Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Front Mol Neurosci ; 15: 841892, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35250480

RESUMO

Alzheimer's disease (AD) is the leading cause of dementia. While impaired glucose homeostasis has been shown to increase AD risk and pathological loss of tau function, the latter has been suggested to contribute to the emergence of the glucose homeostasis alterations observed in AD patients. However, the links between tau impairments and glucose homeostasis, remain unclear. In this context, the present study aimed at investigating the metabolic phenotype of a new tau knock-in (KI) mouse model, expressing, at a physiological level, a human tau protein bearing the P301L mutation under the control of the endogenous mouse Mapt promoter. Metabolic investigations revealed that, while under chow diet tau KI mice do not exhibit significant metabolic impairments, male but not female tau KI animals under High-Fat Diet (HFD) exhibited higher insulinemia as well as glucose intolerance as compared to control littermates. Using immunofluorescence, tau protein was found colocalized with insulin in the ß cells of pancreatic islets in both mouse (WT, KI) and human pancreas. Isolated islets from tau KI and tau knock-out mice exhibited impaired glucose-stimulated insulin secretion (GSIS), an effect recapitulated in the mouse pancreatic ß-cell line (MIN6) following tau knock-down. Altogether, our data indicate that loss of tau function in tau KI mice and, particularly, dysfunction of pancreatic ß cells might promote glucose homeostasis impairments and contribute to metabolic changes observed in AD.

3.
Front Mol Neurosci ; 14: 808603, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35058750

RESUMO

The nuclear bile acid (BA) receptor farnesoid X receptor (FXR) is a major regulator of metabolic/energy homeostasis in peripheral organs. Indeed, enterohepatic-expressed FXR controls metabolic processes (BA, glucose and lipid metabolism, fat mass, body weight). The central nervous system (CNS) regulates energy homeostasis in close interaction with peripheral organs. While FXR has been reported to be expressed in the brain, its function has not been studied so far. We studied the role of FXR in brain control of energy homeostasis by treating wild-type and FXR-deficient mice by intracerebroventricular (ICV) injection with the reference FXR agonist GW4064. Here we show that pharmacological activation of brain FXR modifies energy homeostasis by affecting brown adipose tissue (BAT) function. Brain FXR activation decreases the rate-limiting enzyme in catecholamine synthesis, tyrosine hydroxylase (TH), and consequently the sympathetic tone. FXR activation acts by inhibiting hypothalamic PKA-CREB induction of TH expression. These findings identify a function of brain FXR in the control of energy homeostasis and shed new light on the complex control of energy homeostasis by BA through FXR.

4.
J Biol Chem ; 295(50): 17310-17322, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33037071

RESUMO

In addition to their well-known role in the control of cellular proliferation and cancer, cell cycle regulators are increasingly identified as important metabolic modulators. Several GWAS have identified SNPs near CDKN2A, the locus encoding for p16INK4a (p16), associated with elevated risk for cardiovascular diseases and type-2 diabetes development, two pathologies associated with impaired hepatic lipid metabolism. Although p16 was recently shown to control hepatic glucose homeostasis, it is unknown whether p16 also controls hepatic lipid metabolism. Using a combination of in vivo and in vitro approaches, we found that p16 modulates fasting-induced hepatic fatty acid oxidation (FAO) and lipid droplet accumulation. In primary hepatocytes, p16-deficiency was associated with elevated expression of genes involved in fatty acid catabolism. These transcriptional changes led to increased FAO and were associated with enhanced activation of PPARα through a mechanism requiring the catalytic AMPKα2 subunit and SIRT1, two known activators of PPARα. By contrast, p16 overexpression was associated with triglyceride accumulation and increased lipid droplet numbers in vitro, and decreased ketogenesis and hepatic mitochondrial activity in vivo Finally, gene expression analysis of liver samples from obese patients revealed a negative correlation between CDKN2A expression and PPARA and its target genes. Our findings demonstrate that p16 represses hepatic lipid catabolism during fasting and may thus participate in the preservation of metabolic flexibility.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Ácidos Graxos/metabolismo , Fígado/metabolismo , Mitocôndrias Hepáticas/metabolismo , PPAR alfa/metabolismo , Transdução de Sinais , Sirtuína 1/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Inibidor p16 de Quinase Dependente de Ciclina/genética , Ácidos Graxos/genética , Estudo de Associação Genômica Ampla , Humanos , Gotículas Lipídicas/metabolismo , Camundongos , Camundongos Knockout , Mitocôndrias Hepáticas/genética , Obesidade/genética , Obesidade/metabolismo , Oxirredução , PPAR alfa/genética , Sirtuína 1/genética
5.
Sci Rep ; 10(1): 174, 2020 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-31932631

RESUMO

The gut microbiota participates in the control of energy homeostasis partly through fermentation of dietary fibers hence producing short-chain fatty acids (SCFAs), which in turn promote the secretion of the incretin Glucagon-Like Peptide-1 (GLP-1) by binding to the SCFA receptors FFAR2 and FFAR3 on enteroendocrine L-cells. We have previously shown that activation of the nuclear Farnesoid X Receptor (FXR) decreases the L-cell response to glucose. Here, we investigated whether FXR also regulates the SCFA-induced GLP-1 secretion. GLP-1 secretion in response to SCFAs was evaluated ex vivo in murine colonic biopsies and in colonoids of wild-type (WT) and FXR knock-out (KO) mice, in vitro in GLUTag and NCI-H716 L-cells activated with the synthetic FXR agonist GW4064 and in vivo in WT and FXR KO mice after prebiotic supplementation. SCFA-induced GLP-1 secretion was blunted in colonic biopsies from GW4064-treated mice and enhanced in FXR KO colonoids. In vitro FXR activation inhibited GLP-1 secretion in response to SCFAs and FFAR2 synthetic ligands, mainly by decreasing FFAR2 expression and downstream Gαq-signaling. FXR KO mice displayed elevated colonic FFAR2 mRNA levels and increased plasma GLP-1 levels upon local supply of SCFAs with prebiotic supplementation. Our results demonstrate that FXR activation decreases L-cell GLP-1 secretion in response to inulin-derived SCFA by reducing FFAR2 expression and signaling. Inactivation of intestinal FXR using bile acid sequestrants or synthetic antagonists in combination with prebiotic supplementation may be a promising therapeutic approach to boost the incretin axis in type 2 diabetes.


Assuntos
Colo/metabolismo , Ácidos Graxos Voláteis/farmacologia , Peptídeo 1 Semelhante ao Glucagon/antagonistas & inibidores , Microbiota , Receptores Citoplasmáticos e Nucleares/fisiologia , Animais , Colo/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
6.
Neurobiol Dis ; 125: 14-22, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30665005

RESUMO

Accumulation of hyper-phosphorylated and aggregated Tau proteins is a neuropathological hallmark of Alzheimer's Disease (AD) and Tauopathies. AD patient brains also exhibit insulin resistance. Whereas, under normal physiological conditions insulin signaling in the brain mediates plasticity and memory formation, it can also regulate peripheral energy homeostasis. Thus, in AD, brain insulin resistance affects both cognitive and metabolic changes described in these patients. While a role of Aß oligomers and APOE4 towards the development of brain insulin resistance emerged, contribution of Tau pathology has been largely overlooked. Our recent data demonstrated that one of the physiological function of Tau is to sustain brain insulin signaling. We postulated that under pathological conditions, hyper-phosphorylated/aggregated Tau is likely to lose this function and to favor the development of brain insulin resistance. This hypothesis was substantiated by observations from patient brains with pure Tauopathies. To address the potential link between Tau pathology and brain insulin resistance, we have evaluated the brain response to insulin in a transgenic mouse model of AD-like Tau pathology (THY-Tau22). Using electrophysiological and biochemical evaluations, we surprisingly observed that, at a time when Tau pathology and cognitive deficits are overt and obvious, the hippocampus of THY-Tau22 mice exhibits enhanced response to insulin. In addition, we demonstrated that the ability of i.c.v. insulin to promote body weight loss is enhanced in THY-Tau22 mice. In line with this, THY-Tau22 mice exhibited a lower body weight gain, hypoleptinemia and hypoinsulinemia and finally a metabolic resistance to high-fat diet. The present data highlight that the brain of transgenic Tau mice exhibit enhanced brain response to insulin. Whether these observations are ascribed to the development of Tau pathology, and therefore relevant to human Tauopathies, or unexpectedly results from the Tau transgene overexpression is debatable and discussed.


