Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Med Sci (Paris) ; 38(8-9): 669-678, 2022.
Artigo em Francês | MEDLINE | ID: mdl-36094237

RESUMO

The biological clock is a set of evolutionarily conserved "clock proteins" that generate circadian rhythms in behavior and physiological processes. The clock programs these processes at specific times of the day, allowing the organism to optimize its functions by anticipating predictable daily changes such as day/night, hence sleep/wake or feeding/fasting cycles. Modern lifestyle, i.e., exposure to light at night, shift work and irregular eating patterns and sleep schedules desynchronize the clocks residing in each organ. This dissonance is associated with an increased risk of developing various diseases such as cancer, metabolic, cardiovascular and chronic inflammatory diseases.


Title: Récepteurs nucléaires et rythmes circadiens - Implications dans les maladies inflammatoires. Abstract: L'horloge circadienne programme l'ensemble des processus physiologiques, dont l'activité du système immunitaire, à des moments précis de la journée. Elle permet d'optimiser les fonctions de l'organisme en anticipant les changements quotidiens tels que les cycles jour/nuit. Nos habitudes de vie comme l'exposition à la lumière artificielle ou une prise alimentaire irrégulière désynchronisent cependant cette horloge et provoquent des maladies, par exemple inflammatoires. Au niveau moléculaire, elle consiste en un réseau de facteurs de transcription dont certains sont des récepteurs nucléaires, activables par des ligands. Une meilleure compréhension des rythmes biologiques et du rôle des récepteurs nucléaires de l'horloge circadienne permettrait d'ouvrir un champ thérapeutique nouveau. La chronothérapie qui consiste en l'administration d'un composé pharmacologique au moment de la journée le plus propice, permettrait, en ciblant ces récepteurs, d'optimiser l'efficacité du traitement et d'en réduire les possibles effets secondaires.


Assuntos
Relógios Circadianos , Relógios Circadianos/genética , Ritmo Circadiano/genética , Humanos , Receptores Citoplasmáticos e Nucleares , Sono
2.
Int J Mol Sci ; 22(18)2021 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-34575881

RESUMO

Cardiovascular diseases (CVD) are still the first cause of death worldwide. Their main origin is the development of atherosclerotic plaque, which consists in the accumulation of lipids and inflammatory leucocytes within the vascular wall of large vessels. Beyond dyslipidemia, diabetes, obesity, hypertension and smoking, the alteration of circadian rhythms, in shift workers for instance, has recently been recognized as an additional risk factor. Accordingly, targeting a pro-atherogenic pathway at the right time window, namely chronotherapy, has proven its efficiency in reducing plaque progression without affecting healthy tissues in mice, thus providing the rationale of such an approach to treat CVD and to reduce drug side effects. Nuclear receptors are transcriptional factors involved in the control of many physiological processes. Among them, Rev-erbs and RORs control metabolic homeostasis, inflammatory processes and the biological clock. In this review, we discuss the opportunity to dampen atherosclerosis progression by targeting such ligand-activated core clock components in a (chrono-)therapeutic approach in order to treat CVD.


Assuntos
Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/metabolismo , Relógios Circadianos/genética , Suscetibilidade a Doenças , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Relógios Biológicos/genética , Biomarcadores , Doenças Cardiovasculares/diagnóstico , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Família Multigênica , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Especificidade de Órgãos/genética , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , Transdução de Sinais
3.
Front Endocrinol (Lausanne) ; 12: 630536, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33716981

RESUMO

The innate immune system is the first line of defense specialized in the clearing of invaders whether foreign elements like microbes or self-elements that accumulate abnormally including cellular debris. Inflammasomes are master regulators of the innate immune system, especially in macrophages, and are key sensors involved in maintaining cellular health in response to cytolytic pathogens or stress signals. Inflammasomes are cytoplasmic complexes typically composed of a sensor molecule such as NOD-Like Receptors (NLRs), an adaptor protein including ASC and an effector protein such as caspase 1. Upon stimulation, inflammasome complex components associate to promote the cleavage of the pro-caspase 1 into active caspase-1 and the subsequent activation of pro-inflammatory cytokines including IL-18 and IL-1ß. Deficiency or overactivation of such important sensors leads to critical diseases including Alzheimer diseases, chronic inflammatory diseases, cancers, acute liver diseases, and cardiometabolic diseases. Inflammasomes are tightly controlled by a two-step activation regulatory process consisting in a priming step, which activates the transcription of inflammasome components, and an activation step which leads to the inflammasome complex formation and the subsequent cleavage of pro-IL1 cytokines. Apart from the NF-κB pathway, nuclear receptors have recently been proposed as additional regulators of this pathway. This review will discuss the role of nuclear receptors in the control of the NLRP3 inflammasome and the putative beneficial effect of new modulators of inflammasomes in the treatment of inflammatory diseases including colitis, fulminant hepatitis, cardiac ischemia-reperfusion and brain diseases.


