotic death in the liver that correlated with high Ras dosage. Analysis of the immune tumour microenvironment at the two locations suggests that fast peritoneal tumour growth in the immunocompromised setting is abrogated in immunocompetent animals due to efficient antigen presentation by peritoneal dendritic cells. Furthermore, our data indicate that, in contrast to the metastatic-like outgrowth, strong MAPK signalling is required in the primary liver tumours to resist elimination by NK (natural killer) cells. Overall, this study describes a quantitative aspect of tumour heterogeneity and points to a potential vulnerability of a subtype of hepatocellular carcinoma as a function of MAPK Erk signalling intensity.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Humanos , Camundongos , Carcinoma Hepatocelular/genética , Células Matadoras Naturais , Neoplasias Hepáticas/genética , Sistema de Sinalização das MAP Quinases , Transdução de Sinais , Microambiente Tumoral , Proteínas ras/metabolismo
6.
Mol Metab ; 60: 101483, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35367668

RESUMO

Fibroblast growth factor 19 (FGF19) is a hormone with pleiotropic metabolic functions, leading to ongoing development of analogues for treatment of metabolic disorders. On the other hand, FGF19 is overexpressed in a sub-group of hepatocellular carcinoma (HCC) patients and has oncogenic properties. It is therefore crucial to precisely define FGF19 effects, notably in the context of chronic exposure to elevated concentrations of the hormone. Here, we used hydrodynamic gene transfer to generate a transgenic mouse model with long-term FGF19 hepatic overexpression. We describe a novel effect of FGF19, namely the stimulation of water intake. This phenotype, lasting at least over a 6-month period, depends on signaling in the central nervous system and is independent of FGF21, although it mimics some of its features. We further show that HCC patients with high levels of circulating FGF19 have a reduced natremia, indicating dipsogenic features. The present study provides evidence of a new activity of FGF19, which could be clinically relevant in the context of FGF19 overexpressing cancers and in the course of treatment of metabolic disorders by FGF19 analogues.


Assuntos
Carcinoma Hepatocelular , Fatores de Crescimento de Fibroblastos/metabolismo , Neoplasias Hepáticas , Animais , Carcinoma Hepatocelular/metabolismo , Ingestão de Líquidos , Fatores de Crescimento de Fibroblastos/genética , Hormônios , Humanos , Neoplasias Hepáticas/metabolismo , Camundongos , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/metabolismo
7.
Diabetes ; 69(4): 647-660, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32005707

RESUMO

Aging-dependent changes in tissue function are associated with the development of metabolic diseases. However, the molecular connections linking aging, obesity, and diabetes remain unclear. Lamin A, lamin C, and progerin, products of the Lmna gene, have antagonistic functions on energy metabolism and life span. Lamin C, albeit promoting obesity, increases life span, suggesting that this isoform is crucial for maintaining healthy conditions under metabolic stresses. Because ß-cell loss during obesity or aging leads to diabetes, we investigated the contribution of lamin C to ß-cell function in physiopathological conditions. We demonstrate that aged lamin C only-expressing mice (Lmna LCS/LCS ) become obese but remain glucose tolerant due to adaptive mechanisms including increased ß-cell mass and insulin secretion. Triggering diabetes in young mice revealed that Lmna LCS/LCS animals normalize their fasting glycemia by both increasing insulin secretion and regenerating ß-cells. Genome-wide analyses combined to functional analyses revealed an increase of mitochondrial biogenesis and global translational rate in Lmna LCS/LCS islets, two major processes involved in insulin secretion. Altogether, our results demonstrate for the first time that the sole expression of lamin C protects from glucose intolerance through a ß-cell-adaptive transcriptional program during metabolic stresses, highlighting Lmna gene processing as a new therapeutic target for diabetes treatment.


Assuntos
Envelhecimento/metabolismo , Diabetes Mellitus/metabolismo , Intolerância à Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Lamina Tipo A/metabolismo , Obesidade/metabolismo , Envelhecimento/genética , Animais , Glicemia/metabolismo , Diabetes Mellitus/genética , Metabolismo Energético/fisiologia , Glucagon/metabolismo , Intolerância à Glucose/genética , Insulina/metabolismo , Lamina Tipo A/genética , Camundongos , Camundongos Transgênicos , Obesidade/genética , Pâncreas/metabolismo
8.
Mol Cell ; 68(2): 336-349.e6, 2017 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-29053957

RESUMO

The roles of CDK4 in the cell cycle have been extensively studied, but less is known about the mechanisms underlying the metabolic regulation by CDK4. Here, we report that CDK4 promotes anaerobic glycolysis and represses fatty acid oxidation in mouse embryonic fibroblasts (MEFs) by targeting the AMP-activated protein kinase (AMPK). We also show that fatty acid oxidation (FAO) is specifically induced by AMPK complexes containing the α2 subunit. Moreover, we report that CDK4 represses FAO through direct phosphorylation and inhibition of AMPKα2. The expression of non-phosphorylatable AMPKα2 mutants, or the use of a CDK4 inhibitor, increased FAO rates in MEFs and myotubes. In addition, Cdk4-/- mice have increased oxidative metabolism and exercise capacity. Inhibition of CDK4 mimicked these alterations in normal mice, but not when skeletal muscle was AMPK deficient. This novel mechanism explains how CDK4 promotes anabolism by blocking catabolic processes (FAO) that are activated by AMPK.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Quinase 4 Dependente de Ciclina/metabolismo , Ácidos Graxos/metabolismo , Músculo Esquelético/metabolismo , Condicionamento Físico Animal , Proteínas Quinases Ativadas por AMP/genética , Animais , Quinase 4 Dependente de Ciclina/genética , Embrião de Mamíferos/metabolismo , Ácidos Graxos/genética , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Mutação , Oxirredução
9.
Cell Rep ; 15(5): 1051-1061, 2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-27117420

RESUMO

The endoplasmic reticulum (ER) unfolded protein response (UPR(er)) pathway plays an important role in helping pancreatic ß cells to adapt their cellular responses to environmental cues and metabolic stress. Although altered UPR(er) gene expression appears in rodent and human type 2 diabetic (T2D) islets, the underlying molecular mechanisms remain unknown. We show here that germline and ß cell-specific disruption of the lysine acetyltransferase 2B (Kat2b) gene in mice leads to impaired insulin secretion and glucose intolerance. Genome-wide analysis of Kat2b-regulated genes and functional assays reveal a critical role for Kat2b in maintaining UPR(er) gene expression and subsequent ß cell function. Importantly, Kat2b expression is decreased in mouse and human diabetic ß cells and correlates with UPR(er) gene expression in normal human islets. In conclusion, Kat2b is a crucial transcriptional regulator for adaptive ß cell function during metabolic stress by controlling UPR(er) and represents a promising target for T2D prevention and treatment.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Intolerância à Glucose/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Fatores de Transcrição de p300-CBP/genética , Adaptação Fisiológica , Animais , Linhagem Celular , Retículo Endoplasmático/metabolismo , Humanos , Secreção de Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Estresse Fisiológico , Fatores de Transcrição/metabolismo , Resposta a Proteínas não Dobradas/genética , Fatores de Transcrição de p300-CBP/deficiência
10.
J Clin Invest ; 126(1): 335-48, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26657864

RESUMO

Insulin resistance is a fundamental pathogenic factor that characterizes various metabolic disorders, including obesity and type 2 diabetes. Adipose tissue contributes to the development of obesity-related insulin resistance through increased release of fatty acids, altered adipokine secretion, and/or macrophage infiltration and cytokine release. Here, we aimed to analyze the participation of the cyclin-dependent kinase 4 (CDK4) in adipose tissue biology. We determined that white adipose tissue (WAT) from CDK4-deficient mice exhibits impaired lipogenesis and increased lipolysis. Conversely, lipolysis was decreased and lipogenesis was increased in mice expressing a mutant hyperactive form of CDK4 (CDK4(R24C)). A global kinome analysis of CDK4-deficient mice following insulin stimulation revealed that insulin signaling is impaired in these animals. We determined that insulin activates the CCND3-CDK4 complex, which in turn phosphorylates insulin receptor substrate 2 (IRS2) at serine 388, thereby creating a positive feedback loop that maintains adipocyte insulin signaling. Furthermore, we found that CCND3 expression and IRS2 serine 388 phosphorylation are increased in human obese subjects. Together, our results demonstrate that CDK4 is a major regulator of insulin signaling in WAT.