Assuntos
Encéfalo/metabolismo , Insulina/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Resistência à Insulina/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas tau/genética
7.
J Hepatol ; 69(5): 1099-1109, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29981427

RESUMO

BACKGROUND & AIMS: Embedded into a complex signaling network that coordinates glucose uptake, usage and production, the nuclear bile acid receptor FXR is expressed in several glucose-processing organs including the liver. Hepatic gluconeogenesis is controlled through allosteric regulation of gluconeogenic enzymes and by glucagon/cAMP-dependent transcriptional regulatory pathways. We aimed to elucidate the role of FXR in the regulation of fasting hepatic gluconeogenesis. METHODS: The role of FXR in hepatic gluconeogenesis was assessed in vivo and in mouse primary hepatocytes. Gene expression patterns in response to glucagon and FXR agonists were characterized by quantitative reverse transcription PCR and microarray analysis. FXR phosphorylation by protein kinase A was determined by mass spectrometry. The interaction of FOXA2 with FXR was identified by cistromic approaches and in vitro protein-protein interaction assays. The functional impact of the crosstalk between FXR, the PKA and FOXA2 signaling pathways was assessed by site-directed mutagenesis, transactivation assays and restoration of FXR expression in FXR-deficient hepatocytes in which gene expression and glucose production were assessed. RESULTS: FXR positively regulates hepatic glucose production through two regulatory arms, the first one involving protein kinase A-mediated phosphorylation of FXR, which allowed for the synergistic activation of gluconeogenic genes by glucagon, agonist-activated FXR and CREB. The second arm involves the inhibition of FXR's ability to induce the anti-gluconeogenic nuclear receptor SHP by the glucagon-activated FOXA2 transcription factor, which physically interacts with FXR. Additionally, knockdown of Foxa2 did not alter glucagon-induced and FXR agonist enhanced expression of gluconeogenic genes, suggesting that the PKA and FOXA2 pathways regulate distinct subsets of FXR responsive genes. CONCLUSIONS: Thus, hepatic glucose production is regulated during physiological fasting by FXR, which integrates the glucagon/cAMP signal and the FOXA2 signal, by being post-translationally modified, and by engaging in protein-protein interactions, respectively. LAY SUMMARY: Activation of the nuclear bile acid receptor FXR regulates gene expression networks, controlling lipid, cholesterol and glucose metabolism, which are mostly effective after eating. Whether FXR exerts critical functions during fasting is unknown. The results of this study show that FXR transcriptional activity is regulated by the glucagon/protein kinase A and the FOXA2 signaling pathways, which act on FXR through phosphorylation and protein-protein interactions, respectively, to increase hepatic glucose synthesis.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Jejum/metabolismo , Gluconeogênese , Fator 3-beta Nuclear de Hepatócito/fisiologia , Fígado/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Animais , Regulação da Expressão Gênica , Glucagon/fisiologia , Glucose/metabolismo , Hepatócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação
8.
J Exp Med ; 214(8): 2257-2269, 2017 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-28652303

RESUMO

The molecular pathways underlying tau pathology-induced synaptic/cognitive deficits and neurodegeneration are poorly understood. One prevalent hypothesis is that hyperphosphorylation, misfolding, and fibrillization of tau impair synaptic plasticity and cause degeneration. However, tau pathology may also result in the loss of specific physiological tau functions, which are largely unknown but could contribute to neuronal dysfunction. In the present study, we uncovered a novel function of tau in its ability to regulate brain insulin signaling. We found that tau deletion leads to an impaired hippocampal response to insulin, caused by altered IRS-1 and PTEN (phosphatase and tensin homologue on chromosome 10) activities. Our data also demonstrate that tau knockout mice exhibit an impaired hypothalamic anorexigenic effect of insulin that is associated with energy metabolism alterations. Consistently, we found that tau haplotypes are associated with glycemic traits in humans. The present data have far-reaching clinical implications and raise the hypothesis that pathophysiological tau loss-of-function favors brain insulin resistance, which is instrumental for cognitive and metabolic impairments in Alzheimer's disease patients.


Assuntos
Encéfalo/metabolismo , Resistência à Insulina , Proteínas tau/fisiologia , Animais , Encéfalo/fisiologia , Disfunção Cognitiva/etiologia , Haplótipos , Hipocampo/fisiologia , Humanos , Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
Nat Commun ; 6: 7629, 2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-26134028

RESUMO

Bile acids are signalling molecules, which activate the transmembrane receptor TGR5 and the nuclear receptor FXR. BA sequestrants (BAS) complex bile acids in the intestinal lumen and decrease intestinal FXR activity. The BAS-BA complex also induces glucagon-like peptide-1 (GLP-1) production by L cells which potentiates ß-cell glucose-induced insulin secretion. Whether FXR is expressed in L cells and controls GLP-1 production is unknown. Here, we show that FXR activation in L cells decreases proglucagon expression by interfering with the glucose-responsive factor Carbohydrate-Responsive Element Binding Protein (ChREBP) and GLP-1 secretion by inhibiting glycolysis. In vivo, FXR deficiency increases GLP-1 gene expression and secretion in response to glucose hence improving glucose metabolism. Moreover, treatment of ob/ob mice with the BAS colesevelam increases intestinal proglucagon gene expression and improves glycaemia in a FXR-dependent manner. These findings identify the FXR/GLP-1 pathway as a new mechanism of BA control of glucose metabolism and a pharmacological target for type 2 diabetes.


Assuntos
Células Enteroendócrinas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/genética , Mucosa Intestinal/metabolismo , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Animais , Anticolesterolemiantes/farmacologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Ácidos e Sais Biliares/metabolismo , Glicemia/metabolismo , Cloridrato de Colesevelam/farmacologia , Colo/citologia , Colo/metabolismo , Dieta Hiperlipídica , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Glicólise , Humanos , Íleo/citologia , Íleo/metabolismo , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Intestinos/citologia , Jejuno/citologia , Jejuno/metabolismo , Camundongos , Camundongos Knockout , Camundongos Obesos , Proteínas Nucleares/metabolismo , Obesidade/genética , Obesidade/metabolismo , Proglucagon/efeitos dos fármacos , Proglucagon/genética , Proglucagon/metabolismo , Receptores Acoplados a Proteínas G/genética , Sequestrantes/farmacologia , Transdução de Sinais , Fatores de Transcrição/metabolismo
10.
Diabetes ; 63(10): 3199-209, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24789920

RESUMO

Type 2 diabetes (T2D) is hallmarked by insulin resistance, impaired insulin secretion, and increased hepatic glucose production. The worldwide increasing prevalence of T2D calls for efforts to understand its pathogenesis in order to improve disease prevention and management. Recent genome-wide association studies have revealed strong associations between the CDKN2A/B locus and T2D risk. The CDKN2A/B locus contains genes encoding cell cycle inhibitors, including p16(Ink4a), which have not yet been implicated in the control of hepatic glucose homeostasis. Here, we show that p16(Ink4a) deficiency enhances fasting-induced hepatic glucose production in vivo by increasing the expression of key gluconeogenic genes. p16(Ink4a) downregulation leads to an activation of PKA-CREB-PGC1α signaling through increased phosphorylation of PKA regulatory subunits. Taken together, these results provide evidence that p16(Ink4a) controls fasting glucose homeostasis and could as such be involved in T2D development.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Jejum/metabolismo , Gluconeogênese/fisiologia , Fígado/metabolismo , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Camundongos , Camundongos Knockout , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/metabolismo
11.
Diabetes ; 62(5): 1681-8, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23250356

RESUMO

The τ pathology found in Alzheimer disease (AD) is crucial in cognitive decline. Midlife development of obesity, a major risk factor of insulin resistance and type 2 diabetes, increases the risk of dementia and AD later in life. The impact of obesity on AD risk has been suggested to be related to central insulin resistance, secondary to peripheral insulin resistance. The effects of diet-induced obesity (DIO) on τ pathology remain unknown. In this study, we evaluated effects of a high-fat diet, given at an early pathological stage, in the THY-Tau22 transgenic mouse model of progressive AD-like τ pathology. We found that early and progressive obesity potentiated spatial learning deficits as well as hippocampal τ pathology at a later stage. Surprisingly, THY-Tau22 mice did not exhibit peripheral insulin resistance. Further, pathological worsening occurred while hippocampal insulin signaling was upregulated. Together, our data demonstrate that DIO worsens τ phosphorylation and learning abilities in τ transgenic mice independently from peripheral/central insulin resistance.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Hipocampo/metabolismo , Resistência à Insulina , Obesidade/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Comportamento Animal , Hipocampo/patologia , Proteínas Substratos do Receptor de Insulina/biossíntese , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Deficiências da Aprendizagem/etiologia , Masculino , Transtornos da Memória/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Obesidade/etiologia , Obesidade/patologia , Obesidade/fisiopatologia , Fosforilação , Processamento de Proteína Pós-Traducional , Distribuição Aleatória , Transdução de Sinais , Comportamento Espacial , Tauopatias/etiologia , Tauopatias/patologia , Tauopatias/fisiopatologia , Regulação para Cima , Proteínas tau/genética
12.
PLoS One ; 7(3): e32440, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22403661

