Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 296: 100261, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33837725

RESUMO

GPR56 is a member of the adhesion G-protein-coupled receptor family shown to play important roles in cell adhesion, brain development, immune function, and tumorigenesis. GPR56 is highly upregulated in colorectal cancer and correlates with poor prognosis. Several studies have shown GPR56 couples to the Gα12/13 class of heterotrimeric G-proteins to promote RhoA activation. However, due to its structural complexity and lack of a high-affinity receptor-specific ligand, the complete GPR56 signaling mechanism remains largely unknown. To delineate the activation mechanism and intracellular signaling functions of GPR56, we generated a monoclonal antibody (mAb) that binds with high affinity and specificity to the extracellular domain (ECD). Using deletion mutants, we mapped the mAb binding site to the GAIN domain, which mediates membrane-proximal autoproteolytic cleavage of the ECD. We showed that GPR56 overexpression in 293T cells leads to increased phosphorylation of Src, Fak, and paxillin adhesion proteins and activation of the Gα12/13-RhoA-mediated serum response factor (SRF) pathway. Treatment with the mAb potentiated Src-Fak phosphorylation, RhoA-SRF signaling, and cell adhesion. Consistently, GPR56 knockdown in colorectal cancer cells decreased Src-Fak pathway phosphorylation and cell adhesion. Interestingly, GPR56-mediated activation of Src-Fak phosphorylation occurred independent of RhoA, yet mAb-induced potentiation of RhoA-SRF signaling was Src-dependent. Furthermore, we show that the C-terminal portion of the Serine-Threonine-Proline-rich (STP) region, adjacent to the GAIN domain, was required for Src-Fak activation. However, autoproteolytic cleavage of the ECD was dispensable. These data support a new ECD-dependent mechanism by which GPR56 functions to regulate adhesion through activation of Src-Fak signaling.


Assuntos
Neoplasias Colorretais/genética , Quinase 1 de Adesão Focal/genética , Receptores Acoplados a Proteínas G/genética , Fator de Resposta Sérica/genética , Proteína rhoA de Ligação ao GTP/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Carcinogênese/genética , Adesão Celular/imunologia , Linhagem Celular Tumoral , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Quinase 1 de Adesão Focal/imunologia , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Paxilina/genética , Paxilina/imunologia , Fosforilação/genética , Receptores Acoplados a Proteínas G/imunologia , Fator de Resposta Sérica/imunologia , Transdução de Sinais/genética , Proteína rhoA de Ligação ao GTP/imunologia , Quinases da Família src/genética , Quinases da Família src/imunologia
2.
Curr Opin Hematol ; 22(1): 67-73, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25402621

RESUMO

PURPOSE OF REVIEW: Neutrophils rapidly migrate to sites of injury and infection. Egress of neutrophils from the circulation into tissues is a highly regulated process involving several distinct steps. Cell-cell interactions mediated by selectins and integrins and reorganization of the actin cytoskeleton are key mechanisms facilitating appropriate neutrophil recruitment. Neutrophil function is impaired in inherited and acquired disorders, such as leukocyte adhesion deficiency and myelodysplasia. Since the discovery that deletion of all or part of chromosome 5 is the most common genetic aberration in myelodysplasia, the roles of several of the deleted genes have been investigated in hematopoiesis. Several genes encoding proteins of the serum response factor (SRF) pathway are located on 5q. This review focuses, in particular, on the role of SRF in myeloid maturation and neutrophil function. RECENT FINDINGS: SRF and its pathway fulfill multiple complex roles in the regulation of the innate and adaptive immune system. Loss of SRF leads to defects in B-cell and T-cell development. SRF-deficient macrophages fail to spread, transmigrate, and phagocytose bacteria, and SRF-deficient neutrophils show defective chemotaxis in vitro and in vivo with failure of inside-out activation and trafficking of the Mac1 integrin complex. Loss of the formin mammalian Diaphanous 1, a regulator of linear actin polymerization and mediator of Ras homolog family member A signaling to SRF, results in aberrant myeloid differentiation and hyperactivity of the immune system. SUMMARY: SRF is an essential transcription factor in hematopoiesis and mature myeloid cell function. SRF regulates neutrophil migration, integrin activation, and trafficking. Disruption of the SRF pathway results in myelodysplasia and immune dysfunction.


Assuntos
Movimento Celular/imunologia , Neutrófilos/imunologia , Fator de Resposta Sérica/imunologia , Transdução de Sinais/imunologia , Animais , Movimento Celular/genética , Cromossomos Humanos Par 5/genética , Cromossomos Humanos Par 5/imunologia , Hematopoese/genética , Hematopoese/imunologia , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Integrinas/genética , Integrinas/imunologia , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/imunologia , Síndromes Mielodisplásicas/patologia , Neutrófilos/patologia , Fator de Resposta Sérica/genética , Transdução de Sinais/genética
3.
Fish Shellfish Immunol ; 36(1): 75-82, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24161761

RESUMO

Serum response factor (SRF) function is essential for transcriptional regulation of numerous growth-factor-inducible genes and triggers proliferation, differentiation and apoptosis of the cells. In this report, the first mollusk serum response factor like homolog gene (designated ChSRF) was identified and characterized from the Hong Kong oyster, Crassostrea hongkongensis. The full-length cDNA of ChSRF was 1716 bp in length and encodes a putative protein of 434 amino acids respectively, and shares the MADS domain at the N-terminal. ChSRF is ubiquitously expressed in various tissues, with the highest expression level observed in muscle. Temporal expression of ChSRF following microbe infection shows that the expression of ChSRF in hemocytes increases from 3 to 24 h post-challenge. As a target gene of SRF, ß-actin demonstrates a similar gene expression mode in constitutive tissue and pathogen infection. Furthermore, some protein profiles of ChSRF was revealed, fluorescence microscopy results show that ChSRF located in the nuclei of HeLa cells and over-expression of ChSRF activated the transcriptional activities of MAPK signal pathway in HEK293T cells. These results indicate that ChSRF maybe play an important role in signal transduction in the immunity and development response of oysters. Furthermore, we found that ChSRF could regulate the expression of ß-actin gene, which indicate that ChSRF is a muscle differentiation regulator in the oyster and it will help us to improve aquaculture production.


Assuntos
Crassostrea/imunologia , Regulação da Expressão Gênica/imunologia , Filogenia , Fator de Resposta Sérica/imunologia , Vibrioses/imunologia , Vibrio alginolyticus/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Crassostrea/genética , Crassostrea/virologia , Células HeLa , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , RNA/química , RNA/genética , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real , Alinhamento de Sequência , Análise de Sequência de DNA , Fator de Resposta Sérica/genética , Transfecção/métodos , Vibrioses/virologia
4.
J Basic Clin Physiol Pharmacol ; 24(4): 287-97, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23893683

RESUMO

BACKGROUND: Modulation of inflammatory signaling has been elucidated in several disease models. Acrolein, an environmental pollutant, has been linked to diseases such as atherosclerosis and to the inflammatory process involving nuclear factor κB (NFκB). Serum response factor (SRF), a transcription factor, regulates cell development, differentiation and proliferation through signaling molecules such as extracellular signal-regulated kinase 1/2 (ERK1/2) and CD36. We hypothesized that acrolein toxicity involves SRF in the process of activating NFκB and may involve CD36/ERK1/2. METHODS: Vascular smooth muscle cells (VSMCs) were exposed to acrolein (0.5 µg/mL) in the presence or absence of 10 nM QNZ (NFκB inhibitor), 300 nM CCG1423 (SRF inhibitor) and 50 µM PD98059 (ERK1/2 inhibitor). Protein and RNA were isolated. Changes in expression were determined by Western blot and polymerase chain reaction (PCR) array. RESULTS: Subtoxic doses of acrolein increased ERK1/2, SRF and NFκB protein expression, whereas CD36 expression was unchanged. Increase in NFκB expression was accompanied by an increase in activity. ERK1/2 inhibition only blunted SRF expression. SRF inhibition blunted NFκB expression but not that of ERK1/2. CD36 expression was unchanged in the presence of either inhibitor. PCR array analysis indicated up-regulation of nine genes (>4- to 50-fold) and down-regulation of six genes (>4- to 12-fold) involved in inflammatory signaling. CONCLUSIONS: We propose that SRF is required in acrolein activation of NFκB and is ERK1/2 dependent.


Assuntos
Acroleína/toxicidade , Poluentes Ambientais/toxicidade , Inflamação/induzido quimicamente , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fator de Resposta Sérica/imunologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Inflamação/genética , Inflamação/imunologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Músculo Liso Vascular/imunologia , Miócitos de Músculo Liso/imunologia , NF-kappa B/genética , NF-kappa B/imunologia , Ratos , Fator de Resposta Sérica/antagonistas & inibidores , Fator de Resposta Sérica/genética
5.
Am J Physiol Endocrinol Metab ; 305(1): E1-14, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23632636

RESUMO

Obesity is associated with low-grade chronic inflammation, which contributes to cellular dysfunction promoting metabolic disease. Obesity during pregnancy leads to a proinflammatory milieu in the placenta; however, the underlying causes for obesity-induced placental inflammation remain unclear. Here, we examine the mechanisms by which saturated fatty acids and inflammatory cytokines induce inflammation in placental trophoblasts. We conducted global transcriptomic profiling in BeWo cells following palmitate and/or TNFα treatment and gene/protein expression analyses of MAPK pathways and characterized downstream transcription factors directly regulating inflammatory cytokines. Microarray analysis revealed increased expression of genes regulating inflammation, stress response, and immediate early response in cytotrophoblasts in response to palmitic acid (PA), TNFα, or a combination of both (PA + TNFα). Both gene ontology and gene set enrichment analysis revealed MAPK and EGR-1 signaling to be upregulated in BeWo cells, which was confirmed via immunoblotting. Importantly, activation of JNK signaling was necessary for increased proinflammatory cytokine (IL-6, TNFα, and IL-8) and EGR1 mRNA. Consistent with the requirement of JNK signaling, ChIP analysis confirmed the recruitment of c-Jun and other MAPK-responsive immediate early factors on the EGR1 promoter. Moreover, recruitment of EGR-1 on cytokine promoters (IL-6, TNFα, and IL-8) and an impaired proinflammatory response following knockdown of EGR-1 suggested it as a central component of the mechanism facilitating inflammatory gene expression. Finally, akin to in vitro findings, term placenta from obese women also had both increased JNK and p38 signaling and greater EGR-1 protein relative to lean women. Our results demonstrate that lipotoxic insults induce inflammation in placental cells via activation of JNK/EGR-1 signaling.


Assuntos
Proteína 1 de Resposta de Crescimento Precoce/imunologia , Metabolismo dos Lipídeos/imunologia , Obesidade/imunologia , Placenta/imunologia , Complicações na Gravidez/imunologia , Fator 3 Ativador da Transcrição/imunologia , Fator 3 Ativador da Transcrição/metabolismo , Linhagem Celular , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Feminino , Humanos , Recém-Nascido , Interleucina-6/genética , Interleucina-6/imunologia , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/imunologia , Interleucina-8/metabolismo , Metabolismo dos Lipídeos/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/imunologia , Masculino , Palmitatos/farmacologia , Placenta/citologia , Gravidez , Fator de Resposta Sérica/imunologia , Fator de Resposta Sérica/metabolismo , Transcriptoma/efeitos dos fármacos , Transcriptoma/imunologia , Trofoblastos/citologia , Trofoblastos/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
6.
J Biol Chem ; 287(4): 2459-67, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22157009

RESUMO

Smooth muscle cell (SMC) differentiation is defined largely by a number of cell-restricted genes governed directly by the serum response factor (SRF)/myocardin (MYOCD) transcriptional switch. Here, we describe a new SRF/MYOCD-dependent, SMC-restricted gene known as Leiomodin 1 (Lmod1). Conventional and quantitative RT-PCRs indicate that Lmod1 mRNA expression is enriched in SMC-containing tissues of the mouse, whereas its two paralogs, Lmod2 and Lmod3, exhibit abundant expression in skeletal and cardiac muscle with very low levels in SMC-containing tissues. Western blotting and immunostaining of various adult and embryonic mouse tissues further confirm SMC-specific expression of the LMOD1 protein. Comparative genomic analysis of the human LMOD1 and LMOD2 genes with their respective mouse and rat orthologs shows high conservation between the three exons and several noncoding sequences, including the immediate 5' promoter region. Two conserved CArG boxes are present in both the LMOD1 and LMOD2 promoter regions, although LMOD1 displays much higher promoter activity and is more responsive to SRF/MYOCD stimulation. Gel shift assays demonstrate clear binding between SRF and the two CArG boxes in human LMOD1. Although the CArG boxes in LMOD1 and LMOD2 are similar, only LMOD1 displays SRF or MYOCD-dependent activation. Transgenic mouse studies reveal wild type LMOD1 promoter activity in cardiac and vascular SMC. Such activity is abolished upon mutation of both CArG boxes. Collectively, these data demonstrate that Lmod1 is a new SMC-restricted SRF/MYOCD target gene.


Assuntos
Autoantígenos/biossíntese , Diferenciação Celular/fisiologia , Proteínas do Citoesqueleto/biossíntese , Regulação da Expressão Gênica/fisiologia , Proteínas Musculares/biossíntese , Miócitos de Músculo Liso/metabolismo , Elemento de Resposta Sérica/fisiologia , Fator de Resposta Sérica/imunologia , Animais , Autoantígenos/genética , Células COS , Chlorocebus aethiops , Proteínas do Citoesqueleto/genética , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/biossíntese , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , Miócitos de Músculo Liso/citologia , Células NIH 3T3 , Especificidade de Órgãos/fisiologia , Ratos , Fator de Resposta Sérica/genética
7.
Cardiovasc Res ; 70(1): 136-45, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16451796

RESUMO

OBJECTIVE: Smoothelin-A and -B isoforms are highly restricted to contractile smooth muscle cells (SMCs). Serum response factor (SRF) and myocardin are essential for contractile SMC differentiation. We evaluated the contribution of SRF/myocardin to transcriptional regulation of smoothelins. METHODS: Rat vascular SMCs were transfected with smoothelin-A and smoothelin-B promoter reporter constructs and promoter activity was analyzed. The effects of mutations in the smoothelin-A promoter CArG-boxes and co-transfections with a myocardin expression plasmid were assessed. Electrophoretic mobility shift assays and chromatin immunoprecipitations were performed to investigate SRF-binding to the smoothelin-A CArG-boxes. RESULTS: Smoothelin promoter activity was detected in vascular SMCs. Comparative sequence analysis revealed two conserved CArG elements in the smoothelin-A promoter that bind SRF as shown by chromatin immunoprecipitation. The proximal CArG-near bound SRF stronger than CArG-far in gel shift assays. Mutagenesis studies also indicated that CArG-near is more important than CArG-far in regulating smoothelin-A promoter activity. Myocardin augmented smoothelin-A promoter activity 2.5-fold in a CArG-near-dependent manner. In contrast, myocardin had little effect on the smoothelin-B promoter. CONCLUSION: Smoothelin-A expression is controlled by an intragenic promoter whose activity is, in part, dependent on two CArG boxes that bind SRF. Our data show a role for SRF/myocardin in regulating smoothelin-A whereas the higher smoothelin-B expression appears to be SRF/myocardin-independent.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Regulação da Expressão Gênica , Proteínas Musculares/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Fator de Resposta Sérica/metabolismo , Transativadores/metabolismo , Animais , Sequência de Bases , Células Cultivadas , Proteínas do Citoesqueleto/genética , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Expressão Gênica , Humanos , Imunoprecipitação/métodos , Camundongos , Dados de Sequência Molecular , Proteínas Musculares/genética , Proteínas Nucleares/genética , Reação em Cadeia da Polimerase , Ligação Proteica , RNA Mensageiro/análise , Ratos , Análise de Sequência de DNA , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/imunologia , Transativadores/genética , Transcrição Gênica
8.
Circulation ; 108(4): 407-13, 2003 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-12874181

RESUMO

BACKGROUND: Knowledge about molecular mechanisms leading to heart failure is still limited, but reduced gene activities and modest activation of caspase 3 are hallmarks of end-stage heart failure. We postulated that serum response factor (SRF), a central cardiac transcription factor, might be a cleavage target for modest activated caspase 3, and this cleavage of SRF may play a dominant inhibitory role in propelling hearts toward failure. METHODS AND RESULTS: We examined SRF protein levels from cardiac samples taken at the time of transplantation in 13 patients with end-stage heart failure and 7 normal hearts. Full-length SRF was markedly reduced and processed into 55- and 32-kDa subfragments in all failing hearts. SRF was intact in normal samples. In contrast, the hearts of 10 patients with left ventricular assist devices showed minimal SRF fragmentation. Specific antibodies to N- and C-terminal SRF sequences and site-directed mutagenesis revealed 2 alternative caspase 3 cleavage sites, so that 2 fragments were detected of each containing either the N- or C-terminal SRF. Expression of SRF-N, the 32-kDa fragment, in myogenic cells inhibited the transcriptional activity of alpha-actin gene promoters by 50% to 60%, which suggests that truncated SRF functioned as a dominant-negative transcription factor. CONCLUSIONS: Caspase 3 activation in heart failure sequentially cleaved SRF and generated a dominant-negative transcription factor, which may explain the depression of cardiac-specific genes. Moreover, caspase 3 activation may be reversible in the failing heart with ventricular unloading.


Assuntos
Caspases/metabolismo , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração/fisiopatologia , Fragmentos de Peptídeos/biossíntese , Fator de Resposta Sérica/biossíntese , Fatores de Transcrição/biossíntese , Actinas/genética , Animais , Apoptose , Caspase 3 , Células Cultivadas , Ativação Enzimática , Genes Reporter , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/terapia , Transplante de Coração , Ventrículos do Coração/cirurgia , Coração Auxiliar , Humanos , Camundongos , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/fisiologia , Indução de Remissão , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/imunologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...