Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res ; 73: 103254, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-38035530

RESUMO

A number of genetic variants in the SYNM gene encoding for the intermediate filament synemin have been reported in patients with cardiomyopathies, skeletal myopathies, cancer and certain neurodegenerative disorders. To better understand its role, we generated a human induced pluripotent stem cell line with a homozygous deletion in the SYNM gene by CRISPR/Cas9 genome editing. The synemin-knockout human induced pluripotent stem cells exhibit typical morphology of pluripotent cells, expression of pluripotency markers, normal karyotype and differentiation capacity in the three germ layers. This line will allow us to investigate the role of synemin in cardiomyopathy upon differentiation into beating cardiomyocytes.


Assuntos
Cardiomiopatias , Células-Tronco Pluripotentes Induzidas , Humanos , Sistemas CRISPR-Cas/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Homozigoto , Deleção de Sequência , Cardiomiopatias/genética , Cardiomiopatias/metabolismo
2.
Int J Mol Sci ; 23(13)2022 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-35806398

RESUMO

Myocardin-related transcription factors (MRTFs) play a central role in the regulation of actin expression and cytoskeletal dynamics that are controlled by Rho GTPases. SRF is a ubiquitous transcription factor strongly expressed in muscular tissues. The depletion of SRF in the adult mouse heart leads to severe dilated cardiomyopathy associated with the down-regulation of target genes encoding sarcomeric proteins including α-cardiac actin. The regulatory triad, composed of SRF, its cofactor MRTFA and actin, plays a major role in the coordination of the nuclear transcriptional response to adapt actin filament dynamics associated with changes in cell shape, and contractile and migratory activities. Most of the knowledge on the regulation of the SRF-MRTF-Actin axis has been obtained in non-muscle cells with α-actin and smooth muscle cells with α-smooth actin. Here, we visualized for the first time by a time-lapse video, the nucleocytoplasmic shuttling of MRTFA induced by serum or pro-hypertrophic agonists such as angiotensin II, phenylephrine and endothelin-1, using an MRTFA-GFP adenovirus in cultures of neonatal rat cardiomyocytes. We showed that an inhibitor of the RhoA/ROCK signaling pathway leads to an α-cardiac actin polymerization disruption and inhibition of MRTFA nucleocytoplasmic shuttling. Moreover, inhibition of the PI3K/Akt signaling pathway also prevents the entry of MRTFA into the nuclei. Our findings point out a central role of the SRF-MRTFA-actin axis in cardiac remodeling.


Assuntos
Actinas , Fatores de Transcrição , Actinas/metabolismo , Animais , Camundongos , Miócitos Cardíacos/metabolismo , Proteínas Nucleares , Fosfatidilinositol 3-Quinases , Ratos , Fator de Resposta Sérica/genética , Transativadores , Fatores de Transcrição/metabolismo
3.
FASEB J ; 34(2): 2987-3005, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31908029

RESUMO

The expression of α-cardiac actin, a major constituent of the cytoskeleton of cardiomyocytes, is dramatically decreased in a mouse model of dilated cardiomyopathy triggered by inducible cardiac-specific serum response factor (Srf) gene disruption that could mimic some forms of human dilated cardiomyopathy. To investigate the consequences of the maintenance of α-cardiac actin expression in this model, we developed a new transgenic mouse based on Cre/LoxP strategy, allowing together the induction of SRF loss and a compensatory expression of α-cardiac actin. Here, we report that maintenance of α-cardiac actin within cardiomyocytes temporally preserved cytoarchitecture from adverse cardiac remodeling through a positive impact on both structural and transcriptional levels. These protective effects were accompanied in vivo by the decrease of ROS generation and protein carbonylation and the downregulation of NADPH oxidases NOX2 and NOX4. We also show that ectopic expression of α-cardiac actin protects HEK293 cells against oxidative stress induced by H2 O2 . Oxidative stress plays an important role in the development of cardiac remodeling and contributes also to the pathogenesis of heart failure. Taken together, these findings indicate that α-cardiac actin could be involved in the regulation of oxidative stress that is a leading cause of adverse remodeling during dilated cardiomyopathy development.


Assuntos
Actinas/metabolismo , Cardiomiopatia Dilatada/metabolismo , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Actinas/genética , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/prevenção & controle , Modelos Animais de Doenças , Feminino , Humanos , Peróxido de Hidrogênio/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/patologia , NADPH Oxidase 2/genética , NADPH Oxidase 2/metabolismo , NADPH Oxidase 4/genética , NADPH Oxidase 4/metabolismo
4.
J Cell Biol ; 217(2): 685-700, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29269426

RESUMO

Satellite cells (SCs) are adult muscle stem cells that are mobilized when muscle homeostasis is perturbed. Here, we show that serum response factor (Srf) is needed for optimal SC-mediated hypertrophic growth. We identified Srf as a master regulator of SC fusion required in both fusion partners, whereas it was dispensable for SC proliferation and differentiation. We show that SC-specific Srf deletion leads to impaired actin cytoskeleton and report the existence of finger-like actin-based protrusions at fusion sites in vertebrates that were notoriously absent in fusion-defective myoblasts lacking Srf. Restoration of a polymerized actin network by overexpression of an α-actin isoform in Srf mutant SCs rescued their fusion with a control cell in vitro and in vivo and reestablished overload-induced muscle growth. These findings demonstrate the importance of Srf in controlling the organization of actin cytoskeleton and actin-based protrusions for myoblast fusion in mammals and its requirement to achieve efficient hypertrophic myofiber growth.


Assuntos
Actinas/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Fator de Resposta Sérica/metabolismo , Animais , Fusão Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Camundongos , Camundongos Transgênicos , Células Satélites de Músculo Esquelético/citologia
5.
Circulation ; 137(21): 2256-2273, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29217642

RESUMO

BACKGROUND: Myocardial metabolic impairment is a major feature in chronic heart failure. As the major coenzyme in fuel oxidation and oxidative phosphorylation and a substrate for enzymes signaling energy stress and oxidative stress response, nicotinamide adenine dinucleotide (NAD+) is emerging as a metabolic target in a number of diseases including heart failure. Little is known on the mechanisms regulating homeostasis of NAD+ in the failing heart. METHODS: To explore possible alterations of NAD+ homeostasis in the failing heart, we quantified the expression of NAD+ biosynthetic enzymes in the human failing heart and in the heart of a mouse model of dilated cardiomyopathy (DCM) triggered by Serum Response Factor transcription factor depletion in the heart (SRFHKO) or of cardiac hypertrophy triggered by transverse aorta constriction. We studied the impact of NAD+ precursor supplementation on cardiac function in both mouse models. RESULTS: We observed a 30% loss in levels of NAD+ in the murine failing heart of both DCM and transverse aorta constriction mice that was accompanied by a decrease in expression of the nicotinamide phosphoribosyltransferase enzyme that recycles the nicotinamide precursor, whereas the nicotinamide riboside kinase 2 (NMRK2) that phosphorylates the nicotinamide riboside precursor is increased, to a higher level in the DCM (40-fold) than in transverse aorta constriction (4-fold). This shift was also observed in human failing heart biopsies in comparison with nonfailing controls. We show that the Nmrk2 gene is an AMP-activated protein kinase and peroxisome proliferator-activated receptor α responsive gene that is activated by energy stress and NAD+ depletion in isolated rat cardiomyocytes. Nicotinamide riboside efficiently rescues NAD+ synthesis in response to FK866-mediated inhibition of nicotinamide phosphoribosyltransferase and stimulates glycolysis in cardiomyocytes. Accordingly, we show that nicotinamide riboside supplementation in food attenuates the development of heart failure in mice, more robustly in DCM, and partially after transverse aorta constriction, by stabilizing myocardial NAD+ levels in the failing heart. Nicotinamide riboside treatment also robustly increases the myocardial levels of 3 metabolites, nicotinic acid adenine dinucleotide, methylnicotinamide, and N1-methyl-4-pyridone-5-carboxamide, that can be used as validation biomarkers for the treatment. CONCLUSIONS: The data show that nicotinamide riboside, the most energy-efficient among NAD precursors, could be useful for treatment of heart failure, notably in the context of DCM, a disease with few therapeutic options.


Assuntos
Cardiomiopatia Dilatada/tratamento farmacológico , Niacinamida/análogos & derivados , Proteínas Quinases Ativadas por AMP/metabolismo , Acrilamidas/uso terapêutico , Animais , Ácido Cítrico/metabolismo , Citocinas/genética , Citocinas/metabolismo , Suplementos Nutricionais , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Insuficiência Cardíaca/prevenção & controle , Metaboloma/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , NAD/metabolismo , Niacinamida/uso terapêutico , Nicotinamida Fosforribosiltransferase/genética , Nicotinamida Fosforribosiltransferase/metabolismo , PPAR alfa/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Piperidinas/uso terapêutico , Compostos de Piridínio , Ratos , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética
6.
PLoS One ; 10(10): e0139858, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26440278

RESUMO

Myocardial fibrosis contributes to the remodeling of heart and the loss of cardiac function leading to heart failure. SRF is a transcription factor implicated in the regulation of a large variety of genes involved in cardiac structure and function. To investigate the impact of an SRF overexpression in heart, we developed a new cardiac-specific and tamoxifen-inducible SRF overexpression mouse model by the Cre/loxP strategy. Here, we report that a high level overexpression of SRF leads to severe modifications of cardiac cytoarchitecture affecting the balance between cardiomyocytes and cardiac fibroblasts and also a profound alteration of cardiac gene expression program. The drastic development of fibrosis was characterized by intense sirius red staining and associated with an increased expression of genes encoding extracellular matrix proteins such as fibronectin, procollagen type 1α1 and type 3α1 and especially connective tissue growth factor (CTGF). Furthermore miR-133a, one of the most predominant cardiac miRNAs, is strongly downregulated when SRF is overexpressed. By comparison a low level overexpression of SRF has minor impact on these different processes. Investigation with miR-133a, antimiR-133a and AdSRF-VP16 experiments in H9c2 cardiac cells demonstrated that: 1)-miR-133a acts as a repressor of SRF and CTGF expression; 2)-a simultaneous overexpression of SRF by AdSRF-VP16 and inhibition of miR-133a by a specific antimiR increase CTGF expression; 3)-miR-133a overexpression can block the upregulation of CTGF induced by AdSRF-VP16. Taken together, these findings reveal a key role of the SRF/CTGF/miR-133a axis in the regulation of cardiac fibrosis.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fibrose/metabolismo , Cardiopatias/metabolismo , MicroRNAs/metabolismo , Miocárdio/metabolismo , Fator de Resposta Sérica/metabolismo , Animais , Linhagem Celular , Fator de Crescimento do Tecido Conjuntivo/genética , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Fibrose/patologia , Regulação da Expressão Gênica , Cardiopatias/patologia , Camundongos , Camundongos Transgênicos , MicroRNAs/genética , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Ratos , Fator de Resposta Sérica/genética
7.
Circ Res ; 112(7): 1035-45, 2013 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-23426017

RESUMO

RATIONALE: Vascular smooth muscle (SM) cell phenotypic modulation plays an important role in arterial stiffening associated with aging. Serum response factor (SRF) is a major transcription factor regulating SM genes involved in maintenance of the contractile state of vascular SM cells. OBJECTIVE: We investigated whether SRF and its target genes regulate intrinsic SM tone and thereby arterial stiffness. METHODS AND RESULTS: The SRF gene was inactivated SM-specific knockout of SRF (SRF(SMKO)) specifically in vascular SM cells by injection of tamoxifen into adult transgenic mice. Fifteen days later, arterial pressure and carotid thickness were lower in SRF(SMKO) than in control mice. The carotid distensibility/pressure and elastic modulus/wall stress curves showed a greater arterial elasticity in SRF(SMKO) without modification in collagen/elastin ratio. In SRF(SMKO), vasodilation was decreased in aorta and carotid arteries, whereas a decrease in contractile response was found in mesenteric arteries. By contrast, in mice with inducible SRF overexpression, the in vitro contractile response was significantly increased in all arteries. Without endothelium, the contraction was reduced in SRF(SMKO) compared with control aortic rings owing to impairment of the NO pathway. Contractile components (SM-actin and myosin light chain), regulators of the contractile response (myosin light chain kinase, myosin phosphatase target subunit 1, and protein kinase C-potentiated myosin phosphatase inhibitor) and integrins were reduced in SRF(SMKO). CONCLUSIONS: SRF controls vasoconstriction in mesenteric arteries via vascular SM cell phenotypic modulation linked to changes in contractile protein gene expression. SRF-related decreases in vasomotor tone and cell-matrix attachment increase arterial elasticity in large arteries.


Assuntos
Músculo Liso Vascular/fisiologia , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/fisiologia , Rigidez Vascular/fisiologia , Vasoconstrição/fisiologia , Envelhecimento/fisiologia , Animais , Aorta/fisiologia , Pressão Sanguínea/fisiologia , Artérias Carótidas/fisiologia , Modelos Animais de Doenças , Elasticidade , Artérias Mesentéricas/fisiologia , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Tono Muscular/fisiologia , Músculo Liso Vascular/ultraestrutura , Cadeias Leves de Miosina/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Túnica Média/fisiologia , Vasodilatação/fisiologia
8.
Dis Model Mech ; 5(4): 481-91, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22563064

RESUMO

Cardiac fibrosis is critically involved in the adverse remodeling accompanying dilated cardiomyopathies (DCMs), which leads to cardiac dysfunction and heart failure (HF). Connective tissue growth factor (CTGF), a profibrotic cytokine, plays a key role in this deleterious process. Some beneficial effects of IGF1 on cardiomyopathy have been described, but its potential role in improving DCM is less well characterized. We investigated the consequences of expressing a cardiac-specific transgene encoding locally acting IGF1 propeptide (muscle-produced IGF1; mIGF1) on disease progression in a mouse model of DCM [cardiac-specific and inducible serum response factor (SRF) gene disruption] that mimics some forms of human DCM. Cardiac-specific mIGF1 expression substantially extended the lifespan of SRF mutant mice, markedly improved cardiac functions, and delayed both DCM and HF. These protective effects were accompanied by an overall improvement in cardiomyocyte architecture and a massive reduction of myocardial fibrosis with a concomitant amelioration of inflammation. At least some of the beneficial effects of mIGF1 transgene expression were due to mIGF1 counteracting the strong increase in CTGF expression within cardiomyocytes caused by SRF deficiency, resulting in the blockade of fibroblast proliferation and related myocardial fibrosis. These findings demonstrate that SRF plays a key role in the modulation of cardiac fibrosis through repression of cardiomyocyte CTGF expression in a paracrine fashion. They also explain how impaired SRF function observed in human HF promotes fibrosis and adverse cardiac remodeling. Locally acting mIGF1 efficiently protects the myocardium from these adverse processes, and might thus represent a therapeutic avenue to counter DCM.


Assuntos
Cardiomiopatia Dilatada/fisiopatologia , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Coração/fisiopatologia , Fator de Crescimento Insulin-Like I/metabolismo , Miocárdio/patologia , Peptídeos/metabolismo , Fator de Resposta Sérica/metabolismo , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Proliferação de Células , Fibrose , Regulação da Expressão Gênica , Testes de Função Cardíaca , Humanos , Inflamação/patologia , Longevidade , Camundongos , Camundongos Mutantes , Miocárdio/ultraestrutura , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Especificidade de Órgãos
9.
FEBS Lett ; 580(2): 410-4, 2006 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-16380121

RESUMO

Liver key genes for carbohydrate and lipid homeostasis are regulated by insulin and glucose. The sterol regulatory-element binding protein-1c (SREBP-1c) has emerged as a mediator of insulin effects on gene transcription, particularly on glucokinase (GK). In this paper, we show that despite stimulation of GK promoter transcription by overexpression of mature SREBP-1c, insulin induced GK transcription at least 4h ahead of accumulation of mature SREBP-1c in the nucleus. Importantly, the knockdown of SREBP-1 mRNA using a RNA-interference technique reduced the level of nuclear SREBP-1 protein, diminished fatty acid synthase mRNA level, but did not affect GK and L-pyruvate kinase mRNA levels. We concluded that SREBP-1 is unlikely to be the mediator of the early insulin effect on GK gene transcription.


Assuntos
Regulação Enzimológica da Expressão Gênica , Glucoquinase/metabolismo , Hepatócitos/metabolismo , Insulina/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Células Cultivadas , Inativação Gênica , Glucoquinase/genética , Hepatócitos/citologia , Masculino , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Transcrição Gênica
10.
Biochem J ; 385(Pt 1): 165-71, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15344907

RESUMO

We used mRNA differential display to identify new genes induced early after exposure to insulin. Our screening strategy was based on the comparison of gene expression during the time course of insulin induction in the liver of 12-day-old suckling rats both in vivo and in vitro. A novel, early induced transcript, EIIH, was identified that encodes a 353-amino-acid protein with several features suggesting that it may be secreted or bound to membranes. EIIH is also distantly related to a variety of LRR (leucine-rich repeat) proteins. Insulin treatment increased EIIH mRNA levels in the hepatocytes of suckling, fasted adult and STZ (streptozotocin)-treated diabetic rats, where insulin was required to maintain the basal level of EIIH expression. EIIH expression was induced during the suckling/weaning transition, and remained detectable thereafter. Tissue distribution analysis in adult rats revealed a pattern of expression mainly in the liver, intestine and islets of Langerhans, closely following that of the Glut2 (glucose transporter 2), suggesting that it may play a role in carbohydrate metabolism. EIIH may be a primary target of the transcriptional regulation by insulin, and may therefore constitute a new model to study the mechanisms by which insulin acts on gene transcription.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Insulina/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Proteínas/genética , Envelhecimento/genética , Animais , Animais Lactentes , Células Cultivadas , Diabetes Mellitus Experimental/genética , Estradiol/farmacologia , Jejum , Feminino , Glucoquinase/genética , Glucose/farmacologia , Transportador de Glucose Tipo 2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/citologia , Fígado/embriologia , Masculino , Dados de Sequência Molecular , Proteínas de Transporte de Monossacarídeos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos
11.
J Histochem Cytochem ; 51(3): 319-30, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12588960

RESUMO

We applied automatic quantitative fluorescence imaging of nuclear DNA to rat liver cells obtained from animals at various times after birth up to 3 months of age. We show that, in conditions best preserving the native cellular structures, DNA content measurements, performed on whole single cells in situ after Hoechst staining, were precise and accurate. Cells in the various ploidy and nuclearity classes could thus be identified correctly and their percentages were estimated on a total of 300 cells or more. DNA synthesis was shown to occur asynchronously in all ploidy and nuclearity classes around weaning time. Observation of the labeling patterns, after in vivo BrdU pulse and short-term culture (chase), showed that the cell cycle was shorter in diploid cells compared with cells undergoing polyploidization. These results show that the approach of fluorescence imaging is well suited to investigations on polyploidization mechanisms.


Assuntos
DNA/genética , Hepatócitos/ultraestrutura , Fatores Etários , Animais , Benzimidazóis , Bromodesoxiuridina , DNA/biossíntese , Corantes Fluorescentes , Masculino , Microscopia de Fluorescência , Poliploidia , Ratos , Ratos Wistar
12.
J Biol Chem ; 277(41): 37991-8000, 2002 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-12107181

RESUMO

Liver carnitine palmitoyltransferase I catalyzes the transfer of long-chain fatty acids into mitochondria. L-CPT I is considered the rate-controlling enzyme in fatty acid oxidation. Expression of the L-CPT I gene is induced by starvation in response to glucagon secretion from the pancreas, an effect mediated by cAMP. Here, the molecular mechanisms underlying the induction of L-CPT I gene expression by cAMP were characterized. We demonstrate that the cAMP response unit of the L-CPT I gene is composed of a cAMP-response element motif and a DR1 sequence located 3 kb upstream of the transcription start site. Our data strongly suggest that the coactivator PGC-1 is involved in the regulation of this gene expression by cAMP in combination with HNF4 alpha and cAMP-response element-binding protein (CREB). Indeed, (i) cotransfection of CREB or HNF4 alpha dominant negative mutants completely abolishes the effect of cAMP on the L-CPT I promoter, and (ii) the cAMP-responsive unit binds HNF4 alpha and CREB through the DR1 and the cAMP-response element sequences, respectively. Moreover, cotransfection of PGC-1 strongly activates the L-CPT I promoter through HNF4 alpha bound at the DR1 element. Finally, we show that the transcriptional induction of the PGC-1 gene by glucagon through cAMP in hepatocytes precedes that of L-CPT-1. In addition to the key role that PGC-1 plays in glucose homeostasis, it may also be critical for lipid homeostasis. Taken together these observations suggest that PGC-1 acts to coordinate the process of metabolic adaptation in the liver.


Assuntos
Carnitina O-Palmitoiltransferase/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteínas de Ligação a DNA , Regulação Enzimológica da Expressão Gênica , Fígado/enzimologia , Fosfoproteínas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Carnitina O-Palmitoiltransferase/metabolismo , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Feminino , Genes Reporter , Glucagon/farmacologia , Fator 4 Nuclear de Hepatócito , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Mutação , Regiões Promotoras Genéticas , Ratos , Ratos Wistar , Receptores Citoplasmáticos e Nucleares/metabolismo , Elementos de Resposta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...