RESUMO

OBJECTIVE: A genomic region near the CDKN2A locus, encoding p16(INK4a), has been associated to type 2 diabetes and atherosclerotic vascular disease, conditions in which inflammation plays an important role. Recently, we found that deficiency of p16(INK4a) results in decreased inflammatory signaling in murine macrophages and that p16(INK4a) influences the phenotype of human adipose tissue macrophages. Therefore, we investigated the influence of immune cell p16(INK4a) on glucose tolerance and atherosclerosis in mice. METHODS AND RESULTS: Bone marrow p16(INK4a)-deficiency in C57Bl6 mice did not influence high fat diet-induced obesity nor plasma glucose and lipid levels. Glucose tolerance tests showed no alterations in high fat diet-induced glucose intolerance. While bone marrow p16(INK4a)-deficiency did not affect the gene expression profile of adipose tissue, hepatic expression of the alternative markers Chi3l3, Mgl2 and IL10 was increased and the induction of pro-inflammatory Nos2 was restrained on the high fat diet. Bone marrow p16(INK4a)-deficiency in low density lipoprotein receptor-deficient mice did not affect western diet-induced atherosclerotic plaque size or morphology. In line, plasma lipid levels remained unaffected and p16(INK4a)-deficient macrophages displayed equal cholesterol uptake and efflux compared to wild type macrophages. CONCLUSION: Bone marrow p16(INK4a)-deficiency does not affect plasma lipids, obesity, glucose tolerance or atherosclerosis in mice.


Assuntos
Aterosclerose/metabolismo , Aterosclerose/patologia , Medula Óssea/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Glucose/metabolismo , Homeostase , Obesidade/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/metabolismo , Humanos , Hiperlipidemias/metabolismo , Hiperlipidemias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/induzido quimicamente , Receptores de LDL/deficiência
13.
J Steroid Biochem Mol Biol ; 130(3-5): 126-37, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22033286

RESUMO

Today, we are witnessing a rising incidence of obesity worldwide. This increase is due to a sedentary life style, an increased caloric intake and a decrease in physical activity. Obesity contributes to the appearance of type 2 diabetes, dyslipidemia and cardiovascular complications due to atherosclerosis, and nephropathy. Therefore, the development of new therapeutic strategies may become a necessity. Given the metabolism controlling properties of nuclear receptors in peripheral organs (such as liver, adipose tissues, pancreas) and their implication in various processes underlying metabolic diseases, they constitute interesting therapeutic targets for obesity, dyslipidemia, cardiovascular disease and type 2 diabetes. The recent identification of the central nervous system as a player in the control of peripheral metabolism opens new avenues to our understanding of the pathophysiology of obesity and type 2 diabetes and potential novel ways to treat these diseases. While the metabolic functions of nuclear receptors in peripheral organs have been extensively investigated, little is known about their functions in the brain, in particular with respect to brain control of energy homeostasis. This review provides an overview of the relationships between nuclear receptors in the brain, mainly at the hypothalamic level, and the central regulation of energy homeostasis. In this context, we will particularly focus on the role of PPARα, PPARγ, LXR and Rev-erbα.


Assuntos
Adiposidade/fisiologia , Encéfalo/metabolismo , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Receptores Nucleares Órfãos/metabolismo , PPAR alfa/metabolismo , PPAR gama/metabolismo , Animais , Sistema Nervoso Autônomo/fisiologia , Diabetes Mellitus Tipo 2/metabolismo , Dislipidemias/metabolismo , Ingestão de Energia/fisiologia , Metabolismo Energético/fisiologia , Humanos , Receptores X do Fígado , Camundongos , Obesidade/metabolismo , Ratos
14.
Blood ; 118(9): 2556-66, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21636855

RESUMO

The CDKN2A locus, which contains the tumor suppressor gene p16(INK4a), is associated with an increased risk of age-related inflammatory diseases, such as cardiovascular disease and type 2 diabetes, in which macrophages play a crucial role. Monocytes can polarize toward classically (CAMϕ) or alternatively (AAMϕ) activated macrophages. However, the molecular mechanisms underlying the acquisition of these phenotypes are not well defined. Here, we show that p16(INK4a) deficiency (p16(-/-)) modulates the macrophage phenotype. Transcriptome analysis revealed that p16(-/-) BM-derived macrophages (BMDMs) exhibit a phenotype resembling IL-4-induced macrophage polarization. In line with this observation, p16(-/-) BMDMs displayed a decreased response to classically polarizing IFNγ and LPS and an increased sensitivity to alternative polarization by IL-4. Furthermore, mice transplanted with p16(-/-) BM displayed higher hepatic AAMϕ marker expression levels on Schistosoma mansoni infection, an in vivo model of AAMϕ phenotype skewing. Surprisingly, p16(-/-) BMDMs did not display increased IL-4-induced STAT6 signaling, but decreased IFNγ-induced STAT1 and lipopolysaccharide (LPS)-induced IKKα,ß phosphorylation. This decrease correlated with decreased JAK2 phosphorylation and with higher levels of inhibitory acetylation of STAT1 and IKKα,ß. These findings identify p16(INK4a) as a modulator of macrophage activation and polarization via the JAK2-STAT1 pathway with possible roles in inflammatory diseases.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Genes p16 , Inflamação/genética , Janus Quinase 2/fisiologia , Ativação de Macrófagos , Fator de Transcrição STAT1/fisiologia , Animais , Transplante de Medula Óssea , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Citocinas/biossíntese , Quinase I-kappa B/fisiologia , Interferon gama/farmacologia , Interleucina-4/farmacologia , Lipopolissacarídeos/farmacologia , Fígado/metabolismo , Fígado/patologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Processamento de Proteína Pós-Traducional , Quimera por Radiação , Fator de Transcrição STAT6/fisiologia , Esquistossomose/imunologia , Transdução de Sinais
15.
Atherosclerosis ; 218(1): 69-76, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21529810

RESUMO

BACKGROUND: Atherosclerosis and non-alcoholic fatty liver disease (NAFLD) are complex pathologies characterized by lipid accumulation, chronic inflammation and extensive tissue remodelling. Microparticles (MPs), small membrane vesicles produced by activated and apoptotic cells, might not only be biomarkers, but also functional actors in these pathologies. The apoE2-KI mouse is a model of atherosclerosis and NAFLD. Activation of the nuclear receptor PPARα decreases atherosclerosis and components of non-alcoholic steatohepatitis (NASH) in the apoE2-KI mouse. OBJECTIVES: (1) To determine whether MPs are present in atherosclerotic lesions, liver and plasma during atherosclerosis and NASH progression in apoE2-KI mice, and (2) to study whether PPARα activation modulates MP concentrations. METHODS: ApoE2-KI mice were fed a Western diet to induce atherosclerosis and NASH. MPs were isolated from atherosclerotic lesions, liver and blood and quantified by flow cytometry. RESULTS: An increase of MPs was observed in the atherosclerotic lesions and in the liver of apoE2-KI mice upon Western diet feeding. PPARα activation with fenofibrate decreased MP levels in the atherosclerotic lesions in a PPARα-dependent manner, but did not influence MP concentrations in the liver. CONCLUSION: Here we report that MPs are present in atherosclerotic lesions and in the liver of apoE2-KI mice. Their concentration increased during atherosclerosis and NASH development. PPARα activation differentially modulates MP levels in a tissue-specific manner.


Assuntos
Aterosclerose/metabolismo , Fígado Gorduroso/metabolismo , Fígado/metabolismo , PPAR alfa/metabolismo , Animais , Biomarcadores/metabolismo , Micropartículas Derivadas de Células/metabolismo , Modelos Animais de Doenças , Feminino , Fenofibrato/química , Citometria de Fluxo/métodos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Hepatopatia Gordurosa não Alcoólica
16.
Mol Cell Neurosci ; 37(3): 454-70, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18083596

RESUMO

Neural and mesenchymal stem cells have been proposed as alternative sources of cells for transplantation into the brain in neurodegenerative disorders. However, the endogenous factors controlling their engraftment within the injured parenchyma remain ill-defined. Here, we demonstrate significant engraftment of undifferentiated exogenous mesenchymal or neural stem cells throughout the lesioned area in a rat model for Huntington's disease, as late as 8 weeks post-transplantation. We show that stem cell factor (SCF), strongly up-regulated within host cells in the damaged striatum, is able to activate the SCF receptor c-kit and its signaling pathway and to promote the migration and proliferation of mesenchymal and neural stem cells in vitro. Furthermore, c-kit receptor blockade alters neural stem cell distribution within the lesioned striatum. Host SCF expression is observed in atypical cells expressing glial fibrillary acidic protein and doublecortin in the lesioned striatum and in migrating doublecortin-positive progenitors. Taken together, these data demonstrate that SCF produced in situ in the lesioned striatum is an important factor in promoting the engraftment of stem cells within the lesioned brain.


Assuntos
Doença de Huntington/cirurgia , Neurônios/fisiologia , Fator de Células-Tronco/farmacologia , Transplante de Células-Tronco/métodos , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Análise de Variância , Animais , Contagem de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Células Cultivadas , Corpo Estriado/patologia , Modelos Animais de Doenças , Proteína Duplacortina , Embrião de Mamíferos , Doença de Huntington/etiologia , Doença de Huntington/patologia , Masculino , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Compostos Orgânicos/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Ratos , Ratos Wistar , Células-Tronco/classificação , Fatores de Tempo
17.
J Neuropathol Exp Neurol ; 66(5): 363-71, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17483693

RESUMO

Inhibition of adenosine A2A receptors (A2ARs) is neuroprotective in several experimental models of striatal diseases. However, the mechanisms elicited by A2AR blockade are only partially known, and critical aspects about the potential beneficial effects of A2AR antagonism in models of neurodegeneration still await elucidation. In the present study, we analyzed the influence of the selective A2AR antagonist SCH 58261 in a rat model of striatal excitotoxicity obtained by unilateral intrastriatal injection of quinolinic acid (QA). We found that SCH 58261 differently affected the expression of cyclooxygenase-2 (COX-2) induced by QA in cortex and striatum. The antagonist enhanced COX-2 expression in cortical neurons and prevented it in striatal microglia-like cells. Similarly, SCH 58261 differently regulated astrogliosis and microglial activation in the 2 brain regions. In addition, the A2AR antagonist prevented the QA-induced increase in striatal brain-derived neurotrophic factor levels. Because COX-2 activity has been linked to excitotoxic processes and because brain-derived neurotrophic factor depletion has been observed in mouse models as well as in patients with Huntington disease, we suggest that the final outcome of A2AR blockade (namely neuroprotection vs neurodegeneration) is likely to depend on the balance among its various and region-specific effects.


Assuntos
Antagonistas do Receptor A2 de Adenosina , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Corpo Estriado/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Doenças Neurodegenerativas/tratamento farmacológico , Pirimidinas/farmacologia , Triazóis/farmacologia , Animais , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Interações Medicamentosas , Radicais Livres/metabolismo , Masculino , Doenças Neurodegenerativas/induzido quimicamente , Doenças Neurodegenerativas/patologia , Neuroglia/efeitos dos fármacos , Prostaglandinas/metabolismo , Ácido Quinolínico , Ratos , Ratos Wistar
18.
Neurosci Lett ; 401(1-2): 25-9, 2006 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-16540243

RESUMO

Transgenic rats expressing a mutated form of the human Cu/Zn superoxide dismutase (hSOD1(G93A)) develop an amyotrophic lateral sclerosis (ALS)-like phenotype, including motor neurone degeneration and reactive gliosis in the spinal cord. This study aimed at examining the presence of endogenous neural progenitors in the lumbar spinal cord of these rats at the end-stage of the disease. Immunohistochemical data clearly demonstrated the induced expression of the stem cell factor reported as a chemoattractant and survival factor for neural stem cells as well as nestin (neuro-epithelial stem cell intermediate filament) in the spinal cord sections. While the stem cell factor immunolabelling appeared diffuse throughout the gray matter, nestin labelling was restricted to clusters within the ventral horn. Interestingly, as paralysis regularly develops asymmetrically, induction of nestin was only detected on the ipsilateral side of the predominant symptoms. Finally, immunohistochemical detection of the stem cell factor receptor (c-Kit) revealed its specific induction which coincided with nestin immunolabelling. Together, these results are indicative of endogenous recruitment of neural progenitors within lesioned tissues and could support the development of treatments involving endogenous or exogenous stem cells.


Assuntos
Esclerose Lateral Amiotrófica/genética , Regeneração Nervosa/genética , Paralisia/genética , Medula Espinal/metabolismo , Células-Tronco/metabolismo , Superóxido Dismutase/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular/genética , Movimento Celular/genética , Modelos Animais de Doenças , Lateralidade Funcional/genética , Membro Posterior/inervação , Membro Posterior/fisiopatologia , Humanos , Imuno-Histoquímica , Proteínas de Filamentos Intermediários/metabolismo , Neurônios Motores/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurônios/metabolismo , Paralisia/metabolismo , Paralisia/fisiopatologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Ratos , Recuperação de Função Fisiológica/genética , Medula Espinal/citologia , Medula Espinal/fisiopatologia , Fator de Células-Tronco/metabolismo , Células-Tronco/citologia , Superóxido Dismutase-1
19.
Neurobiol Dis ; 18(1): 206-17, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15649711

RESUMO

Minocycline has been shown to be neuroprotective in various models of neurodegenerative diseases. However, its potential in Huntington's disease (HD) models characterized by calpain-dependent degeneration and inflammation has not been investigated. Here, we have tested minocycline in phenotypic models of HD using 3-nitropropionic acid (3NP) intoxication and quinolinic acid (QA) injections. In the 3NP rat model, where the development of striatal lesions involves calpain, we found that minocycline was not protective, although it attenuated the development of inflammation induced after the onset of striatal degeneration. The lack of minocycline activity on calpain-dependent cell death was also confirmed in vitro using primary striatal cells. Conversely, we found that minocycline reduced lesions and inflammation induced by QA. In cultured cells, minocycline protected against mutated huntingtin and staurosporine, stimulations known to promote caspase-dependent cell death. Altogether, these data suggested that, in HD, minocycline may counteract the development of caspase-dependent neurodegeneration, inflammation, but not calpain-dependent neuronal death.


Assuntos
Doença de Huntington/tratamento farmacológico , Minociclina/farmacologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Animais , Calpaína/efeitos dos fármacos , Calpaína/metabolismo , Caspases/efeitos dos fármacos , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Células Cultivadas , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Encefalite/tratamento farmacológico , Encefalite/fisiopatologia , Encefalite/prevenção & controle , Ácido Glutâmico/metabolismo , Proteína Huntingtina , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Masculino , Minociclina/uso terapêutico , Degeneração Neural/patologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Nitrocompostos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Fenótipo , Propionatos , Ácido Quinolínico , Ratos , Ratos Endogâmicos Lew , Ratos Wistar , Estaurosporina/antagonistas & inibidores
20.
Neurobiol Dis ; 15(1): 152-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14751780

RESUMO

An important aspect of Huntington's disease (HD) pathogenesis which may have important therapeutic implications is that the cellular events leading to cell death may be different in cortical and striatal neurons. In the present study, we characterized cellular changes in cortical and striatal neurons treated with the mitochondrial toxin 3-nitropropionic acid (3NP) in culture. Degeneration induced by 3NP was similar in both striatal and cortical neurons as observed using markers of cell viability and DNA fragmentation. However, in striatal neurons, 3NP produced a marked delocalization of Bad, Bax, cytochrome c and Smac while this was not observed in cortical neurons. Death of striatal neurons was preceded by activation of calpain and was blocked by calpain inhibitor I. In cortical neurons, calpain was not activated and calpain inhibitor I was without effect. In both cell types, caspase-9 and -3 were not activated by 3NP and the caspase inhibitor zVAD-fmk did not provide neuroprotective effect. Interestingly, treatment with staurosporine (STS) triggered caspase-9 and -3 in cortical and striatal cells, suggesting that the molecular machinery related to caspase-dependent apoptosis was functional in both cell types even though this machinery was not involved in 3NP toxicity. The present results clearly demonstrate that under mitochondrial inhibition, striatal and cortical neurons die through different pathways. This suggests that mitochondrial defects in HD may trigger the death of cortical and striatal neurons through different molecular events.


Assuntos
Córtex Cerebral/enzimologia , Doença de Huntington/enzimologia , Mitocôndrias/enzimologia , Neostriado/enzimologia , Degeneração Neural/enzimologia , Proteínas Proto-Oncogênicas c-bcl-2 , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Inibidores de Caspase , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Respiração Celular/efeitos dos fármacos , Respiração Celular/fisiologia , Células Cultivadas , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Citocromos c/efeitos dos fármacos , Citocromos c/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Feto , Doença de Huntington/fisiopatologia , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Neostriado/patologia , Neostriado/fisiopatologia , Degeneração Neural/induzido quimicamente , Degeneração Neural/fisiopatologia , Neurotoxinas/toxicidade , Nitrocompostos , Propionatos/toxicidade , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteína X Associada a bcl-2 , Proteína de Morte Celular Associada a bcl
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA