Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 10(1): 5281, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32210317

RESUMO

Traumatic injury of peripheral nerves typically also damages nerve surrounding tissue including muscles. Hence, molecular and cellular interactions of neighboring damaged tissues might be decisive for successful axonal regeneration of injured nerves. So far, the contribution of muscles and muscle-derived molecules to peripheral nerve regeneration has only poorly been studied. Herein, we conditionally ablated SRF (serum response factor), an important myofiber transcription factor, in skeletal muscles of mice. Subsequently, the impact of this myofiber-restricted SRF deletion on peripheral nerve regeneration, i.e. facial nerve injury was analyzed. Quantification of facial nerve regeneration by retrograde tracer transport, inspection of neuromuscular junctions (NMJs) and recovery of whisker movement revealed reduced axonal regeneration upon muscle specific Srf deletion. In contrast, responses in brainstem facial motor neuron cell bodies such as regeneration-associated gene (RAG) induction of Atf3, synaptic stripping and neuroinflammation were not overly affected by SRF deficiency. Mechanistically, SRF in myofibers appears to stimulate nerve regeneration through regulation of muscular satellite cell (SC) proliferation. In summary, our data suggest a role of muscle cells and SRF expression within muscles for regeneration of injured peripheral nerves.


Assuntos
Músculos Faciais/metabolismo , Traumatismos do Nervo Facial/fisiopatologia , Nervo Facial/fisiologia , Músculo Masseter/metabolismo , Regeneração Nervosa/fisiologia , Fator de Resposta Sérica/fisiologia , Fator 3 Ativador da Transcrição/biossíntese , Fator 3 Ativador da Transcrição/genética , Animais , Tronco Encefálico/fisiopatologia , Músculos Faciais/inervação , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Lábio/inervação , Músculo Masseter/inervação , Camundongos , Neurônios Motores/fisiologia , Especificidade de Órgãos , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/metabolismo , Células Satélites de Músculo Esquelético/fisiologia , Fator de Resposta Sérica/biossíntese , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética , Regulação para Cima , Vibrissas/inervação
2.
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
3.
Mol Neurobiol ; 54(10): 8242-8262, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-27914009

RESUMO

Stress experience modulates behavior, metabolism, and energy expenditure of organisms. One molecular hallmark of an acute stress response is a rapid induction of immediate early genes (IEGs) such as c-Fos and Egr family members. IEG transcription in neurons is mediated by the neuronal activity-driven gene regulator serum response factor (SRF). We show a first role of SRF in immediate and long-lasting acute restraint stress (AS) responses. For this, we employed a standardized mouse phenotyping protocol at the German Mouse Clinic (GMC) including behavioral, metabolic, and cardiologic tests as well as gene expression profiling to analyze the consequences of forebrain-specific SRF deletion in mice exposed to AS. Adult mice with an SRF deletion in glutamatergic neurons (Srf; CaMKIIa-CreERT2 ) showed hyperactivity, decreased anxiety, and impaired working memory. In response to restraint AS, instant stress reactivity including locomotor behavior and corticosterone induction was impaired in Srf mutant mice. Interestingly, even several weeks after previous AS exposure, SRF-deficient mice showed long-lasting AS-associated changes including altered locomotion, metabolism, energy expenditure, and cardiovascular changes. This suggests a requirement of SRF for mediating long-term stress coping mechanisms in wild-type mice. SRF ablation decreased AS-mediated IEG induction and activity of the actin severing protein cofilin. In summary, our data suggest an SRF function in immediate AS reactions and long-term post-stress-associated coping mechanisms.


Assuntos
Adaptação Psicológica/fisiologia , Genes Precoces/fisiologia , Prosencéfalo/metabolismo , Fator de Resposta Sérica/deficiência , Estresse Psicológico/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Prosencéfalo/patologia , Fator de Resposta Sérica/genética , Estresse Psicológico/genética , Estresse Psicológico/patologia , Fatores de Tempo
4.
Mol Neurobiol ; 53(3): 1478-1493, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25636686

RESUMO

Although the transcription factor serum response factor (SRF) has been suggested to play a role in activity-dependent gene expression and mediate plasticity-associated structural changes in the hippocampus, no unequivocal evidence has been provided for its role in brain pathology, such as epilepsy. A genome-wide program of activity-induced genes that are regulated by SRF also remains unknown. In the present study, we show that the inducible and conditional deletion of SRF in the adult mouse hippocampus increases the epileptic phenotype in the kainic acid model of epilepsy, reflected by more severe and frequent seizures. Moreover, we observe a robust decrease in activity-induced gene transcription in SRF knockout mice. We characterize the genetic program controlled by SRF in neurons and using functional annotation, we find that SRF target genes are associated with synaptic plasticity and epilepsy. Several of these SRF targets function as regulators of inhibitory or excitatory balance and the structural plasticity of neurons. Interestingly, mutations in those SRF targets have found to be associated with such human neuropsychiatric disorders, as autism and intellectual disability. We also identify novel direct SRF targets in hippocampus: Npas4, Gadd45g, and Zfp36. Altogether, our data indicate that proteins that are highly upregulated by neuronal stimulation, identified in the present study as SRF targets, may function as endogenous protectors against overactivation. Thus, the lack of these effector proteins in SRF knockout animals may lead to uncontrolled excitation and eventually epilepsy.


Assuntos
Epilepsia/genética , Fator de Resposta Sérica/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Epilepsia/induzido quimicamente , Epilepsia/metabolismo , Deleção de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Ontologia Genética , Hipocampo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/genética , Ácido Caínico/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Plasticidade Neuronal , Neurônios/metabolismo , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética , Tristetraprolina/biossíntese , Tristetraprolina/genética , Proteínas GADD45
5.
PLoS One ; 10(8): e0133751, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26241044

RESUMO

Genome-scale expression data on the absolute numbers of gene isoforms offers essential clues in cellular functions and biological processes. Smooth muscle cells (SMCs) perform a unique contractile function through expression of specific genes controlled by serum response factor (SRF), a transcription factor that binds to DNA sites known as the CArG boxes. To identify SRF-regulated genes specifically expressed in SMCs, we isolated SMC populations from mouse small intestine and colon, obtained their transcriptomes, and constructed an interactive SMC genome and CArGome browser. To our knowledge, this is the first online resource that provides a comprehensive library of all genetic transcripts expressed in primary SMCs. The browser also serves as the first genome-wide map of SRF binding sites. The browser analysis revealed novel SMC-specific transcriptional variants and SRF target genes, which provided new and unique insights into the cellular and biological functions of the cells in gastrointestinal (GI) physiology. The SRF target genes in SMCs, which were discovered in silico, were confirmed by proteomic analysis of SMC-specific Srf knockout mice. Our genome browser offers a new perspective into the alternative expression of genes in the context of SRF binding sites in SMCs and provides a valuable reference for future functional studies.


Assuntos
Proteínas Musculares/genética , Miócitos de Músculo Liso/metabolismo , RNA Mensageiro/genética , Elemento de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Navegador , Animais , Sítios de Ligação , Proteínas de Transporte/genética , Colo/citologia , Simulação por Computador , Biblioteca Gênica , Genes Reporter , Proteínas de Fluorescência Verde , Código das Histonas , Histonas/metabolismo , Canais Iônicos/genética , Jejuno/citologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Especificidade de Órgãos , Proteômica , Fator de Resposta Sérica/deficiência , Transcriptoma
6.
J Clin Invest ; 125(7): 2877-90, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26098211

RESUMO

Systemic lupus erythematosus (SLE) is a severe autoimmune disease that is associated with increased circulating apoptotic cell autoantigens (AC-Ags) as well as increased type I IFN signaling. Here, we describe a pathogenic mechanism in which follicular translocation of marginal zone (MZ) B cells in the spleens of BXD2 lupus mice disrupts marginal zone macrophages (MZMs), which normally clear AC debris and prevent follicular entry of AC-Ags. Phagocytosis of ACs by splenic MZMs required the megakaryoblastic leukemia 1 (MKL1) transcriptional coactivator-mediated mechanosensing pathway, which was maintained by MZ B cells through expression of membrane lymphotoxin-α1ß2 (mLT). Specifically, type I IFN-induced follicular shuttling of mLT-expressing MZ B cells disengaged interactions between these MZ B cells and LTß receptor-expressing MZMs, thereby downregulating MKL1 in MZMs. Loss of MKL1 expression in MZMs led to defective F-actin polymerization, inability to clear ACs, and, eventually, MZM dissipation. Aggregation of plasmacytoid DCs in the splenic perifollicular region, follicular translocation of MZ B cells, and loss of MKL1 and MZMs were also observed in an additional murine lupus model and in the spleens of patients with SLE. Collectively, the results suggest that lupus might be interrupted by strategies that maintain or enhance mechanosensing signaling in the MZM barrier to prevent follicular entry of AC-Ags.


Assuntos
Apoptose/imunologia , Interferon Tipo I/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Mecanotransdução Celular/imunologia , Animais , Autoanticorpos/biossíntese , Linfócitos B/imunologia , Linfócitos B/patologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Modelos Animais de Doenças , Feminino , Humanos , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Macrófagos/imunologia , Macrófagos/patologia , Mecanotransdução Celular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores Imunológicos/metabolismo , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética , Baço/imunologia , Baço/patologia , Transativadores/deficiência , Transativadores/genética
7.
Mech Dev ; 133: 23-35, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25020278

RESUMO

Mesoderm formation in the mouse embryo initiates around E6.5 at the primitive streak and continues until the end of axis extension at E12.5. It requires the process of epithelial-to-mesenchymal transition (EMT), wherein cells detach from the epithelium, adopt mesenchymal cell morphology, and gain competence to migrate. It was shown previously that, prior to mesoderm formation, the transcription factor SRF (Serum Response Factor) is essential for the formation of the primitive streak. To elucidate the role of murine Srf in mesoderm formation during axis extension we conditionally inactivated Srf in nascent mesoderm using the T(s)::Cre driver mouse. Defects in mutant embryos became apparent at E8.75 in the heart and in the allantois. From E9.0 onwards body axis elongation was arrested. Using genome-wide expression analysis, combined with SRF occupancy data from ChIP-seq analysis, we identified a set of direct SRF target genes acting in posterior nascent mesoderm which are enriched for transcripts associated with migratory function. We further show that cell migration is impaired in Srf mutant embryos. Thus, the primary role for SRF in the nascent mesoderm during elongation of the embryonic body axis is the activation of a migratory program, which is a prerequisite for axis extension.


Assuntos
Mesoderma/embriologia , Mesoderma/metabolismo , Fator de Resposta Sérica/metabolismo , Animais , Padronização Corporal/genética , Padronização Corporal/fisiologia , Caderinas/metabolismo , Movimento Celular/genética , Movimento Celular/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Proteínas Fetais/deficiência , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Adesões Focais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética , Fibras de Estresse/metabolismo , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Vimentina/metabolismo
8.
Blood ; 123(19): 3027-36, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24574460

RESUMO

Serum response factor (SRF) is a ubiquitously expressed transcription factor and master regulator of the actin cytoskeleton. We have previously shown that SRF is essential for megakaryocyte maturation and platelet formation and function. Here we elucidate the role of SRF in neutrophils, the primary defense against infections. To study the effect of SRF loss in neutrophils, we crossed Srf(fl/fl) mice with select Cre-expressing mice and studied neutrophil function in vitro and in vivo. Despite normal neutrophil numbers, neutrophil function is severely impaired in Srf knockout (KO) neutrophils. Srf KO neutrophils fail to polymerize globular actin to filamentous actin in response to N-formyl-methionine-leucine-phenylalanine, resulting in significantly disrupted cytoskeletal remodeling. Srf KO neutrophils fail to migrate to sites of inflammation in vivo and along chemokine gradients in vitro. Polarization in response to cytokine stimuli is absent and Srf KO neutrophils show markedly reduced adhesion. Integrins play an essential role in cellular adhesion, and although integrin expression levels are maintained with loss of SRF, integrin activation and trafficking are disrupted. Migration and cellular adhesion are essential for normal cell function, but also for malignant processes such as metastasis, underscoring an essential function for SRF and its pathway in health and disease.


Assuntos
Movimento Celular/genética , Inflamação/genética , Neutrófilos/metabolismo , Fator de Resposta Sérica/genética , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Western Blotting , Adesão Celular/genética , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Quimiocinas/metabolismo , Expressão Gênica/efeitos dos fármacos , Inflamação/fisiopatologia , Integrinas/genética , Integrinas/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , Polimerização/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/fisiologia , Transdução de Sinais/genética
9.
Cardiovasc Res ; 98(3): 372-80, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23436819

RESUMO

AIMS: The expression of the sodium/calcium exchanger NCX1 increases during cardiac hypertrophy and heart failure, playing an important role in Ca(2+) extrusion. This increase is presumed to result from stress signalling induced changes in the interplay between transcriptional and post-transcriptional regulations. We aimed to determine the impact of the SRF transcription factor known to regulate the NCX1 promoter and microRNA genes, on the expression of NCX1 mRNA and protein and annexin A5 (AnxA5), a Ca(2+)-binding protein interacting with NCX1 and increased during HF. METHODS AND RESULTS: NCX1 mRNA was decreased while the protein was increased in the failing heart of the cardiomyocyte-restricted SRF knock-out mice (SRF(HKO)). The induction of NCX1 mRNA by the pro-hypertrophic drug phenylephrine observed in control mice was abolished in the SRF(HKO) though the protein was strongly increased. AnxA5 protein expression profile paralleled the expression of NCX1 protein in the SRF(HKO). MiR-1, a microRNA regulated by SRF, was decreased in the SRF(HKO) and repressed by phenylephrine. In vitro and in vivo manipulation of miR-1 levels and site-directed mutagenesis showed that NCX1 and AnxA5 mRNAs are targets of miR-1. AnxA5 overexpression slowed down Ca(2+) extrusion during caffeine application in adult rat cardiomyocytes. CONCLUSION: Our study reveals the existence of a complex regulatory loop where SRF regulates the transcription of NCX1 and miR-1, which in turn functions as a rheostat limiting the translation of NCX1 and AnxA5 proteins. The decrease of miR-1 and increase of AnxA5 appear as important modulators of NCX1 expression and activity during heart failure.


Assuntos
Anexina A5/metabolismo , Cardiomiopatia Dilatada/metabolismo , Insuficiência Cardíaca/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Fator de Resposta Sérica/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Animais , Anexina A5/genética , Cafeína/farmacologia , Cálcio/metabolismo , Sinalização do Cálcio , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/fisiopatologia , Linhagem Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica , Genótipo , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Miócitos Cardíacos/efeitos dos fármacos , Fenótipo , Fenilefrina/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética , Trocador de Sódio e Cálcio/genética , Fatores de Tempo , Transfecção
10.
J Invest Dermatol ; 133(3): 608-617, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23151848

RESUMO

Serum response factor (SRF) is a transcription factor that regulates the expression of growth-related immediate-early, cytoskeletal, and muscle-specific genes to control growth, differentiation, and cytoskeletal integrity in different cell types. To investigate the role for SRF in epidermal development and homeostasis, we conditionally knocked out SRF in epidermal keratinocytes. We report that SRF deletion disrupted epidermal barrier function leading to early postnatal lethality. Mice lacking SRF in epidermis displayed morphogenetic defects, including an eye-open-at-birth phenotype and lack of whiskers. SRF-null skin exhibited abnormal morphology, hyperplasia, aberrant expression of differentiation markers and transcriptional regulators, anomalous actin organization, enhanced inflammation, and retarded hair follicle (HF) development. Transcriptional profiling experiments uncovered profound molecular changes in SRF-null E17.5 epidermis and revealed that many previously identified SRF target CArG box-containing genes were markedly upregulated in SRF-null epidermis, indicating that SRF may function to repress transcription of a subset of its target genes in epidermis. Remarkably, when transplanted onto nude mice, engrafted SRF-null skin lacked hair but displayed normal epidermal architecture with proper expression of differentiation markers, suggesting that although keratinocyte SRF is essential for HF development, a cross-talk between SRF-null keratinocytes and the surrounding microenvironment is likely responsible for the barrier-deficient mutant epidermal phenotype.


Assuntos
Epiderme/fisiopatologia , Folículo Piloso/crescimento & desenvolvimento , Morfogênese/fisiologia , Fator de Resposta Sérica/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Animais , Comunicação Celular/fisiologia , Proliferação de Células , Epiderme/patologia , Feminino , Folículo Piloso/fisiologia , Queratinócitos/patologia , Camundongos , Camundongos Knockout , Camundongos Nus , Modelos Animais , Fenótipo , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
11.
Arterioscler Thromb Vasc Biol ; 33(2): 339-46, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23264443

RESUMO

OBJECTIVE: In resistance arteries, diameter adjustment in response to pressure changes depends on the vascular cytoskeleton integrity. Serum response factor (SRF) is a dispensable transcription factor for cellular growth, but its role remains unknown in resistance arteries. We hypothesized that SRF is required for appropriate microvascular contraction. METHODS AND RESULTS: We used mice in which SRF was specifically deleted in smooth muscle or endothelial cells, and their control. Myogenic tone and pharmacological contraction was determined in resistance arteries. mRNA and protein expression were assessed by quantitative real-time PCR (qRT-PCR) and Western blot. Actin polymerization was determined by confocal microscopy. Stress-activated channel activity was measured by patch clamp. Myogenic tone developing in response to pressure was dramatically decreased by SRF deletion (5.9±2.3%) compared with control (16.3±3.2%). This defect was accompanied by decreases in actin polymerization, filamin A, myosin light chain kinase and myosin light chain expression level, and stress-activated channel activity and sensitivity in response to pressure. Contractions induced by phenylephrine or U46619 were not modified, despite a higher sensitivity to p38 blockade; this highlights a compensatory pathway, allowing normal receptor-dependent contraction. CONCLUSIONS: This study shows for the first time that SRF has a major part to play in the control of local blood flow via its central role in pressure-induced myogenic tone in resistance arteries.


Assuntos
Pressão Arterial , Músculo Liso Vascular/metabolismo , Fator de Resposta Sérica/metabolismo , Cauda/irrigação sanguínea , Resistência Vascular , Vasodilatação , Actinas/metabolismo , Animais , Pressão Arterial/efeitos dos fármacos , Artérias/metabolismo , Western Blotting , Sinalização do Cálcio , Proteínas Contráteis/metabolismo , Relação Dose-Resposta a Droga , Filaminas , Regulação da Expressão Gênica , Masculino , Mecanotransdução Celular , Potenciais da Membrana , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Microscopia Confocal , Músculo Liso Vascular/efeitos dos fármacos , Miografia , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Técnicas de Patch-Clamp , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética , Fatores de Tempo , Resistência Vascular/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
J Neurosci ; 32(22): 7577-84, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22649236

RESUMO

The molecular mechanism underlying induction by cocaine of ΔFosB, a transcription factor important for addiction, remains unknown. Here, we demonstrate a necessary role for two transcription factors, cAMP response element binding protein (CREB) and serum response factor (SRF), in mediating this induction within the mouse nucleus accumbens (NAc), a key brain reward region. CREB and SRF are both activated in NAc by cocaine and bind to the fosB gene promoter. Using viral-mediated Cre recombinase expression in the NAc of single- or double-floxed mice, we show that deletion of both transcription factors from this brain region completely blocks cocaine induction of ΔFosB in NAc, whereas deletion of either factor alone has no effect. Furthermore, deletion of both SRF and CREB from NAc renders animals less sensitive to the rewarding effects of moderate doses of cocaine when tested in the conditioned place preference (CPP) procedure and also blocks locomotor sensitization to higher doses of cocaine. Deletion of CREB alone has the opposite effect and enhances both cocaine CPP and locomotor sensitization. In contrast to ΔFosB induction by cocaine, ΔFosB induction in NAc by chronic social stress, which we have shown previously requires activation of SRF, is unaffected by the deletion of CREB alone. These surprising findings demonstrate the involvement of distinct transcriptional mechanisms in mediating ΔFosB induction within this same brain region by cocaine versus stress. Our results also establish a complex mode of regulation of ΔFosB induction in response to cocaine, which requires the concerted activities of both SRF and CREB.


Assuntos
Proteína de Ligação a CREB/metabolismo , Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fator de Resposta Sérica/metabolismo , Análise de Variância , Animais , Proteína de Ligação a CREB/deficiência , Imunoprecipitação da Cromatina , Condicionamento Operante/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Núcleo Accumbens/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/metabolismo , Fator de Resposta Sérica/deficiência , Estresse Psicológico/genética , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Transdução Genética
13.
J Neuroinflammation ; 9: 78, 2012 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-22537405

RESUMO

BACKGROUND: The transcription factor SRF (serum response factor) mediates neuronal survival in vitro. However, data available so far suggest that SRF is largely dispensable for neuron survival during physiological brain function. FINDINGS: Here, we demonstrate that upon neuronal injury, that is facial nerve transection, constitutively-active SRF-VP16 enhances motorneuron survival. SRF-VP16 suppressed active caspase 3 abundance in vitro and enhanced neuron survival upon camptothecin induced apoptosis. Following nerve fiber injury in vitro, SRF-VP16 improved survival of neurons and re-growth of severed neurites. Further, SRF-VP16 enhanced immune responses (that is microglia and T cell activation) associated with neuronal injury in vivo. Genome-wide transcriptomics identified target genes associated with axonal injury and modulated by SRF-VP16. CONCLUSION: In sum, this is a first report describing a neuronal injury-related survival function for SRF.


Assuntos
Axônios/patologia , Traumatismos do Nervo Facial/patologia , Neurônios/patologia , Traumatismos dos Nervos Periféricos/patologia , Fator de Resposta Sérica/fisiologia , Animais , Axônios/fisiologia , Sobrevivência Celular/genética , Modelos Animais de Doenças , Traumatismos do Nervo Facial/genética , Camundongos , Camundongos Knockout , Neurônios/fisiologia , Traumatismos dos Nervos Periféricos/genética , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética
14.
J Bone Miner Res ; 27(8): 1659-68, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22434656

RESUMO

Serum response factor (SRF) plays vital roles in numerous cellular processes; however, the physiological function of SRF in skeletal tissue remains unknown. In several organ systems, SRF regulates the expression of insulin-like growth factor-1 (IGF-1), which is crucial for normal development of mineralized skeleton and bone remodeling throughout life. Here, we show that conditional deletion of SRF in osteoblasts by osteocalcin-Cre generated viable mice with normal body size and body weight. Compared with normal siblings, osteoblast-specific SRF-deficient adult mice exhibited a marked decrease in bone mineral density and bone formation rate. Deletion of SRF in primary mouse calvarial osteoblasts reduced cell differentiation and mineralization in vitro. This was accompanied by a decrease in IGF-1 expression and secretion. Addition of IGF-1 in the culture media enhanced osteoblast differentiation in control cells and partially restored the mineralization defect of SRF-deficient cells, supporting an important role of SRF in regulating IGF-1 and IGF-1-mediated osteoblast differentiation. IGF-1-induced Akt activation was inhibited in SRF-deficient calvarial cells and enhanced in the SRF overexpressed cells. In addition, SRF deficiency decreased the transcriptional activity of Runx2, the key transcription factor for osteogenesis. Overexpression of SRF induced Runx2 transactivity in control cells and restored Runx2 transactivity in the SRF-deficient cells. Taken together, we conclude that SRF is important for IGF-1-induced osteoblast differentiation and mineralization via regulating IGF-1 expression and Runx2 transactivity.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Osteogênese , Fator de Resposta Sérica/metabolismo , Transdução de Sinais , Envelhecimento/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , DNA/metabolismo , Densitometria , Feminino , Deleção de Genes , Fator de Crescimento Insulin-Like I/farmacologia , Masculino , Camundongos , Tamanho do Órgão/efeitos dos fármacos , Especificidade de Órgãos/efeitos dos fármacos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Fator de Resposta Sérica/deficiência , Transdução de Sinais/efeitos dos fármacos , Crânio/citologia , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética , Microtomografia por Raio-X
15.
Eur J Neurosci ; 35(5): 735-41, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22356487

RESUMO

The high susceptibility of dopaminergic (DA) neurons to cellular stress is regarded as a primary cause of Parkinson's disease. Here we investigate the role of the serum response factor (SRF), an important regulator of anti-apoptotic responses, for the survival of DA neurons in mice. We show that loss of SRF in DA neurons does not affect their viability and does not influence dopamine-dependent behaviors. However, ablation of SRF causes exacerbated sensitivity to 1-methyl 4-phenyl 1,2,3,6-tetrahydropyridine (MPTP), leading to significantly greater loss of DA neurons in the substantia nigra, compared with DA neurons located in the ventral tegmental area. In addition, loss of SRF decreases levels of the anti-apoptotic proteins brain-derived neurotrophic factor (BDNF) and Bcl-2, a plausible underlying cause of increased sensitivity to oxidative stress. These observations support the notion that dysfunction of the SRF-activating mitogen-associated kinase pathway may be part of Parkinson's disease etiology.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Intoxicação por MPTP/metabolismo , Intoxicação por MPTP/patologia , Estresse Oxidativo/fisiologia , Fator de Resposta Sérica/deficiência , Animais , Suscetibilidade a Doenças/metabolismo , Suscetibilidade a Doenças/patologia , Predisposição Genética para Doença , Intoxicação por MPTP/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estresse Oxidativo/genética , Transtornos Parkinsonianos/genética , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/patologia , Fator de Resposta Sérica/genética , Substância Negra/metabolismo , Substância Negra/patologia
16.
J Neurosci ; 31(46): 16651-64, 2011 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-22090492

RESUMO

Previous studies have shown that neuron-specific deletion of serum response factor (SRF) results in deficits in tangential cell migration, guidance-dependent circuit assembly, activity-dependent gene expression, and synaptic plasticity in the hippocampus. Furthermore, SRF deletion in mouse embryonic stem cells causes cell death in vitro. However, the requirement of SRF for early neuronal development including neural stem cell homeostasis, neurogenesis, and axonal innervations remains unknown. Here, we report that SRF is critical for development of major axonal tracts in the forebrain. Conditional mutant mice lacking SRF in neural progenitor cells (Srf-Nestin-cKO) exhibit striking deficits in cortical axonal projections including corticostriatal, corticospinal, and corticothalamic tracts, and they show a variable loss of the corpus callosum. Neurogenesis and interneuron specification occur normally in the absence of SRF and the deficits in axonal projections were not due to a decrease or loss in cell numbers. Radial migration of neurons and neocortical lamination were also not affected. No aberrant cell death was observed during development, whereas there was an increase in the number of proliferative cells in the ventricular zone from embryonic day 14 to day 18. Similar axonal tract deficits were also observed in mutant mice lacking SRF in the developing excitatory neurons of neocortex and hippocampus (Srf-NEX-cKO). Together, these findings suggest distinct roles for SRF during neuronal development; SRF is specifically required in a cell-autonomous manner for axonal tract development but is dispensable for cell survival, neurogenesis, neocortical lamination, and neuronal differentiation.


Assuntos
Axônios/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neocórtex/citologia , Neurogênese/fisiologia , Neurônios/citologia , Fator de Resposta Sérica/metabolismo , Fatores Etários , Aminoácidos , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Caspase 3/metabolismo , Contagem de Células/métodos , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Marcação In Situ das Extremidades Cortadas/métodos , Proteínas de Filamentos Intermediários/genética , Camundongos , Mutação/genética , Neocórtex/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina , Vias Neurais/embriologia , Vias Neurais/crescimento & desenvolvimento , Neurogênese/genética , Neurônios/classificação , Fator de Resposta Sérica/deficiência , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
17.
FASEB J ; 25(8): 2592-603, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21525490

RESUMO

Serum response factor (SRF) is an essential regulator of myogenic and neurogenic genes and the ubiquitously expressed immediate-early genes. The purpose of this study is to determine SRF expression pattern in murine pancreas and examine the role of SRF in pancreatic gene expression. Immunohistochemical analysis of wild-type pancreas and LacZ staining of pancreas from SRF LacZ knock-in animals showed that SRF expression is restricted to ß cells. SRF bound to the rat insulin promoter II (RIP II) serum response element, an element conserved in both rat I and murine I and II insulin promoters. SRF activated RIP II, and SRF binding to RIP II and the exon 5-encoded 64-aa subdomain of SRF was required for this activation. Transient or stable knockdown of SRF leads to down-regulation of insulin gene expression, suggesting that SRF is required for insulin gene expression. Further, SRF physically interacted with the pancreas and duodenum homeobox-1 (Pdx-1) and synergistically activated RIP II. Elevated glucose concentration down-regulated SRF binding to RIP II SRE, and this down-regulation was associated with decreased RIP II activity and increased SRF phosphorylation on serine 103. Together, our results demonstrate that SRF is a glucose concentration-sensitive regulator of insulin gene expression.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/genética , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Ratos , Fator de Resposta Sérica/deficiência , Transativadores/metabolismo , Transfecção
18.
Behav Brain Res ; 220(2): 312-8, 2011 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-21329726

RESUMO

Serum response factor (SRF) is a ubiquitously expressed stimulus-dependent transcription factor that regulates gene expression by binding to serum response element in the promoter region of target genes. Recent studies in mice have shown that SRF is important for activity-dependent gene expression and synaptic plasticity in the adult brain but is dispensable for neuronal survival. Given these important functions of SRF in the CNS, it is expected to play a critical role in several aspects of learning and memory. Here we evaluated the role of SRF in conditioned reinforcement using two lines of conditional SRF mutant mice. These SRF mutant mice exhibited different spatial patterns of SRF deletion in the post-natal forebrain and notably within the hippocampus. SRF deletion was more widespread in SRF-CKCre mutants than in SRF-SynCre mutants, particularly in areas of the cortex and striatum. Mutant and wild-type mice were trained to associate one auditory cue (CS+) with reward, whereas a second cue remained relatively neutral (CS-). All mice readily acquired this discrimination, entering the food cup during CS+ but not during CS-. In a subsequent test of conditioned reinforcement, in the absence of food, wild-type control mice and SRF-SynCre mice learned to selectively perform an instrumental response that yielded CS+ presentation rather than another response that produced CS-. SRF-CKCre mutants failed to show this preferential responding for CS+. These results suggest a role for SRF in conditioned reinforcement, a manifestation of incentive learning that has been implicated in many aspects of adaptive and maladaptive behavior, such as substance abuse and eating disorders.


Assuntos
Condicionamento Clássico/fisiologia , Condicionamento Operante/fisiologia , Reforço Psicológico , Fator de Resposta Sérica/deficiência , Análise de Variância , Animais , Comportamento Animal , Encéfalo/citologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Privação de Alimentos , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Sinapsinas/metabolismo
19.
Mol Cell Biol ; 31(4): 861-75, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21135125

RESUMO

Cells of the monocyte/macrophage lineage play essential roles in tissue homeostasis and immune responses, but mechanisms underlying the coordinated expression of cytoskeletal genes required for specialized functions of these cells, such as directed migration and phagocytosis, remain unknown. Here, using genetic and genomic approaches, we provide evidence that serum response factor (SRF) regulates both general and cell type-restricted components of the cytoskeletal gene expression program in macrophages. Genome-wide location analysis of SRF in macrophages demonstrates enrichment of SRF binding at ubiquitously expressed target gene promoters, as expected, but also reveals that the majority of SRF binding sites associated with cell type-restricted target genes are at distal inter- and intragenic locations. Most of these distal SRF binding sites are established by the prior binding of the macrophage- and the B cell-specific transcription factor PU.1 and exhibit histone modifications characteristic of enhancers. Consistent with this, representative cytoskeletal target genes associated with these elements require both SRF and PU.1 for full expression. These findings suggest that SRF uses two distinct molecular strategies to regulate programs of cytoskeletal gene expression: a promoter-based strategy for ubiquitously expressed target genes and an enhancer-based strategy at target genes that exhibit cell type-restricted patterns of expression.


Assuntos
Citoesqueleto/genética , Citoesqueleto/metabolismo , Macrófagos/metabolismo , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Células Cultivadas , Primers do DNA/genética , Elementos Facilitadores Genéticos , Hematopoese/genética , Hematopoese/fisiologia , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Interferente Pequeno/genética , Fator de Resposta Sérica/deficiência , Transativadores/deficiência , Transativadores/genética , Transativadores/metabolismo , Ativação Transcricional
20.
Genetics ; 186(1): 147-57, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20610412

RESUMO

Cell hyperproliferation, inflammation, and angiogenesis are biological processes central to the pathogenesis of corneal disease, as well as other conditions including tumorigenesis and chronic inflammatory disorders. Due to the number of disease conditions that arise as a result of these abnormalities, identifying the molecular mechanisms underlying these processes is critical. The avascular and transparent cornea serves as a good in vivo model to study the pathogenesis of cell hyperproliferation, inflammation, and angiogenesis. Corneal disease 1 (Dstn(corn1)) mice are homozygous for a spontaneous null allele of the destrin (Dstn) gene, which is also known as actin depolymerizing factor (ADF). These mice exhibit abnormalities in the cornea including epithelial cell hyperproliferation, stromal inflammation, and neovascularization. We previously identified that the transcription factor, serum response factor (SRF) and a number of its target genes are upregulated in the cornea of these mice. In this study, we show that conditional ablation of Srf in the corneal epithelium of a diseased Dstn(corn1) cornea results in the rescue of the epithelial cell hyperproliferation, inflammation, and neovascularization phenotypes, delineating an epithelial cell-specific role for SRF in the development of all of these abnormalities. Our study also demonstrates that Dstn is genetically upstream of Srf and defines a new functional role for SRF as the master regulator of a hyperproliferative, inflammatory phenotype accompanied by neovascularization.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Epitélio Corneano/metabolismo , Epitélio Corneano/patologia , Fator de Resposta Sérica/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Destrina/genética , Destrina/metabolismo , Doxiciclina/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitélio Corneano/citologia , Epitélio Corneano/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Fenótipo , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...