Assuntos
Adipócitos/metabolismo , Quinase 4 Dependente de Ciclina/fisiologia , Insulina/farmacologia , Células 3T3-L1 , Tecido Adiposo Branco/metabolismo , Animais , Ciclina D3/fisiologia , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Fator de Transcrição E2F1/fisiologia , Feminino , Humanos , Proteínas Substratos do Receptor de Insulina/metabolismo , Resistência à Insulina , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Transdução de Sinais
11.
EMBO Rep ; 15(5): 529-39, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24639560

RESUMO

Alternative RNA processing of LMNA pre-mRNA produces three main protein isoforms, that is, lamin A, progerin, and lamin C. De novo mutations that favor the expression of progerin over lamin A lead to Hutchinson-Gilford progeria syndrome (HGPS), providing support for the involvement of LMNA processing in pathological aging. Lamin C expression is mutually exclusive with the splicing of lamin A and progerin isoforms and occurs by alternative polyadenylation. Here, we investigate the function of lamin C in aging and metabolism using mice that express only this isoform. Intriguingly, these mice live longer, have decreased energy metabolism, increased weight gain, and reduced respiration. In contrast, progerin-expressing mice show increased energy metabolism and are lipodystrophic. Increased mitochondrial biogenesis is found in adipose tissue from HGPS-like mice, whereas lamin C-only mice have fewer mitochondria. Consistently, transcriptome analyses of adipose tissues from HGPS and lamin C-only mice reveal inversely correlated expression of key regulators of energy expenditure, including Pgc1a and Sfrp5. Our results demonstrate that LMNA encodes functionally distinct isoforms that have opposing effects on energy metabolism and lifespan in mammals.


Assuntos
Tecido Adiposo/fisiologia , Metabolismo Energético/genética , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Adipócitos/citologia , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Envelhecimento , Processamento Alternativo , Animais , Células Cultivadas , Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lamina Tipo A/biossíntese , Longevidade/genética , Camundongos , Camundongos Transgênicos , Mitocôndrias , Proteínas Nucleares/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Progéria/genética , Isoformas de Proteínas , Precursores de Proteínas/genética , Transdução de Sinais , Fatores de Transcrição/metabolismo
12.
Nat Cell Biol ; 13(9): 1146-52, 2011 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-21841792

RESUMO

Cells respond to stress by coordinating proliferative and metabolic pathways. Starvation restricts cell proliferative (glycolytic) and activates energy productive (oxidative) pathways. Conversely, cell growth and proliferation require increased glycolytic and decreased oxidative metabolism levels. E2F transcription factors regulate both proliferative and metabolic genes. E2Fs have been implicated in the G1/S cell-cycle transition, DNA repair, apoptosis, development and differentiation. In pancreatic ß-cells, E2F1 gene regulation facilitated glucose-stimulated insulin secretion. Moreover, mice lacking E2F1 (E2f1(-/-)) were resistant to diet-induced obesity. Here, we show that E2F1 coordinates cellular responses by acting as a regulatory switch between cell proliferation and metabolism. In basal conditions, E2F1 repressed key genes that regulate energy homeostasis and mitochondrial functions in muscle and brown adipose tissue. Consequently, E2f1(-/-) mice had a marked oxidative phenotype. An association between E2F1 and pRB was required for repression of genes implicated in oxidative metabolism. This repression was alleviated in a constitutively active CDK4 (CDK4(R24C)) mouse model or when adaptation to energy demand was required. Thus, E2F1 represents a metabolic switch from oxidative to glycolytic metabolism that responds to stressful conditions.


Assuntos
Tecido Adiposo Marrom/metabolismo , Fator de Transcrição E2F1/metabolismo , Metabolismo Energético , Músculo Esquelético/metabolismo , Tecido Adiposo Marrom/citologia , Animais , Proliferação de Células , Células Cultivadas , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Metilação de DNA , Fator de Transcrição E2F1/genética , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Immunoblotting , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/ultraestrutura , Mioblastos/citologia , Mioblastos/metabolismo , Consumo de Oxigênio , Interferência de RNA , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Environ Health Perspect ; 118(4): 465-71, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20064776

RESUMO

BACKGROUND: The incidence of the insulin resistance syndrome has increased at an alarming rate worldwide, creating a serious challenge to public health care in the 21st century. Recently, epidemiological studies have associated the prevalence of type 2 diabetes with elevated body burdens of persistent organic pollutants (POPs). However, experimental evidence demonstrating a causal link between POPs and the development of insulin resistance is lacking. OBJECTIVE: We investigated whether exposure to POPs contributes to insulin resistance and metabolic disorders. METHODS: Sprague-Dawley rats were exposed for 28 days to lipophilic POPs through the consumption of a high-fat diet containing either refined or crude fish oil obtained from farmed Atlantic salmon. In addition, differentiated adipocytes were exposed to several POP mixtures that mimicked the relative abundance of organic pollutants present in crude salmon oil. We measured body weight, whole-body insulin sensitivity, POP accumulation, lipid and glucose homeostasis, and gene expression and we performed microarray analysis. RESULTS: Adult male rats exposed to crude, but not refined, salmon oil developed insulin resistance, abdominal obesity, and hepatosteatosis. The contribution of POPs to insulin resistance was confirmed in cultured adipocytes where POPs, especially organochlorine pesticides, led to robust inhibition of insulin action. Moreover, POPs induced down-regulation of insulin-induced gene-1 (Insig-1) and Lpin1, two master regulators of lipid homeostasis. CONCLUSION: Our findings provide evidence that exposure to POPs commonly present in food chains leads to insulin resistance and associated metabolic disorders.


Assuntos
Poluentes Ambientais/toxicidade , Resistência à Insulina , Células 3T3-L1 , Animais , Metabolismo dos Carboidratos/efeitos dos fármacos , Glucose/metabolismo , Técnica Clamp de Glucose , Hidrocarbonetos Clorados/toxicidade , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Síndrome Metabólica/induzido quimicamente , Síndrome Metabólica/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Praguicidas/toxicidade , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley
14.
PLoS One ; 4(10): e7422, 2009 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-19823686

RESUMO

The cell surface low-density lipoprotein receptor-related protein 1, LRP1, plays a major role in lipid metabolism. The question that remains open concerns the function of LRP1 in adipogenesis. Here, we show that LRP1 is highly expressed in murine preadipocytes as well as in primary culture of human adipocytes. Moreover, LRP1 remains abundantly synthesised during mouse and human adipocyte differentiation. We demonstrate that LRP1 silencing in 3T3F442A murine preadipocytes significantly inhibits the expression of PPARgamma, HSL and aP2 adipocyte differentiation markers after adipogenesis induction, and leads to lipid-depleted cells. We further show that the absence of lipids in LRP1-silenced preadipocytes is not caused by lipolysis induction. In addition, we provide the first evidences that LRP1 is significantly up-regulated in obese C57BI6/J mouse adipocytes and obese human adipose tissues. Interestingly, silencing of LRP1 in fully-differentiated adipocytes also reduces cellular lipid level and is associated with an increase of basal lipolysis. However, the ability of mature adipocytes to induce lipolysis is independent of LRP1 expression. Altogether, our findings highlight the dual role of LRP1 in the control of adipogenesis and lipid homeostasis, and suggest that LRP1 may be an important therapeutic target in obesity.


Assuntos
Adipogenia , Tecido Adiposo/metabolismo , Obesidade/metabolismo , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Adipócitos/metabolismo , Animais , Diferenciação Celular , Feminino , Inativação Gênica , Humanos , Lipídeos/química , Camundongos , Camundongos Endogâmicos C57BL , Regulação para Cima
15.
Int J Biochem Cell Biol ; 41(11): 2173-80, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19782949

RESUMO

The insulin/insulin-like growth factor 1 (IGF-1) signaling pathway is a major regulator of adipose tissue growth and differentiation. We recently demonstrated that human protein tyrosine phosphatase (PTP) L1, a large cytoplasmic phosphatase also known as PTP-BAS/PTPN13/PTP-1E, is a negative regulator of IGF-1R/IRS-1/Akt pathway in breast cancer cells. This triggered us to investigate the potential role of PTPL1 in adipogenesis. To evaluate the implication of PTP-BL, the mouse orthologue of PTPL1, in adipose tissue biology, we analyzed PTP-BL mRNA expression in adipose tissue in vivo and during proliferation and differentiation of 3T3-L1 pre-adipocytes. To elucidate the role of PTP-BL and of its catalytic activity during adipogenesis we use siRNA techniques in 3T3-L1 pre-adipocytes, and mouse embryonic fibroblasts that lack wildtype PTP-BL and instead express a variant without the PTP domain (Delta P/Delta P MEFs). Here we show that PTP-BL is strongly expressed in white adipose tissue and that PTP-BL transcript and protein levels increase during proliferation and differentiation of 3T3-L1 pre-adipocytes. Strikingly, knockdown of PTP-BL expression in 3T3-L1 adipocytes caused a dramatic decrease in adipogenic gene expression levels (PPAR gamma, aP2) and lipid accumulation but did not interfere with the insulin/Akt pathway. Delta P/Delta P MEFs differentiate into the adipogenic lineage as efficiently as wildtype MEFs. However, when expression of either PTP-BL or PTP-BL Delta P was inhibited a dramatic reduction in the number of MEF-derived adipocytes was observed. These findings demonstrate a key role for PTP-BL in 3T3-L1 and MEF-derived adipocyte differentiation that is independent of its enzymatic activity.


Assuntos
Adipogenia/genética , Regulação para Baixo/genética , Proteína Tirosina Fosfatase não Receptora Tipo 13/genética , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/enzimologia , Animais , Diferenciação Celular , Proliferação de Células , Células Clonais , Regulação Enzimológica da Expressão Gênica , Técnicas de Silenciamento de Genes , Camundongos , Proteína Tirosina Fosfatase não Receptora Tipo 13/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 13/metabolismo
16.
Nat Cell Biol ; 11(8): 1017-23, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19597485

RESUMO

CDK4-pRB-E2F1 cell-cycle regulators are robustly expressed in non-proliferating beta cells, suggesting that besides the control of beta-cell number the CDK4-pRB-E2F1 pathway has a role in beta-cell function. We show here that E2F1 directly regulates expression of Kir6.2, which is a key component of the K(ATP) channel involved in the regulation of glucose-induced insulin secretion. We demonstrate, through chromatin immunoprecipitation analysis from tissues, that Kir6.2 expression is regulated at the promoter level by the CDK4-pRB-E2F1 pathway. Consistently, inhibition of CDK4, or genetic inactivation of E2F1, results in decreased expression of Kir6.2, impaired insulin secretion and glucose intolerance in mice. Furthermore we show that rescue of Kir6.2 expression restores insulin secretion in E2f1(-/-) beta cells. Finally, we demonstrate that CDK4 is activated by glucose through the insulin pathway, ultimately resulting in E2F1 activation and, consequently, increased expression of Kir6.2. In summary we provide evidence that the CDK4-pRB-E2F1 regulatory pathway is involved in glucose homeostasis, defining a new link between cell proliferation and metabolism.


Assuntos
Quinase 4 Dependente de Ciclina/metabolismo , Fator de Transcrição E2F1/metabolismo , Insulina/metabolismo , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Animais , Western Blotting , Células COS , Linhagem Celular , Chlorocebus aethiops , Imunoprecipitação da Cromatina , Fator de Transcrição E2F1/genética , Perfilação da Expressão Gênica , Glucose/farmacologia , Imuno-Histoquímica , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Fosforilação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ligação Proteica , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
17.
Cell Metab ; 9(4): 339-49, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19356715

RESUMO

We show here high levels of expression and secretion of the chemokine CXC ligand 5 (CXCL5) in the macrophage fraction of white adipose tissue (WAT). Moreover, we find that CXCL5 is dramatically increased in serum of human obese compared to lean subjects. Conversely, CXCL5 concentration is decreased in obese subjects after a weight reduction program, or in obese non-insulin-resistant, compared to insulin-resistant, subjects. Most importantly we demonstrate that treatment with recombinant CXCL5 blocks insulin-stimulated glucose uptake in muscle in mice. CXCL5 blocks insulin signaling by activating the Jak2/STAT5/SOCS2 pathway. Finally, by treating obese, insulin-resistant mice with either anti-CXCL5 neutralizing antibodies or antagonists of CXCR2, which is the CXCL5 receptor, we demonstrate that CXCL5 mediates insulin resistance. Furthermore CXCR2-/- mice are protected against obesity-induced insulin resistance. Taken together, these results show that secretion of CXCL5 by WAT resident macrophages represents a link between obesity, inflammation, and insulin resistance.


Assuntos
Tecido Adiposo/metabolismo , Quimiocina CXCL5/metabolismo , Resistência à Insulina , Obesidade/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/efeitos dos fármacos , Animais , Quimiocina CXCL5/deficiência , Quimiocina CXCL5/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Rosiglitazona , Transdução de Sinais/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
18.
Aging (Albany NY) ; 1(7): 674-7, 2009 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-20157549

RESUMO

We have recently shown that the CXCL5 chemokine is secreted by adipose tissue in the obese state. We demonstrated that adipose tissue-derived CXCL5 mediates insulin resistance in muscle. We speculate in this paper that CXCL5 could also mediate other obesity, and diabetes-derived pathologies, such as cardiovascular disease, retinopathy, or inflammatory bowel disease. In this scenario CXCL5 targeted therapy would prevent not only the development of type II diabetes in obese subjects, but also several other obesity-related co morbidities. Finally we propose to analyze the CXCL5 gene to find particular polymorphisms that could predict the development of type II diabetes in obese subjects.


Assuntos
Quimiocina CXCL5/metabolismo , Complicações do Diabetes/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Obesidade/metabolismo , Animais , Quimiocina CXCL5/genética , Complicações do Diabetes/etiologia , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/terapia , Humanos , Resistência à Insulina/fisiologia , Obesidade/complicações , Polimorfismo Genético/genética , Receptores de Interleucina-8B/metabolismo
19.
Mol Pharmacol ; 74(5): 1359-66, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18669446

RESUMO

We have reported recently that the chemokine interleukin 8 (IL-8)/CXCL8 was overexpressed in invasive estrogen receptor (ERalpha)-negative breast cancer cells compared with ERalpha-positive breast cancer cells. We now demonstrate that histone deacetylases (HDACs) play an essential role in the regulation of IL-8 gene expression in ERalpha-positive MCF-7 breast cancer cells. Treatment of MCF-7 cells with the HDAC inhibitor trichostatin A (TSA) led to a strong up-regulation of IL-8 protein and RNA levels in MCF-7 cells. The up-regulation of IL-8 in MCF-7 cells was time- and concentration-dependent. Moreover, run-on and transfection experiments demonstrated that IL-8 induction by HDAC inhibitors was transcriptional and involved mainly the nuclear factor-kappaB (NF-kappaB) site of the IL-8 promoter. These observations are corroborated by an up-regulation of NF-kappaB activity in MCF-7 cells in the presence of TSA. In addition, blocking NF-kappaB pathway by adenoviral delivery of a dominant-negative IkappaBorIkappaB kinase complex 2 (IKK2) mutant abolished IL-8 gene induction by histone deacetylase inhibitors. HDAC inhibitors triggered IKK phosphorylation and up-regulated p65 nuclear translocation, although they decreased the protein levels of IkappaBalpha, which accounts for NF-kappaB activation. TSA increased binding of acetylated histone 3 to the IL-8 gene promoter. In summary, our results demonstrate that NF-kappaB pathway repression by HDAC is responsible for the low expression of IL-8 in ERalpha-positive breast cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Histona Desacetilases/metabolismo , Interleucina-8/metabolismo , NF-kappa B/metabolismo , Adenoviridae/genética , Sequência de Bases , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Humanos , Interleucina-8/genética , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Recombinação Genética , Transdução de Sinais
20.
Endocrinology ; 149(11): 5706-12, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18635652

RESUMO

Circulating free fatty acids are a reflection of the balance between lipogenesis and lipolysis that takes place mainly in adipose tissue. We found that mice deficient for regulator of G protein signaling (RGS)-4 have increased circulating catecholamines, and increased free fatty acids. Consequently, RGS4-/- mice have increased concentration of circulating free fatty acids; abnormally accumulate fatty acids in liver, resulting in liver steatosis; and show a higher degree of glucose intolerance and decreased insulin secretion in pancreas. We show in this study that RGS4 controls adipose tissue lipolysis through regulation of the secretion of catecholamines by adrenal glands. RGS4 controls the balance between adipose tissue lipolysis and lipogenesis, secondary to its role in the regulation of catecholamine secretion by adrenal glands. RGS4 therefore could be a good target for the treatment of metabolic diseases.


Assuntos
Ácidos Graxos/metabolismo , Glucose/metabolismo , Homeostase/genética , Proteínas RGS/fisiologia , Células 3T3-L1 , Tecido Adiposo/metabolismo , Animais , Células Cultivadas , Dieta Aterogênica , Jejum/sangue , Ácidos Graxos/sangue , Fígado Gorduroso/complicações , Fígado Gorduroso/genética , Hiperglicemia/complicações , Hiperglicemia/genética , Hiperinsulinismo/complicações , Hiperinsulinismo/genética , Insulina/metabolismo , Secreção de Insulina , Lipogênese/genética , Lipólise/genética , Camundongos , Camundongos Knockout , Proteínas RGS/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...