Assuntos
Inflamassomos/metabolismo , Inflamação/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Humanos , Transdução de Sinais/fisiologia
4.
Acta Diabetol ; 57(7): 819-826, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32086613

RESUMO

AIMS: Not all people with obesity become glucose intolerant, suggesting differential activation of cellular pathways. The unfolded protein response (UPR) may contribute to the development of insulin resistance in several organs, but its role in skeletal muscle remains debated. Therefore, we explored the UPR activation in muscle from non-diabetic glucose tolerant or intolerant patients with obesity and the impact of bariatric procedures. METHODS: Muscle biopsies from 22 normoglycemic (NG, blood glucose measured 120 min after an oral glucose tolerance test, G120 < 7.8 mM) and 22 glucose intolerant (GI, G120 between 7.8 and 11.1 mM) patients with obesity were used to measure UPR activation by RTqPCR and western blot. Then, UPR was studied in biopsies from 7 NG and 7 GI patients before and 1 year after bariatric surgery. RESULTS: Binding immunoglobulin protein (BIP) protein was ~ 40% higher in the GI compared to NG subjects. Contrastingly, expression of the UPR-related genes BIP, activating transcription factor 6 (ATF6) and unspliced X-box binding protein 1 (XBP1u) were significantly lower and C/EBP homologous protein (CHOP) tended to decrease (p = 0.08) in GI individuals. While BIP protein positively correlated with fasting blood glucose (r = 0.38, p = 0.01), ATF6 and CHOP were associated with G120 (r = - 0.38 and r = - 0.41, p < 0.05) and the Matsuda index (r = 0.37 and r = 0.38, p < 0.05). Bariatric surgery improved metabolic parameters, associated with higher CHOP expression in GI patients, while ATF6 tended to increase (p = 0.08). CONCLUSIONS: CHOP and ATF6 expression decreased in non-diabetic GI patients with obesity and was modified by bariatric surgery. These genes may contribute to glucose homeostasis in human skeletal muscle.


Assuntos
Cirurgia Bariátrica , Intolerância à Glucose/cirurgia , Músculo Esquelético/metabolismo , Obesidade Mórbida/cirurgia , Resposta a Proteínas não Dobradas , Fator 6 Ativador da Transcrição/genética , Fator 6 Ativador da Transcrição/metabolismo , Adulto , Biópsia , Glicemia/metabolismo , Estudos de Casos e Controles , Estudos de Coortes , Estresse do Retículo Endoplasmático/genética , Estresse do Retículo Endoplasmático/fisiologia , Feminino , Regulação da Expressão Gênica , Intolerância à Glucose/complicações , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Teste de Tolerância a Glucose , Humanos , Resistência à Insulina/fisiologia , Masculino , Músculo Esquelético/patologia , Obesidade Mórbida/complicações , Obesidade Mórbida/metabolismo , Obesidade Mórbida/patologia , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo
5.
Cell Rep ; 29(6): 1410-1418.e6, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31693883

RESUMO

Browning induction or transplantation of brown adipose tissue (BAT) or brown/beige adipocytes derived from progenitor or induced pluripotent stem cells (iPSCs) can represent a powerful strategy to treat metabolic diseases. However, our poor understanding of the mechanisms that govern the differentiation and activation of brown adipocytes limits the development of such therapy. Various genetic factors controlling the differentiation of brown adipocytes have been identified, although most studies have been performed using in vitro cultured pre-adipocytes. We investigate here the differentiation of brown adipocytes from adipose progenitors in the mouse embryo. We demonstrate that the formation of multiple lipid droplets (LDs) is initiated within clusters of glycogen, which is degraded through glycophagy to provide the metabolic substrates essential for de novo lipogenesis and LD formation. Therefore, this study uncovers the role of glycogen in the generation of LDs.


Assuntos
Adipócitos Marrons/metabolismo , Adipogenia/genética , Tecido Adiposo Marrom/metabolismo , Embrião de Mamíferos/metabolismo , Glicogênio/metabolismo , Gotículas Lipídicas/metabolismo , Adipócitos Marrons/ultraestrutura , Tecido Adiposo Marrom/embriologia , Tecido Adiposo Marrom/ultraestrutura , Animais , Autofagia/efeitos dos fármacos , Autofagia/genética , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Células Cultivadas , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Glicogênio/ultraestrutura , Humanos , Gotículas Lipídicas/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , PPAR gama/genética , PPAR gama/metabolismo , RNA Interferente Pequeno , Transcriptoma
6.
Proc Natl Acad Sci U S A ; 115(47): E11033-E11042, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30397120

RESUMO

The nuclear receptor REV-ERBα integrates the circadian clock with hepatic glucose and lipid metabolism by nucleating transcriptional comodulators at genomic regulatory regions. An interactomic approach identified O-GlcNAc transferase (OGT) as a REV-ERBα-interacting protein. By shielding cytoplasmic OGT from proteasomal degradation and favoring OGT activity in the nucleus, REV-ERBα cyclically increased O-GlcNAcylation of multiple cytoplasmic and nuclear proteins as a function of its rhythmically regulated expression, while REV-ERBα ligands mostly affected cytoplasmic OGT activity. We illustrate this finding by showing that REV-ERBα controls OGT-dependent activities of the cytoplasmic protein kinase AKT, an essential relay in insulin signaling, and of ten-of-eleven translocation (TET) enzymes in the nucleus. AKT phosphorylation was inversely correlated to REV-ERBα expression. REV-ERBα enhanced TET activity and DNA hydroxymethylated cytosine (5hmC) levels in the vicinity of REV-ERBα genomic binding sites. As an example, we show that the REV-ERBα/OGT complex modulates SREBP-1c gene expression throughout the fasting/feeding periods by first repressing AKT phosphorylation and by epigenomically priming the Srebf1 promoter for a further rapid response to insulin. Conclusion: REV-ERBα regulates cytoplasmic and nuclear OGT-controlled processes that integrate at the hepatic SREBF1 locus to control basal and insulin-induced expression of the temporally and nutritionally regulated lipogenic SREBP-1c transcript.


Assuntos
Insulina/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/biossíntese , Animais , Linhagem Celular Tumoral , Relógios Circadianos/fisiologia , Regulação da Expressão Gênica/genética , Glucose/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Metabolismo dos Lipídeos/fisiologia , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Acetilglucosaminiltransferases/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
7.
J Biol Methods ; 5(3): e94, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-31453244

RESUMO

The nuclear receptors are transcription factors involved in the regulation of a variety of physiological processes whose activity can be modulated by binding to relevant small molecule ligands. Their dysfunction has been shown to play a role in disease states such as diabetes, cancer, inflammatory diseases, and hormonal resistance ailments, which makes them interesting targets for drug discovery. The nuclear receptor REV-ERBα is involved in regulating the circadian rhythm and metabolism. Its natural ligand is heme and there is significant interest in identifying novel synthetic modulators to serve as tools to characterize its function and to serve as drugs in treating metabolic disorders. To do so, we established a mammalian cell-based two-hybrid assay system capable of measuring the interaction between REV-ERBα and its co-repressor, nuclear co-repressor 1. This assay was validated to industry standard criteria and was used to screen a subset of the LOPAC®1280 library and 29568 compounds from a diverse compound library. Profiling of the primary hits in a panel of counter and selectivity assays confirmed that REV-ERBα activity can be modulated pharmacologically and chemical scaffolds have been identified for optimization.

8.
Gastroenterology ; 154(5): 1449-1464.e20, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29277561

RESUMO

BACKGROUND & AIMS: The innate immune system responds not only to bacterial signals, but also to non-infectious danger-associated molecular patterns that activate the NLRP3 inflammasome complex after tissue injury. Immune functions vary over the course of the day, but it is not clear whether these changes affect the activity of the NLRP3 inflammasome. We investigated whether the core clock component nuclear receptor subfamily 1 group D member 1 (NR1D1, also called Rev-erbα) regulates expression, activity of the NLRP3 inflammasome, and its signaling pathway. METHODS: We collected naïve peritoneal macrophages and plasma, at multiple times of day, from Nr1d1-/- mice and their Nr1d1+/+ littermates (controls) and analyzed expression NLRP3, interleukin 1ß (IL1B, in plasma), and IL18 (in plasma). We also collected bone marrow-derived primary macrophages from these mice. Levels of NR1D1 were knocked down with small hairpin RNAs in human primary macrophages. Bone marrow-derived primary macrophages from mice and human primary macrophages were incubated with lipopolysaccharide (LPS) to induce expression of NLRP3, IL1B, and IL18; cells were incubated with LPS and adenosine triphosphate to activate the NLRP3 complex. We analyzed caspase 1 activity and cytokine secretion. NR1D1 was activated in primary mouse and human macrophages by incubation with SR9009; some of the cells were also incubated with an NLRP3 inhibitor or inhibitors of caspase 1. Nr1d1-/- mice and control mice were given intraperitoneal injections of LPS to induce peritoneal inflammation; plasma samples were isolated and levels of cytokines were measured. Nr1d1-/- mice, control mice, and control mice given injections of SR9009 were given LPS and D-galactosamine to induce fulminant hepatitis and MCC950 to specifically inhibit NLRP3; plasma was collected to measure cytokines and a marker of liver failure (alanine aminotransferase); liver tissues were collected and analyzed by quantitative polymerase chain reaction, immunohistochemistry, and flow cytometry. RESULTS: In peritoneal macrophages, expression of NLRP3 and activation of its complex varied with time of day (circadian rhythm)-this regulation required NR1D1. Primary macrophages from Nr1d1-/- mice and human macrophages with knockdown of NR1D1 had altered expression patterns of NLRP3, compared to macrophages that expressed NR1D1, and altered patterns of IL1B and 1L18 production. Mice with disruption of Nr1d1 developed more-severe acute peritoneal inflammation and fulminant hepatitis than control mice. Incubation of macrophage with the NR1D1 activator SR9009 reduced expression of NLRP3 and secretion of cytokines. Mice given SR9009 developed less-severe liver failure and had longer survival times than mice given saline (control). CONCLUSIONS: In studies of Nr1d1-/- mice and human macrophages with pharmacologic activation of NR1D1, we found NR1D1 to regulate the timing of NLRP3 expression and production of inflammatory cytokines by macrophages. Activation of NR1D1 reduced the severity of peritoneal inflammation and fulminant hepatitis in mice.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Ritmo Circadiano , Inflamassomos/metabolismo , Falência Hepática Aguda/prevenção & controle , Fígado/metabolismo , Macrófagos Peritoneais/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Animais , Caspase 1/metabolismo , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/imunologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Galactosamina , Predisposição Genética para Doença , Inflamassomos/genética , Inflamassomos/imunologia , Lipopolissacarídeos , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/patologia , Falência Hepática Aguda/imunologia , Falência Hepática Aguda/metabolismo , Falência Hepática Aguda/patologia , Ativação de Macrófagos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/patologia , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/agonistas , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/deficiência , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Peritonite/imunologia , Peritonite/metabolismo , Peritonite/prevenção & controle , Fenótipo , Pirrolidinas/farmacologia , Interferência de RNA , Índice de Gravidade de Doença , Transdução de Sinais , Tiofenos/farmacologia , Fatores de Tempo , Transfecção
9.
Lancet ; 391(10115): 59-69, 2018 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-29107324

RESUMO

BACKGROUND: On-pump cardiac surgery provokes a predictable perioperative myocardial ischaemia-reperfusion injury which is associated with poor clinical outcomes. We determined the occurrence of time-of-the-day variation in perioperative myocardial injury in patients undergoing aortic valve replacement and its molecular mechanisms. METHODS: We studied the incidence of major adverse cardiac events in a prospective observational single-centre cohort study of patients with severe aortic stenosis and preserved left ventricular ejection fraction (>50%) who were referred to our cardiovascular surgery department at Lille University Hospital (Lille, France) for aortic valve replacement and underwent surgery in the morning or afternoon. Patients were matched into pairs by propensity score. We also did a randomised study, in which we evaluated perioperative myocardial injury and myocardial samples of patients randomly assigned (1:1) via permuted block randomisation (block size of eight) to undergo isolated aortic valve replacement surgery either in the morning or afternoon. We also evaluated human and rodent myocardium in ex-vivo hypoxia-reoxygenation models and did a transcriptomic analysis in myocardial samples from the randomised patients to identify the signalling pathway(s) involved. The primary objective of the study was to assess whether myocardial tolerance of ischaemia-reperfusion differed depending on the timing of aortic valve replacement surgery (morning vs afternoon), as measured by the occurrence of major adverse cardiovascular events (cardiovascular death, myocardial infarction, and admission to hospital for acute heart failure). The randomised study is registered with ClinicalTrials.gov, number NCT02812901. FINDINGS: In the cohort study (n=596 patients in matched pairs who underwent either morning surgery [n=298] or afternoon surgery [n=298]), during the 500 days following aortic valve replacement, the incidence of major adverse cardiac events was lower in the afternoon surgery group than in the morning group: hazard ratio 0·50 (95% CI 0·32-0·77; p=0·0021). In the randomised study, 88 patients were randomly assigned to undergo surgery in the morning (n=44) or afternoon (n=44); perioperative myocardial injury assessed with the geometric mean of perioperative cardiac troponin T release was significantly lower in the afternoon group than in the morning group (estimated ratio of geometric means for afternoon to morning of 0·79 [95% CI 0·68-0·93; p=0·0045]). Ex-vivo analysis of human myocardium revealed an intrinsic morning-afternoon variation in hypoxia-reoxygenation tolerance, concomitant with transcriptional alterations in circadian gene expression with the nuclear receptor Rev-Erbα being highest in the morning. In a mouse Langendorff model of hypoxia-reoxygenation myocardial injury, Rev-Erbα gene deletion or antagonist treatment reduced injury at the time of sleep-to-wake transition, through an increase in the expression of the ischaemia-reperfusion injury modulator CDKN1a/p21. INTERPRETATION: Perioperative myocardial injury is transcriptionally orchestrated by the circadian clock in patients undergoing aortic valve replacement, and Rev-Erbα antagonism seems to be a pharmacological strategy for cardioprotection. Afternoon surgery might provide perioperative myocardial protection and lead to improved patient outcomes compared with morning surgery. FUNDING: Fondation de France, Fédération Française de Cardiologie, EU-FP7-Eurhythdia, Agence Nationale pour la Recherche ANR-10-LABX-46, and CPER-Centre Transdisciplinaire de Recherche sur la Longévité.


Assuntos
Estenose da Valva Aórtica/cirurgia , Ritmo Circadiano , Implante de Prótese de Valva Cardíaca/efeitos adversos , Traumatismo por Reperfusão Miocárdica/epidemiologia , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Complicações Pós-Operatórias/epidemiologia , Idoso , Idoso de 80 Anos ou mais , Estenose da Valva Aórtica/metabolismo , Estudos de Casos e Controles , Estudos de Coortes , Feminino , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Traumatismo por Reperfusão Miocárdica/metabolismo , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/antagonistas & inibidores , Complicações Pós-Operatórias/metabolismo , Pontuação de Propensão , Transdução de Sinais , Resultado do Tratamento
10.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(9): 901-916, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28529179

RESUMO

Obesity and its associated metabolic disorders represent a major health burden, with economic and social consequences. Although adapted lifestyle and bariatric surgery are effective in reducing body weight, obesity prevalence is still rising. Obese individuals often become insulin-resistant. Obesity impacts on insulin responsive organs, such as the liver, adipose tissue and skeletal muscle, and increases the risk of cardiovascular diseases, type 2 diabetes and cancer. In this review, we discuss the effects of obesity and insulin resistance on skeletal muscle, an important organ for the control of postprandial glucose. The roles of mitochondria and the endoplasmic reticulum in insulin signaling are highlighted and potential innovative research and treatment perspectives are proposed.


Assuntos
Retículo Endoplasmático/metabolismo , Resistência à Insulina/fisiologia , Insulina/metabolismo , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Animais , Glucose/metabolismo , Humanos
11.
Sci Rep ; 6: 35047, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27756900

RESUMO

Cell and animal studies have demonstrated that circadian rhythm is governed by autonomous rhythmicity of clock genes. Although disturbances in circadian rhythm have been implicated in metabolic disease development, it remains unknown whether muscle circadian rhythm is altered in human models of type 2 diabetes. Here we used human primary myotubes (HPM) to investigate if rhythmicity of clock- and metabolic gene expression is altered in donors with obesity or type 2 diabetes compared to metabolically healthy donors. HPM were obtained from skeletal muscle biopsies of four groups: type 2 diabetic patients and their BMI- and age-matched obese controls and from lean, healthy and young endurance trained athletes and their age-matched sedentary controls. HPM were differentiated for 7 days before synchronization by serum shock followed by gene expression profiling over the next 72 hours. HPM display robust circadian rhythms in clock genes, but REVERBA displayed dampened rhythmicity in type 2 diabetes. Furthermore, rhythmicity in NAMPT and SIRT1 expression was only observed in HPM from trained athletes. Rhythmicity in expression of key-regulators of carbohydrate and lipid metabolism was modest. We demonstrate that in human skeletal muscle REVERBA/B, NAMPT and SIRT1 circadian rhythms are affected in donors of sedentary life style and poor health status.


Assuntos
Proteínas CLOCK/genética , Diabetes Mellitus Tipo 2/genética , Fibras Musculares Esqueléticas/fisiologia , Obesidade/genética , Biópsia , Células Cultivadas , Ritmo Circadiano , Diabetes Mellitus Tipo 2/patologia , Exercício Físico , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/patologia , Obesidade/patologia , Adulto Jovem
12.
Cell Rep ; 17(4): 1087-1097, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27760313

RESUMO

To maintain energy homeostasis despite variable energy supply and consumption along the diurnal cycle, the liver relies on a circadian clock synchronized to food timing. Perturbed feeding and fasting cycles have been associated with clock disruption and metabolic diseases; however, the mechanisms are unclear. To address this question, we have constructed a mathematical model of the mammalian circadian clock, incorporating the metabolic sensors SIRT1 and AMPK. The clock response to various temporal patterns of AMPK activation was simulated numerically, mimicking the effects of a normal diet, fasting, and a high-fat diet. The model reproduces the dampened clock gene expression and NAD+ rhythms reported for mice on a high-fat diet and predicts that this effect may be pharmacologically rescued by timed REV-ERB agonist administration. Our model thus identifies altered AMPK signaling as a mechanism leading to clock disruption and its associated metabolic effects and suggests a pharmacological approach to resetting the clock in obesity.


Assuntos
Relógios Circadianos/fisiologia , Jejum/fisiologia , Comportamento Alimentar/fisiologia , Fígado/fisiologia , Modelos Biológicos , Monofosfato de Adenosina/metabolismo , Adenilato Quinase/metabolismo , Animais , Relógios Circadianos/genética , Criptocromos/genética , Criptocromos/metabolismo , Dieta Hiperlipídica , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Camundongos Knockout , Camundongos Obesos , NAD/metabolismo , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/agonistas , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Fenótipo , Sirtuína 1/metabolismo , Fatores de Tempo
13.
Biochem Pharmacol ; 83(7): 823-32, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22172989

RESUMO

Although being a primary objective in the management of type 2 diabetes, optimal glycaemic control is difficult to achieve and usually not maintained over time. Type 2 diabetes is a complex pathology, comprising altered insulin sensitivity and impaired insulin secretion. Recent advances in the understanding of the physiological functions of incretins and their degrading enzyme dipeptidyl-peptidase (DPP)-4 have led to the 'discovery' of a new class of oral anti-diabetic drugs. Several DPP-4 inhibitors (or gliptins) with different chemical structures are now available. These agents inhibit the degradation of the incretins glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) and hence potentiate glucose-dependent insulin secretion. DPP-4 inhibitors inhibit DPP-4 activity by almost 100% in vitro, maintaining a ≥ 80% inhibition throughout the treatment period in vivo, thus prolonging GLP-1 half-life, and significantly reducing HbA1c generally by -0.7 to 0.8% as well as fasting and post-prandial glycaemia. They are well-tolerated with no weight gain and few adverse effects, and, of particular interest, no increase in hypoglycaemic episodes. Although different by their chemical structure and pharmacokinetic properties, the DPP4 inhibitors currently available have proven similar glucose lowering efficacy.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Administração Oral , Animais , Glicemia/metabolismo , Ensaios Clínicos como Assunto , Diabetes Mellitus Tipo 2/metabolismo , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/fisiologia , Inibidores da Dipeptidil Peptidase IV/efeitos adversos , Inibidores da Dipeptidil Peptidase IV/química , Inibidores da Dipeptidil Peptidase IV/farmacocinética , Humanos , Estrutura Molecular
14.
Endocrinology ; 150(1): 56-62, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18801896

RESUMO

Pharmacological approaches that enhance incretin action for the treatment of type 2 diabetes mellitus have recently been developed, i.e. injectable glucagon-like peptide-1 receptor (GLP-1R) agonists with prolonged plasma half-lives and orally available inhibitors of dipeptidyl peptidase (DPP)-4, the main enzyme responsible for the rapid degradation of circulating glucagon-like peptide-1 and glucose-dependent insulinotropic peptide. The mechanism(s) underlying the glucose-lowering effect of these two pharmacotherapies differs and is not yet fully understood. Here we investigated whether acute GLP-1R activation (exendin-4) or DPP-4 inhibition (des-F-sitagliptin) modulates insulin action in mice using a hyperinsulinemic euglycemic clamp. A single iv bolus of des-F-sitagliptin (11 mg/kg) was administered to mice 15 min after the start of the clamp, and its effect was compared with a 50-ng bolus of exendin-4 or the same volume of saline. Despite matched levels of plasma glucose and insulin, within 15 min the glucose infusion rate required to maintain euglycemia was significantly greater after des-F-sitagliptin compared with saline or exendin-4. This difference was entirely due to enhancement of insulin-mediated suppression of endogenous glucose production by des-F-sitagliptin, with no difference in glucose disposal rate. These findings illustrate that DPP-4 inhibition modulates glucose homeostasis through pathways distinct from those used by GLP-1R agonists in mice.


Assuntos
Glicemia/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Insulina/farmacologia , Animais , Glicemia/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Exenatida , Técnica Clamp de Glucose , Teste de Tolerância a Glucose , Hiperinsulinismo/sangue , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Secreção de Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/farmacologia , Pirazinas/farmacologia , Fosfato de Sitagliptina , Triazóis/farmacologia , Peçonhas/farmacologia
15.
Gastroenterology ; 135(2): 689-98, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18565334

RESUMO

BACKGROUND & AIMS: Conversion into bile acids represents an important route to remove excess cholesterol from the body. Rev-erbalpha is a nuclear receptor that participates as one of the clock genes in the control of circadian rhythmicity and plays a regulatory role in lipid metabolism and adipogenesis. Here, we investigate a potential role for Rev-erbalpha in the control of bile acid metabolism via the regulation of the neutral bile acid synthesis pathway. METHODS: Bile acid synthesis and CYP7A1 gene expression were studied in vitro and in vivo in mice deficient for or over expressing Rev-erbalpha. RESULTS: Rev-erbalpha-deficient mice display a lower synthesis rate and an impaired excretion of bile acids into the bile and feces. Expression of CYP7A1, the rate-limiting enzyme of the neutral pathway, is decreased in livers of Rev-erbalpha-deficient mice, whereas adenovirus-mediated hepatic Rev-erbalpha overexpression induces its expression. Moreover, bile acid feeding resulted in a more pronounced suppression of hepatic CYP7A1 expression in Rev-erbalpha-deficient mice. Hepatic expression of E4BP4 and the orphan nuclear receptor small heterodimer partner (SHP), both negative regulators of CYP7A1 expression, is increased in Rev-erbalpha-deficient mice. Promoter analysis and chromatin immunoprecipitation experiments demonstrated that SHP and E4BP4 are direct Rev-erbalpha target genes. Finally, the circadian rhythms of liver CYP7A1, SHP, and E4BP4 messenger RNA levels were perturbed in Rev-erbalpha-deficient mice. CONCLUSIONS: These data identify a role for Rev-erbalpha in the regulatory loop of bile acid synthesis, likely acting by regulating both hepatic SHP and E4BP4 expression.


Assuntos
Ácidos e Sais Biliares/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fígado/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Bile/metabolismo , Linhagem Celular Tumoral , Colesterol 7-alfa-Hidroxilase/genética , Colesterol 7-alfa-Hidroxilase/metabolismo , Ritmo Circadiano , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Fezes/química , Regulação Enzimológica da Expressão Gênica , Humanos , Fígado/enzimologia , Camundongos , Camundongos Knockout , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Tempo , Transfecção
16.
Mol Endocrinol ; 22(8): 1797-811, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18511497

RESUMO

A role of the nuclear receptor Rev-erbalpha in the regulation of transcription pathways involving other nuclear receptors is emerging. Indeed, Rev-erbalpha is a negative regulator of transcription by binding to overlapping response elements shared with various nuclear receptors, including the peroxisome proliferator-activated receptors and the retinoid-related orphan receptor alpha (RORalpha). Here, we show that Rev-erbalpha is expressed in primary human macrophages and that its expression is induced by synthetic ligands for the liver X receptors (LXRs), which control cholesterol homeostasis, inflammation, and the immune response in macrophages. LXRalpha binds to a specific response element in the human Rev-erbalpha promoter, thus inducing Rev-erbalpha transcriptional expression. Interestingly, Rev-erbalpha does not influence basal or LXR-regulated cholesterol homeostasis. However, Rev-erbalpha overexpression represses the induction of toll-like receptor (TLR)-4 by LXR agonists, whereas Rev-erbalpha silencing by short interfering RNA results in enhanced TLR-4 expression upon LXR activation. Electrophoretic mobility shift, chromatin immunoprecipitation, and transient transfection experiments demonstrate that Rev-erbalpha represses human TLR-4 promoter activity by binding as a monomer to a RevRE site overlapping with the LXR response element site in the TLR-4 promoter. These data identify Rev-erbalpha as a new LXR target gene, inhibiting LXR-induction of TLR-4 in a negative transcriptional feedback loop, but not cholesterol homeostasis gene expression.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Retroalimentação Fisiológica , Macrófagos/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Sequência de Bases , Células Cultivadas , Colesterol/metabolismo , Dimerização , Retroalimentação Fisiológica/efeitos dos fármacos , Humanos , Lipopolissacarídeos/farmacologia , Receptores X do Fígado , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Modelos Genéticos , Dados de Sequência Molecular , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares , Receptores Nucleares Órfãos , Ligação Proteica/efeitos dos fármacos , Proteínas Repressoras/metabolismo , Elementos de Resposta/genética , Receptor X Retinoide alfa/metabolismo , Receptor 4 Toll-Like/genética , Ativação Transcricional/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
17.
Mol Pharmacol ; 67(1): 241-9, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15470081

RESUMO

UDP-glucuronosyltransferase (UGT) enzymes catalyze the glucuronidation reaction, which is a major pathway in the catabolism and elimination of numerous endo- and xenobiotics. Among the UGT enzyme family members, the UGT1A7, UGT1A8, UGT1A9, and UGT1A10 isoforms are issued from a single gene through differential splicing. However, these enzymes display distinct tissue-specific expression patterns. Indeed, UGT1A7, UGT1A8, and UGT1A10 are exclusively expressed in extrahepatic tissues, whereas UGT1A9 transcripts are found at high concentrations in liver. In the present study, we report that the liver-enriched hepatocyte nuclear factor 4 (HNF4)-alpha controls the hepatic expression of the UGT1A9 enzyme. Liver-specific disruption of the HNF4alpha gene in mice drastically decreases liver UGT1A9 mRNA levels. Furthermore, an HNF4alpha response element (HNF4alpha RE) was identified in the promoter of human UGT1A9 at position -372 to -360 base pairs by transient transfection, electrophoretic mobility shift assays, and chromatin immunoprecipitation experiments. It is interesting that this response element is absent in the proximal UGT1A7, UGT1A8, and UGT1A10 gene promoters. In conclusion, the present study identifies HNF4alpha as a major factor for the control of UGT1A9 hepatic expression and suggests that the absence of UGT1A7, UGT1A8, and UGT1A10 expression in the liver is caused by, at least in part, a few base pair changes in their promoter sequences in the region corresponding to the HNF4alpha RE of the UGT1A9 gene.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Glucuronosiltransferase/genética , Fígado/enzimologia , Fosfoproteínas/fisiologia , Fatores de Transcrição/fisiologia , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Clonagem Molecular , Fator 4 Nuclear de Hepatócito , Humanos , Mutagênese Sítio-Dirigida , Plasmídeos , Regiões Promotoras Genéticas , Proteínas Recombinantes/metabolismo , Mapeamento por Restrição , Reação em Cadeia da Polimerase Via Transcriptase Reversa , UDP-Glucuronosiltransferase 1A
18.
J Biol Chem ; 279(44): 45512-8, 2004 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-15337740

RESUMO

Statins are drugs widely used in humans to treat hypercholesterolemia. Statins act by inhibiting cholesterol synthesis resulting in the activation of the transcription factor sterol-responsive element-binding protein-2 that controls the expression of genes involved in cholesterol homeostasis. Statin therapy also decreases plasma triglyceride and non-esterified fatty acid levels, but the mechanism behind this effect remains more elusive. Liver fatty acid-binding protein (L-FABP) plays a role in the influx of long-chain fatty acids into hepatocytes. Here we show that L-FABP is a target for statins. In rat hepatocytes, simvastatin treatment induced L-FABP mRNA levels in a dose-dependent manner. Moreover, L-FABP promoter activity was induced by statin treatment. Progressive 5'-deletion analysis revealed that the peroxisome proliferator-activated receptor (PPAR)-responsive element located at position -67/-55 was responsible for the statin-mediated transactivation of the rat L-FABP promoter. Moreover, treatment with simvastatin and the PPARalpha agonist Wy14,649 resulted in a synergistic induction of L-FABP expression (mRNA and protein) in rat Fao hepatoma cells. This effect was also observed in vivo in wild-type mice but not in PPARalpha-null animals demonstrating the direct implication of PPARalpha in L-FABP regulation by statin treatment. Statin treatment resulted in a rise in PPARalpha mRNA levels both in vitro and in vivo and activated the mouse PPARalpha promoter in a reporter assay. Altogether, these data demonstrate that L-FABP expression is up-regulated by statins through a mechanism involving PPARalpha. Moreover, PPARalpha might be a statin target gene. These effects might contribute to the triglyceride/non-esterified fatty acid-lowering properties of statins.


Assuntos
Proteínas de Transporte/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , PPAR alfa/fisiologia , Sinvastatina/farmacologia , Animais , Sequência de Bases , Células CACO-2 , Proteínas de Ligação a Ácido Graxo , Hepatócitos/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , PPAR alfa/genética , Regiões Promotoras Genéticas , Pirimidinas/farmacologia , RNA Mensageiro/análise , Ratos
19.
J Biol Chem ; 277(51): 49275-81, 2002 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-12377782

RESUMO

The Rev-erb and retinoic acid-related orphan receptors (ROR) are two related families of orphan nuclear receptors that recognize similar response elements but have opposite effects on transcription. Recently, the Rev-erbalpha gene promoter has been characterized and shown to harbor a functional Rev-erbalpha-binding site known as Rev-DR2, responsible for negative feedback down-regulation of promoter activity by Rev-erbalpha itself. The present study aimed to investigate whether Rev-erbalpha gene expression is regulated by RORalpha. Gel shift analysis demonstrated that in vitro translated hRORalpha1 protein binds to the Rev-DR2 site, both as monomer and dimer. Chromatin immunoprecipitation assays demonstrated that binding of RORalpha to this site also occurred in vivo in human hepatoma HepG2 cells. The Rev-DR2 site was further shown to be functional as it conferred hRORalpha1 responsiveness to a heterologous promoter and to the natural human Rev-erbalpha gene promoter in these cells. Mutation of this site in the context of the natural Rev-erbalpha gene promoter abolished its activation by RORalpha, indicating that this site plays a key role in hRORalpha1 action. Finally, adenoviral overexpression of hRORalpha1 in HepG2 cells led to enhanced hRev-erbalpha mRNA accumulation, further confirming the physiological importance of RORalpha1 in the regulation of Rev-erbalpha expression.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transativadores/metabolismo , Transcrição Gênica , Tretinoína/metabolismo , Adenoviridae/genética , Sítios de Ligação , Linhagem Celular , Cromatina/metabolismo , Dimerização , Regulação da Expressão Gênica , Humanos , Mutação , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares , Plasmídeos/metabolismo , Testes de Precipitina , Regiões Promotoras Genéticas , Ligação Proteica , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Proteínas Recombinantes/metabolismo , Fatores de Tempo
20.
J Clin Invest ; 109(7): 961-71, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11927623

RESUMO

Serum levels of HDL are inversely correlated with the risk of coronary heart disease. The anti-atherogenic effect of HDL is partially mediated by its major protein constituent apoA-I. In this study, we identify bile acids that are activators of the nuclear receptor farnesoid X receptor (FXR) as negative regulators of human apoA-I expression. Intrahepatocellular accumulation of bile acids, as seen in patients with progressive familial intrahepatic cholestasis and biliary atresia, was associated with diminished apoA-I serum levels. In human apoA-I transgenic mice, treatment with the FXR agonist taurocholic acid strongly decreased serum concentrations and liver mRNA levels of human apoA-I, which was associated with reduced serum HDL levels. Incubation of human primary hepatocytes and hepatoblastoma HepG2 cells with bile acids resulted in a dose-dependent downregulation of apoA-I expression. Promoter mutation analysis and gel-shift experiments in HepG2 cells demonstrated that bile acid-activated FXR decreases human apoA-I promoter activity by a negative FXR response element mapped to the C site. FXR bound this site and repressed transcription in a manner independent of retinoid X receptor. The nonsteroidal synthetic FXR agonist GW4064 likewise decreased apoA-I mRNA levels and promoter activity in HepG2 cells.


Assuntos
Apolipoproteína A-I/genética , Ácidos e Sais Biliares/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Regiões Promotoras Genéticas , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Apolipoproteína A-I/sangue , Sítios de Ligação , Proteínas Sanguíneas/metabolismo , Células Cultivadas , Colestase Intra-Hepática/metabolismo , Mapeamento Cromossômico , Dimerização , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Isoxazóis/farmacologia , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Elementos de Resposta , Células Tumorais Cultivadas , gama-Glutamiltransferase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA