Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
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.
Brain Struct Funct ; 224(8): 2691-2701, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31375980

RESUMO

Serum response factor (SRF) is a major transcription factor that regulates the expression of several plasticity-associated genes in the brain. Although the developmental expression of SRF in excitatory neurons is crucial for establishing proper hippocampal circuitry, no substantial evidence of its role in unstimulated mature neurons has been provided. The present study used time-controlled, conditional SRF knockout mice and found that the lack of SRF in adult neurons led to decreased actin levels and inactivation of the actin-severing protein cofilin 1 through its increase in phosphorylation at Ser3. The augmentation of cofilin 1 phosphorylation correlated with an alteration of dendritic spine morphology in the dentate gyrus, which was reflected by an increase in the number of spines that clustered into the long-spine category. The changes in spine morphology coincided with a lower amplitude and frequency of miniature excitatory postsynaptic currents. Moreover, SRF knockout animals were hyperactive and exhibited impairments in hippocampus-dependent behaviors, such as digging, marble burying, and nesting. Altogether, our data indicate that the adult deletion of neuronal SRF leads to alterations of spine morphology and function and hippocampus-dependent behaviors. Thus, SRF deletion in adult neurons recapitulates some aspects of morphological, electrophysiological, and behavioral changes that are observed in such psychiatric disorders as schizophrenia and autism spectrum disorders.


Assuntos
Comportamento Animal/fisiologia , Espinhas Dendríticas/fisiologia , Giro Denteado/citologia , Giro Denteado/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Fator de Resposta Sérica/fisiologia , Animais , Potenciais Pós-Sinápticos Excitadores , Feminino , Hipocampo/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasticidade Neuronal , Fator de Resposta Sérica/genética
3.
Sci Rep ; 9(1): 4631, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30874621

RESUMO

Methylmercury is an environmental pollutant that causes specific and serious damage to the central nervous system. We have previously shown that C-C motif chemokine ligand 4 (CCL4) protects cultured neural cells from methylmercury toxicity and expression of CCL4 is specifically induced in mouse brain by methylmercury. In this study, we examined the transcriptional regulatory mechanism that induces CCL4 expression by methylmercury using C17.2 mouse neural stem cells. The promoter region of the CCL4 gene was analyzed by a reporter assay, revealing that the region up to 50 bp upstream from the transcription start site was necessary for inducing expression of CCL4 by methylmercury. Nine transcription factors that might bind to this upstream region and be involved in the induction of CCL4 expression by methylmercury were selected, and the induction of CCL4 expression by methylmercury was suppressed by the knockdown of serum response factor (SRF). In addition, the nuclear level of SRF was elevated by methylmercury, and an increase in the amount bound to the CCL4 gene promoter was also observed. Furthermore, we examined the upstream signaling pathway involved in the induction of CCL4 expression by SRF, and confirmed that activation of p38 and ERK, which are part of the MAPK pathway, are involved. These results suggest that methylmercury induces the expression of CCL4 by activating SRF via the p38 and ERK signaling pathway. Our findings are important for elucidating the mechanism involved in the brain-specific induction of CCL4 expression by methylmercury.


Assuntos
Quimiocina CCL4/metabolismo , Compostos de Metilmercúrio/efeitos adversos , Fator de Resposta Sérica/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular , Células Cultivadas , Quimiocina CCL4/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases , Compostos de Metilmercúrio/metabolismo , Compostos de Metilmercúrio/toxicidade , Camundongos , NF-kappa B/metabolismo , Células-Tronco Neurais/metabolismo , Regiões Promotoras Genéticas/genética , Fator de Resposta Sérica/fisiologia , Transdução de Sinais , Fatores de Transcrição/metabolismo
4.
J Am Soc Nephrol ; 29(2): 416-422, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29114040

RESUMO

Podocytes contain an intricate actin cytoskeleton that is essential for the specialized function of this cell type in renal filtration. Serum response factor (SRF) is a master transcription factor for the actin cytoskeleton, but the in vivo expression and function of SRF in podocytes are unknown. We found that SRF protein colocalizes with podocyte markers in human and mouse kidneys. Compared with littermate controls, mice in which the Srf gene was conditionally inactivated with NPHS2-Cre exhibited early postnatal proteinuria, hypoalbuminemia, and azotemia. Histologic changes in the mutant mice included glomerular capillary dilation and mild glomerulosclerosis, with reduced expression of multiple canonical podocyte markers. We also noted tubular dilation, cell proliferation, and protein casts as well as reactive changes in mesangial cells and interstitial inflammation. Ultrastructure analysis disclosed foot process effacement with loss of slit diaphragms. To ascertain the importance of SRF cofactors in podocyte function, we disabled the myocardin-related transcription factor A and B genes. Although loss of either SRF cofactor alone had no observable effect in the kidney, deficiency of both recapitulated the Srf-null phenotype. These results establish a vital role for SRF and two SRF cofactors in the maintenance of podocyte structure and function.


Assuntos
Actinas/metabolismo , Podócitos/metabolismo , Podócitos/ultraestrutura , Fator de Resposta Sérica/fisiologia , Transativadores/genética , Fatores de Transcrição/genética , Actinina/genética , Actinas/genética , Animais , Citoesqueleto , Dilatação Patológica/genética , Feminino , Humanos , Túbulos Renais Distais/patologia , Túbulos Renais Proximais/patologia , Masculino , Camundongos , Camundongos Knockout , Podócitos/fisiologia , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Proteínas Repressoras/genética , Fator de Resposta Sérica/genética , Proteínas WT1
5.
FEBS J ; 284(11): 1644-1656, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28342289

RESUMO

Vascular smooth muscle cells (VSMCs) do not terminally differentiate; they modulate their phenotype between proliferative and differentiated states, which is a major factor contributing to vascular diseases. TGFß signalling has been implicated in inducing VSMC differentiation, although the exact mechanism remains largely unknown. Our goal was to assess the network of transcription factors involved in the induction of VSMC differentiation, and to determine the role of TAZ in promoting the quiescent VSMC phenotype. TGFß robustly induces VSMC marker genes in 10T1/2 mouse embryonic fibroblast cells and the potent transcriptional regulator TAZ has been shown to retain Smad complexes on DNA. Thus, the role of TAZ in regulation of VSMC differentiation was studied. Using primary aortic VSMCs coupled with siRNA-mediated gene silencing, our studies reveal that TAZ is required for TGFß induction of smooth muscle genes and is also required for the differentiated VSMC phenotype; synergy between TAZ and SRF, and TAZ and Myocardin (MyoC856), in regulating smooth muscle gene activation was observed. These data provide evidence of components of a novel signalling pathway that links TGFß signalling to induction of smooth muscle genes through a mechanism involving regulation of TAZ and SRF proteins. In addition, we report a physical interaction of TAZ and MyoC856. These observations elucidate a novel level of control of VSMC induction which may have implications for vascular diseases and congenital vascular malformations.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Desenvolvimento Muscular/fisiologia , Miócitos de Músculo Liso/citologia , Proteínas Nucleares/fisiologia , Fator de Resposta Sérica/fisiologia , Transativadores/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Actinas/biossíntese , Actinas/genética , Animais , Aorta , Linhagem Celular , Células Cultivadas , Fibroblastos , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Camundongos Endogâmicos C3H , Desenvolvimento Muscular/genética , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Mapeamento de Interação de Proteínas , Interferência de RNA , RNA Interferente Pequeno/genética , Transdução de Sinais
6.
PLoS One ; 12(2): e0171262, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28152551

RESUMO

Serum response factor (SRF) transcriptionally regulates expression of contractile genes in smooth muscle cells (SMC). Lack or decrease of SRF is directly linked to a phenotypic change of SMC, leading to hypomotility of smooth muscle in the gastrointestinal (GI) tract. However, the molecular mechanism behind SRF-induced hypomotility in GI smooth muscle is largely unknown. We describe here how SRF plays a functional role in the regulation of the SMC contractility via myotonic dystrophy protein kinase (DMPK) and L-type calcium channel CACNA1C. GI SMC expressed Dmpk and Cacna1c genes into multiple alternative transcriptional isoforms. Deficiency of SRF in SMC of Srf knockout (KO) mice led to reduction of SRF-dependent DMPK, which down-regulated the expression of CACNA1C. Reduction of CACNA1C in KO SMC not only decreased intracellular Ca2+ spikes but also disrupted their coupling between cells resulting in decreased contractility. The role of SRF in the regulation of SMC phenotype and function provides new insight into how SMC lose their contractility leading to hypomotility in pathophysiological conditions within the GI tract.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Contração Muscular/fisiologia , Músculo Liso/fisiologia , Miotonina Proteína Quinase/fisiologia , Fator de Resposta Sérica/fisiologia , Animais , Western Blotting , Feminino , Masculino , Camundongos , Camundongos Knockout , Microscopia Confocal , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/ultraestrutura , Reação em Cadeia da Polimerase , Proteômica , Tamoxifeno/farmacologia
7.
Proc Natl Acad Sci U S A ; 113(13): 3651-6, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26976584

RESUMO

Behavioral changes in response to stressful stimuli can be controlled via adaptive epigenetic changes in neuronal gene expression. Here we indicate a role for the transcriptional corepressor Lysine-Specific Demethylase 1 (LSD1) and its dominant-negative splicing isoform neuroLSD1, in the modulation of emotional behavior. In mouse hippocampus, we show that LSD1 and neuroLSD1 can interact with transcription factor serum response factor (SRF) and set the chromatin state of SRF-targeted genes early growth response 1 (egr1) and c-fos Deletion or reduction of neuro LSD1 in mutant mice translates into decreased levels of activating histone marks at egr1 and c-fos promoters, dampening their psychosocial stress-induced transcription and resulting in low anxiety-like behavior. Administration of suberoylanilide hydroxamine to neuroLSD1(KO)mice reactivates egr1 and c-fos transcription and restores the behavioral phenotype. These findings indicate that LSD1 is a molecular transducer of stressful stimuli as well as a stress-response modifier. Indeed, LSD1 expression itself is increased acutely at both the transcriptional and splicing levels by psychosocial stress, suggesting that LSD1 is involved in the adaptive response to stress.


Assuntos
Emoções/fisiologia , Genes Precoces , Histona Desmetilases/fisiologia , Processamento Alternativo , Animais , Proteína 1 de Resposta de Crescimento Precoce/genética , Epigênese Genética , Genes fos , Histona Desmetilases/deficiência , Histona Desmetilases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Plasticidade Neuronal , Fenótipo , Fator de Resposta Sérica/fisiologia , Estresse Psicológico , Transcrição Gênica
8.
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
9.
Cell Death Dis ; 6: e1944, 2015 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-26512955

RESUMO

Exposure to metabolic disease during fetal development alters cellular differentiation and perturbs metabolic homeostasis, but the underlying molecular regulators of this phenomenon in muscle cells are not completely understood. To address this, we undertook a computational approach to identify cooperating partners of the myocyte enhancer factor-2 (MEF2) family of transcription factors, known regulators of muscle differentiation and metabolic function. We demonstrate that MEF2 and the serum response factor (SRF) collaboratively regulate the expression of numerous muscle-specific genes, including microRNA-133a (miR-133a). Using tandem mass spectrometry techniques, we identify a conserved phosphorylation motif within the MEF2 and SRF Mcm1 Agamous Deficiens SRF (MADS)-box that regulates miR-133a expression and mitochondrial function in response to a lipotoxic signal. Furthermore, reconstitution of MEF2 function by expression of a neutralizing mutation in this identified phosphorylation motif restores miR-133a expression and mitochondrial membrane potential during lipotoxicity. Mechanistically, we demonstrate that miR-133a regulates mitochondrial function through translational inhibition of a mitophagy and cell death modulating protein, called Nix. Finally, we show that rodents exposed to gestational diabetes during fetal development display muscle diacylglycerol accumulation, concurrent with insulin resistance, reduced miR-133a, and elevated Nix expression, as young adult rats. Given the diverse roles of miR-133a and Nix in regulating mitochondrial function, and proliferation in certain cancers, dysregulation of this genetic pathway may have broad implications involving insulin resistance, cardiovascular disease, and cancer biology.


Assuntos
Diferenciação Celular/genética , Fatores de Transcrição MEF2/química , Mitocôndrias/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator de Resposta Sérica/química , Motivos de Aminoácidos , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Diabetes Gestacional , Feminino , Regulação da Expressão Gênica , Humanos , Fatores de Transcrição MEF2/metabolismo , Fatores de Transcrição MEF2/fisiologia , Potencial da Membrana Mitocondrial/genética , MicroRNAs/metabolismo , Mitocôndrias/genética , Fibras Musculares Esqueléticas/citologia , Mutagênese Sítio-Dirigida , Miócitos Cardíacos/citologia , Miócitos de Músculo Liso/citologia , Fosforilação , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Fator de Resposta Sérica/metabolismo , Fator de Resposta Sérica/fisiologia , Espectrometria de Massas em Tandem
10.
Mol Oncol ; 9(8): 1510-27, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25999144

RESUMO

Radiotherapy is a standard treatment after conservative breast cancer surgery. However, cancers relapsing within a previously irradiated area have an increased probability to metastasize. The mechanisms responsible for this aggressiveness remain unclear. Here, we used the clinically relevant 4T1 breast cancer model mimicking aggressive local relapse after radiotherapy to identify differences between tumors grown in untreated versus preirradiated mammary glands. Tumors grown within preirradiated beds were highly enriched in transcripts encoding collagens and other proteins building or modifying the extracellular matrix, such as laminin-332, tenascins, lysyl oxidases and matrix metalloproteinases. Type I collagen, known to directly contribute to tissue stiffening, and the pro-metastatic megakaryoblastic leukemia-1 (Mkl1) target gene tenascin-C were further investigated. Mammary tissue preirradiation induced Mkl1 nuclear translocation in the tumor cells in vivo, indicating activation of Mkl1 signaling. Transcript profiling of cultured 4T1 cells revealed that the majority of the Mkl1 target genes, including tenascin-C, required serum response factor (SRF) for their expression. However, application of dynamic strain or matrix stiffness to 4T1 cells converted the predominant SRF/Mkl1 action into SAP domain-dependent Mkl1 signaling independent of SRF, accompanied by a switch to SAP-dependent tumor cell migration. 4T1 tumors overexpressing intact Mkl1 became more metastatic within preirradiated beds, while tumors expressing Mkl1 lacking the SAP domain exhibited impaired growth and metastatic spread, and decreased Mkl1 target gene expression. Thus, we identified SAP-dependent Mkl1 signaling as a previously unrecognized mediator of aggressive progression of mammary tumors locally relapsing after radiotherapy, and provide a novel signaling pathway for therapeutic intervention.


Assuntos
Neoplasias da Mama/patologia , Transformação Celular Neoplásica/efeitos da radiação , Neoplasias Induzidas por Radiação/genética , Segunda Neoplasia Primária/genética , Domínios e Motivos de Interação entre Proteínas/fisiologia , Transativadores/química , Transativadores/fisiologia , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/radioterapia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Neoplasias Induzidas por Radiação/patologia , Segunda Neoplasia Primária/patologia , Domínios e Motivos de Interação entre Proteínas/genética , Fator de Resposta Sérica/fisiologia , Transdução de Sinais/genética , Transativadores/genética
11.
J Clin Invest ; 125(4): 1569-78, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25774500

RESUMO

Maintenance of skeletal muscle structure and function requires a precise stoichiometry of sarcomeric proteins for proper assembly of the contractile apparatus. Absence of components of the sarcomeric thin filaments causes nemaline myopathy, a lethal congenital muscle disorder associated with aberrant myofiber structure and contractility. Previously, we reported that deficiency of the kelch-like family member 40 (KLHL40) in mice results in nemaline myopathy and destabilization of leiomodin-3 (LMOD3). LMOD3 belongs to a family of tropomodulin-related proteins that promote actin nucleation. Here, we show that deficiency of LMOD3 in mice causes nemaline myopathy. In skeletal muscle, transcription of Lmod3 was controlled by the transcription factors SRF and MEF2. Myocardin-related transcription factors (MRTFs), which function as SRF coactivators, serve as sensors of actin polymerization and are sequestered in the cytoplasm by actin monomers. Conversely, conditions that favor actin polymerization de-repress MRTFs and activate SRF-dependent genes. We demonstrated that the actin nucleator LMOD3, together with its stabilizing partner KLHL40, enhances MRTF-SRF activity. In turn, SRF cooperated with MEF2 to sustain the expression of LMOD3 and other components of the contractile apparatus, thereby establishing a regulatory circuit to maintain skeletal muscle function. These findings provide insight into the molecular basis of the sarcomere assembly and muscle dysfunction associated with nemaline myopathy.


Assuntos
Fatores de Transcrição MEF2/fisiologia , Proteínas dos Microfilamentos/deficiência , Miopatias da Nemalina/genética , Actinas/química , Animais , Células COS , Chlorocebus aethiops , Sequência Consenso , Creatina Quinase Forma MM/genética , Insuficiência de Crescimento/genética , Insuficiência de Crescimento/patologia , Insuficiência de Crescimento/terapia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Terapia Genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/fisiologia , Contração Muscular , Proteínas Musculares/fisiologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miopatias da Nemalina/metabolismo , Especificidade de Órgãos , Polimerização , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/metabolismo , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura , Fator de Resposta Sérica/fisiologia , Transativadores/fisiologia , Transgenes
12.
Blood ; 125(8): 1244-55, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25573994

RESUMO

Chemokine signaling is important for the seeding of different sites by hematopoietic stem cells (HSCs) during development. Serum response factor (SRF) controls multiple genes governing adhesion and migration, mainly by recruiting members of the myocardin-related transcription factor (MRTF) family of G-actin-regulated cofactors. We used vav-iCre to inactivate MRTF-SRF signaling early during hematopoietic development. In both Srf- and Mrtf-deleted animals, hematopoiesis in fetal liver and spleen is intact but does not become established in fetal bone marrow. Srf-null HSC progenitor cells (HSC/Ps) fail to effectively engraft in transplantation experiments, exhibiting normal proximal signaling responses to SDF-1, but reduced adhesiveness, F-actin assembly, and reduced motility. Srf-null HSC/Ps fail to polarize in response to SDF-1 and cannot migrate through restrictive membrane pores to SDF-1 or Scf in vitro. Mrtf-null HSC/Ps were also defective in chemotactic responses to SDF-1. Srf-null HSC/Ps exhibit substantial deficits in cytoskeletal gene expression. MRTF-SRF signaling is thus critical for expression of genes required for the response to chemokine signaling during hematopoietic development.


Assuntos
Medula Óssea/embriologia , Medula Óssea/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Fator de Resposta Sérica/fisiologia , Nicho de Células-Tronco , Transativadores/fisiologia , Fatores de Transcrição/fisiologia , Animais , Medula Óssea/crescimento & desenvolvimento , Movimento Celular/genética , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/genética , Nicho de Células-Tronco/genética
13.
Hepatology ; 61(3): 979-89, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25266280

RESUMO

UNLABELLED: The ubiquitously expressed transcriptional regulator serum response factor (SRF) is controlled by both Ras/MAPK (mitogen-activated protein kinase) and Rho/actin signaling pathways, which are frequently activated in hepatocellular carcinoma (HCC). We generated SRF-VP16iHep mice, which conditionally express constitutively active SRF-VP16 in hepatocytes, thereby controlling subsets of both Ras/MAPK- and Rho/actin-stimulated target genes. All SRF-VP16iHep mice develop hyperproliferative liver nodules that progresses to lethal HCC. Some murine (m)HCCs acquire Ctnnb1 mutations equivalent to those in human (h)HCC. The resulting transcript signatures mirror those of a distinct subgroup of hHCCs, with shared activation of oncofetal genes including Igf2, correlating with CpG hypomethylation at the imprinted Igf2/H19 locus. CONCLUSION: SRF-VP16iHep mHCC reveal convergent Ras/MAPK and Rho/actin signaling as a highly oncogenic driver mechanism for hepatocarcinogenesis. This suggests simultaneous inhibition of Ras/MAPK and Rho/actin signaling as a treatment strategy in hHCC therapy.


Assuntos
Neoplasias Hepáticas Experimentais/etiologia , Fator de Resposta Sérica/fisiologia , Animais , Proliferação de Células , Ilhas de CpG , Metilação de DNA , Perfilação da Expressão Gênica , Hepatócitos/patologia , Proteína Vmw65 do Vírus do Herpes Simples/genética , Humanos , Fator de Crescimento Insulin-Like II/genética , Linfócitos/patologia , Camundongos , Mutação , beta Catenina/genética
14.
J Gastroenterol Hepatol ; 29 Suppl 4: 112-23, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25521743

RESUMO

Angiogenesis (also referred to as neovascularization-formation of new blood vessels from existing vessels) is a fundamental process essential for healing of tissue injury and ulcers because regeneration of blood microvessels is a critical requirement for oxygen and nutrient delivery to the healing site. This review article updates the current views on angiogenesis in gastric mucosa following injury and during ulcer healing, its sequential events, the underlying mechanisms, and the impairment of angiogenesis in aging gastric mucosa. We focus on the time sequence and ultrastructural features of angiogenesis, hypoxia as a trigger, role of vascular endothelial growth factor signaling (VEGF), serum response factor, Cox2 and prostaglandins, nitric oxide, and importin. Recent reports indicate that gastric mucosa of aging humans and experimental animals exhibits increased susceptibility to injury and delayed healing. Gastric mucosa of aging rats has increased susceptibility to injury by a variety of damaging agents such as ethanol, aspirin, and other non-steroidal anti-inflammatory drugs because of structural and functional abnormalities including: reduced gastric mucosal blood flow, hypoxia, reduced expression of vascular endothelial growth factor and survivin, and increased expression of early growth response protein 1 (egr-1) and phosphatase and tensin homolog (PTEN). Until recently, postnatal neovascularization was assumed to occur solely through angiogenesis sprouting of endothelial cells and formation of new blood vessels from pre-existing blood vessels. New studies in the last decade have challenged this paradigm and indicate that in some tissues, including gastric mucosa, the homing of bone marrow-derived endothelial progenitor cells to the site of injury can also contribute to neovascularization by a process termed vasculogenesis.


Assuntos
Envelhecimento/patologia , Envelhecimento/fisiologia , Mucosa Gástrica/irrigação sanguínea , Mucosa Gástrica/patologia , Mucosa Gástrica/fisiologia , Neovascularização Patológica , Regeneração/fisiologia , Úlcera Gástrica/patologia , Úlcera Gástrica/fisiopatologia , Células da Medula Óssea , Ciclo-Oxigenase 2/fisiologia , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Células Progenitoras Endoteliais/fisiologia , Humanos , Carioferinas/fisiologia , Neovascularização Patológica/genética , Óxido Nítrico/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , Prostaglandinas/fisiologia , Regeneração/genética , Fator de Resposta Sérica/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia
15.
Cell Death Differ ; 21(12): 1900-13, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25080937

RESUMO

Dysregulation of transcription factors (TFs) is associated with tumor progression, but little is known about TF expression patterns in the context of gastric cancer (GC) metastasis. Using array-based profile analysis, we found that 22 TFs showed differential activities between GC cell lines with low- and high-metastatic potential. Of this group of TFs, serum response factor (SRF) was significantly upregulated in metastatic GC cells. SRF expression was frequently elevated in a panel of metastatic GC cells and tissues, and high-level expression of SRF was significantly associated with a more aggressive phenotype and poor prognosis in patients with GC. In GC cell lines, overexpression of SRF potently promoted cell migration and invasion in vitro as well as the formation of intrahepatic and pulmonary metastases in vivo, whereas loss of SRF inhibited GC cell invasion and metastasis. We also performed a microRNA microarray to screen for transcriptional targets of SRF and found that SRF transactivates miR-199a-5p and miR-199a-3p by directly binding to their promoters. We further determined that overexpression of miR-199a-5p, but not miR-199a-3p, increased GC cell invasion and metastasis. In contrast, inhibition of miR-199a-5p impaired the metastatic potential of GC cells in vitro and in vivo, and E-cadherin was identified as a direct and functional target of miR-199a-5p in GC cells. Specifically, our results showed that SRF promotes GC metastasis and the epithelial to mesenchymal transition (EMT) though miR-199a-5p-mediated downregulation of E-cadherin. The present study thus provides insight into the specific biological behavior of SRF in GC metastasis. As increased activity of the SRF/miR-199a-5p/E-cadherin pathway appears to promote GC cell EMT and metastasis, these regulators may therefore be developed as therapeutic targets or biomarkers for GC progression.


Assuntos
Transição Epitelial-Mesenquimal , MicroRNAs/metabolismo , Fator de Resposta Sérica/fisiologia , Neoplasias Gástricas/metabolismo , Antígenos CD , Sequência de Bases , Sítios de Ligação , Caderinas/genética , Caderinas/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Linfática , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Invasividade Neoplásica , Regiões Promotoras Genéticas , Interferência de RNA , Neoplasias Gástricas/patologia , Transcriptoma , Via de Sinalização Wnt
16.
Invest Ophthalmol Vis Sci ; 55(7): 4560-7, 2014 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-25056592

RESUMO

Tissue contraction and fibrosis are major causes of morbidity in the human body. In the eye in particular, fibrosis and scarring are responsible for the pathogenesis or failure of treatment of all major blinding diseases, with postoperative wound healing responses posing a major problem for most ocular surgery on a worldwide scale. This is one of the largest areas of unmet need in ophthalmology, with currently no antifibrotic treatments available clinically. This review focuses on the ubiquitous myocardin-related transcription factor/serum response factor (MRTF-A/SRF) transcription pathway as a potential novel therapeutic target in fibrotic eye diseases. It describes how the MRTF-A/SRF pathway is intricately linked to all the key regulators and pathways in ocular fibrosis, and how it could potentially lead to a new avenue of antifibrotic therapies in the future.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Olho/patologia , Proteínas de Fusão Oncogênica/fisiologia , Fator de Resposta Sérica/fisiologia , Olho/enzimologia , Fibrose , Humanos , Metaloproteinases da Matriz/metabolismo , Transativadores
17.
Expert Opin Ther Targets ; 18(6): 633-49, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24660778

RESUMO

INTRODUCTION: Prostate cancer is a leading cause of cancer-related death in men and current treatments offer only a modest survival benefit in advanced stages of the disease. RNA interference (RNAi) is a therapeutic option that has received great attention in recent years with the potential to treat a variety of disorders, including prostate cancer. Transcription factors are cellular proteins that can up-regulate or down-regulate the transcription of genes and offer promising therapeutic targets. AREAS COVERED: This review will focus on transcription factors that have been identified as key molecules in drug resistance, disease progression and metastases in prostate cancer, which may offer potential as therapeutic targets for RNAi in the future. EXPERT OPINION: By identifying therapeutically viable transcription factor targets in prostate cancer, it is hoped that treatment strategies using RNAi will augment the effect of current chemotherapy regimens, slow disease progression and reduce metastases in prostate cancer, resulting in disease regression.


Assuntos
Neoplasias da Próstata/terapia , Interferência de RNA , Fatores de Transcrição/fisiologia , Antagonistas de Androgênios/uso terapêutico , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/fisiologia , Humanos , Masculino , NF-kappa B/metabolismo , Receptores Androgênicos/fisiologia , Fatores de Transcrição STAT/fisiologia , Fator de Resposta Sérica/fisiologia , Fator de Transcrição AP-1/fisiologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
18.
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
19.
Curr Biol ; 23(18): R843-5, 2013 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-24070445

RESUMO

Flies can form a visually-guided working memory. A new study shows that the gene termed ellipsoid body open influences multiple signals to regulate a competence factor in the ellipsoid body to support normal working memory.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Regulação da Expressão Gênica , Carioferinas/fisiologia , Memória de Curto Prazo , Proteínas dos Microfilamentos/fisiologia , Fator de Resposta Sérica/fisiologia , Animais
20.
Curr Biol ; 23(18): 1756-63, 2013 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-24012317

RESUMO

BACKGROUND: Navigation through the environment requires a working memory for the chosen target and path integration facilitating an approach when the target becomes temporarily hidden. We have previously shown that this visual orientation memory resides in the ellipsoid body, which is part of the central complex in the Drosophila brain. Former analysis of foraging and ignorant mutants have revealed that a hierarchical PKG and RSKII kinase signaling cascade in a subset of the ellipsoid-body ring neurons is required for this type of working memory in flies. RESULTS: Here we show that mutants in the ellipsoid body open (ebo) gene, which encodes the actin-binding protein Exportin 6, exhibit excessive nuclear accumulation of actin during development and in the adult brain. ebo mutants lack the orientation memory independent of the structural defect in the ellipsoid-body neuropil, and EBO activity in any type of adult ring neurons is sufficient for orientation-memory function. Moreover, genetic interaction studies revealed that nuclear actin accumulation in ebo mutants inhibits the Drosophila coactivator myocardin-related transcription factor A (dMRTF) and therefore the transcriptional activator serum response factor (dSRF). dSRF also functions in different ring neurons, suggesting that it regulates abundance of a diffusible factor that enables a working memory in ellipsoid-body ring neurons. CONCLUSIONS: To date, SRF has only been implicated in longer forms of memory formation like synaptic long-term potentiation and depression. This study provides the first evidence that SRF-mediated gene regulation is also required for a working memory that lasts only for a few seconds.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Regulação da Expressão Gênica , Carioferinas/fisiologia , Memória de Curto Prazo , Proteínas dos Microfilamentos/fisiologia , Fator de Resposta Sérica/fisiologia , Actinas/metabolismo , Animais , Núcleo Celular/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiologia , Carioferinas/genética , Carioferinas/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Modelos Genéticos , Mutação , Orientação , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Percepção Visual/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...