Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Exp Biol Med (Maywood) ; 237(5): 593-607, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22619371

RESUMO

Peri-transplant surgical trauma and ischemia/reperfusion injury in accepted murine heterotopic heart grafts has been associated with myofibroblast differentiation, cardiac fibrosis and biomechanical-stress activation of the fetal myocardial smooth muscle α-actin (SMαA) gene. The wound-healing agonists, transforming growth factor ß1 and thrombin, are known to coordinate SMαA mRNA transcription and translation in activated myofibroblasts by altering the subcellular localization and mRNA-binding affinity of the Y-box binding protein-1 (YB-1) cold-shock domain (CSD) protein that governs a variety of cellular responses to metabolic stress. YB-1 accumulated in polyribosome-enriched regions of the sarcoplasm proximal to cardiac intercalated discs in accepted heart grafts. YB-1 binding to a purine-rich motif in exon 3 of SMαA mRNA that regulates translational efficiency increased substantially in perfusion-isolated, rod-shaped adult rat cardiomyocytes during phenotypic de-differentiation in the presence of serum-derived growth factors. Cardiomyocyte de-differentiation was accompanied by the loss of a 60 kDa YB-1 variant that was highly expressed in both adult myocardium and freshly isolated myocytes and replacement with the 50 kDa form of YB-1 (p50) typically expressed in myofibroblasts that demonstrated sequence-specific interaction with SMαA mRNA. Accumulation of p50 YB-1 in reprogrammed, de-differentiated myocytes was associated with a 10-fold increase in SMαA protein expression. Endomyocardial biopsies collected from patients up to 14 years after heart transplant showed variable yet coordinately elevated expression of SMαA and p50 YB-1 protein and demonstrable p50 YB-1:SMαA mRNA interaction. The p60 YB-1 variant in human heart graft samples, but neither mouse p60 nor mouse or human p50, reacted with an antibody specific for the phosphoserine 102 modification in the YB-1 CSD. Modulation of YB-1 subcellular compartmentalization and mRNA-binding activity may be linked with reprogramming of contractile protein gene expression in ventricular cardiomyocytes that could contribute to maladaptive remodeling in accepted, long-term heart grafts.


Assuntos
Transplante de Coração , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Proteína 1 de Ligação a Y-Box/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Animais Recém-Nascidos , Ensaio de Desvio de Mobilidade Eletroforética , Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Músculo Liso Vascular/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miofibroblastos/metabolismo , Regiões Promotoras Genéticas , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transcrição Gênica , Transplante Heterotópico , Cicatrização , Proteína 1 de Ligação a Y-Box/genética
3.
Mol Biol Cell ; 20(8): 2174-85, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19261809

RESUMO

Transforming growth factor (TGF) beta1 is a mediator of myofibroblast differentiation in healing wounds in which it activates transcription of the smooth muscle alpha-actin (SMalphaA) gene via dynamic interplay of nuclear activators and repressors. Targeting components of TGFbeta1 signaling may be an effective strategy for controlling myofibroblasts in chronic fibrotic diseases. We examined the ability of proinflammatory tumor necrosis factor (TNF)-alpha to antagonize TGFbeta1-mediated human pulmonary myofibroblast differentiation. TNF-alpha abrogated TGFbeta1-induced SMalphaA gene expression at the level of transcription without disrupting phosphorylation of regulatory Smads. Intact mitogen-activated protein kinase kinase (Mek)-extracellular signal-regulated kinase (Erk) kinase signaling was required for myofibroblast repression by TNF-alpha via induction of the early growth response factor-1 (Egr-1) DNA-binding protein. Egr-1 bound to the GC-rich SPUR activation element in the SMalphaA promoter and potently suppressed Smad3- and TGFbeta1-mediated transcription. Reduction in Smad binding to the SMalphaA promoter in TNF-alpha-treated myofibroblasts was accompanied by an increase in Egr-1 and YB-1 repressor binding, suggesting that the molecular mechanism underlying repression may involve competitive interplay between Egr-1, YB-1, and Smads. The ability of TNF-alpha to attenuate myofibroblast differentiation via modulation of a Mek1/Erk/Egr-1 regulatory axis may be useful in designing new therapeutic targets to offset destructive tissue remodeling in chronic fibrotic disease.


Assuntos
Actinas/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Fibroblastos/enzimologia , Regulação da Expressão Gênica/efeitos dos fármacos , MAP Quinase Quinase 1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Humanos , Pulmão/citologia , Modelos Genéticos , Músculo Liso/metabolismo , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Proteínas Repressoras/metabolismo , Proteína Smad2/metabolismo , Proteína Smad7/metabolismo , Transcrição Gênica/efeitos dos fármacos , Proteína 1 de Ligação a Y-Box/metabolismo
4.
Am J Physiol Cell Physiol ; 294(3): C702-14, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18344281

RESUMO

Mouse hearts subjected to repeated transplant surgery and ischemia-reperfusion injury develop substantial interstitial and perivascular fibrosis that was spatially associated with dysfunctional activation of fetal smooth muscle alpha-actin (SM alpha A) gene expression in graft ventricular cardiomyocytes. Compared with cardiac fibroblasts in which nuclear levels of the Sp1 and Smad 2/3 transcriptional-activating proteins increased markedly after transplant injury, the most abundant SM alpha A gene-activating protein in cardiomyocyte nuclei was serum response factor (SRF). Additionally, cardiac intercalated discs in heart grafts contained substantial deposits of Pur alpha, an mRNA-binding protein and known negative modulator of SRF-activated SM alpha A gene transcription. Activation of fetal SM alpha A gene expression in perfusion-isolated adult cardiomyocytes was linked to elevated binding of a novel protein complex consisting of SRF and Pur alpha to a purine-rich DNA element in the SM alpha A promoter called SPUR, previously shown to be required for induction of SM alpha A gene transcription in injury-activated myofibroblasts. Increased SRF binding to SPUR DNA plus one of two nearby CArG box consensus elements was observed in SM alpha A-positive cardiomyocytes in parallel with enhanced Pur alpha:SPUR protein:protein interaction. The data suggest that de novo activation of the normally silent SM alpha A gene in reprogrammed adult cardiomyocytes is linked to elevated interaction of SRF with fetal-specific CArG and injury-activated SPUR elements in the SM alpha A promoter as well as the appearance of novel Pur alpha protein complexes in both the nuclear and cytosolic compartments of these cells.


Assuntos
Actinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Miócitos Cardíacos/metabolismo , Proteínas Repressoras/metabolismo , Fator de Resposta Sérica/metabolismo , Estresse Fisiológico/metabolismo , Abdome/cirurgia , Actinas/genética , Animais , Células COS , Chlorocebus aethiops , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Feminino , Fibroblastos/metabolismo , Fibrose , Rejeição de Enxerto/genética , Rejeição de Enxerto/metabolismo , Transplante de Coração , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Músculo Liso Vascular/embriologia , Músculo Liso Vascular/metabolismo , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/patologia , Proteínas do Tecido Nervoso/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Transdução de Sinais , Estresse Fisiológico/genética , Estresse Fisiológico/patologia , Estresse Fisiológico/fisiopatologia , Fatores de Tempo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transfecção , Transplante Heterotópico , Remodelação Ventricular
5.
J Biol Chem ; 282(49): 35899-909, 2007 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-17906292

RESUMO

Previous studies from our laboratories have implicated two members of the Pur family of single-stranded DNA/RNA-binding proteins, Pur alpha and Pur beta, in transcriptional repression of the smooth muscle alpha-actin gene in vascular cell types. Although Pur alpha and Pur beta share substantial sequence homology and nucleic acid binding properties, genomic promoter and cis-element occupancy studies reported herein suggest that Pur beta is the dominant factor in gene regulation. To dissect the molecular basis of Pur beta repressor activity, site-directed mutagenesis was used to map amino acids critical to the physical and functional interaction of Pur beta with the smooth muscle alpha-actin promoter. Of all the various acidic, basic, and aromatic residues studied, mutation of positionally conserved arginines in the class I or class II repeat modules significantly attenuated Pur beta repressor activity in transfected vascular smooth muscle cells and fibroblasts. DNA binding and protein-protein interaction assays were conducted with purified recombinant Pur beta and selected mutants to reveal the physical basis for loss-of-function. Mutants R57E, R57E/R96E, and R57A/R96A each exhibited reduced single-stranded DNA binding affinity for an essential promoter element and diminished interaction with corepressor YB-1/MSY1. Structural analyses of the R57A/R96A and R57E/R96E double mutants in comparison to the wild-type Pur beta homodimer revealed aberrant self-association into higher order oligomeric complexes, which correlated with decreased alpha-helical content and defective DNA and protein binding in vitro. These findings point to a previously unrecognized structural role for certain core arginine residues in forming a conformationally stable Pur beta protein capable of physical interactions necessary for smooth muscle alpha-actin gene repression.


Assuntos
Actinas/biossíntese , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/metabolismo , Inativação Gênica , Músculo Liso Vascular/metabolismo , Proteínas Repressoras/metabolismo , Elementos de Resposta/fisiologia , Actinas/genética , Substituição de Aminoácidos , Animais , Linhagem Celular , DNA de Cadeia Simples/genética , Proteínas de Ligação a DNA/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Camundongos , Músculo Liso Vascular/citologia , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica/genética , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/genética , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
6.
Mol Biol Cell ; 18(12): 4837-46, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17881730

RESUMO

The phenotypic switch of cardiac fibroblasts (CFs) to myofibroblasts is essential for normal and pathological wound healing. Relative hyperoxic challenge during reoxygenation causes myocardial remodeling. Here, we sought to characterize the novel O(2)-sensitive molecular mechanisms responsible for triggering the differentiation of CFs to myofibroblasts. Exposure of CFs to hyperoxic challenge-induced transcription of smooth muscle actin (SMA) and enhanced the stability of both Acta2 transcript as well as of SMA protein. Both p21 deficiency as well as knockdown blunted hyperoxia-induced Acta2 and SMA response. Strikingly, overexpression of p21 alone markedly induced differentiation of CFs under normoxia. Overexpression of p21 alone induced SMA transcription by down-regulating YB1 and independent of TGFbeta1. In vivo, hyperoxic challenge induced p21-dependent differentiation of CFs to myofibroblasts in the infarct boundary region of ischemia-reperfused heart. Tissue elements were laser-captured from infarct boundary and from a noninfarct region 0.5 mm away. Reperfusion caused marked p21 induction in the infarct region. Acta2 as well as SMA expression were markedly up-regulated in CF-rich infarct boundary region. Of note, ischemia-reperfusion-induced up-regulation of Acta2 in the infarct region was completely abrogated in p21-deficient mice. This observation establishes p21 as a central regulator of reperfusion-induced phenotypic switch of CFs to myofibroblasts.


Assuntos
Actinas/metabolismo , Diferenciação Celular , Músculo Liso/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Animais , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21 , Fibroblastos , Regulação da Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/farmacologia , RNA Mensageiro/genética
7.
Am J Physiol Cell Physiol ; 293(1): C238-45, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17392380

RESUMO

Isoforms of the smooth muscle myosin motor, SM1 and SM2, differ in length at the carboxy terminal tail region. Their proportion changes with development, hormonal status and disease, but their function is unknown. We developed mice carrying the myosin heavy chain (MyHC) transgenes SM1, cMyc-tagged SM1, SM2, and V5-tagged SM2, and all transgenes corresponded to the SMa NH(2)-terminal isoform. Transgene expression was targeted to smooth muscle by the smooth muscle alpha-actin promoter. Immunoblot analysis showed substantial expression of the cMyc-tagged SM1 and V5-tagged SM2 MyHC protein in aorta and bladder and transgene mRNA was expressed in mice carrying unlabeled SM1 or SM2 transgenes. Despite significant protein expression of tagged MyHCs we found only small changes in the SM1:SM2 protein ratio. Significant changes in functional phenotype were observed in mice carrying unlabeled SM1 or SM2 transgenes. Force in aorta and bladder was increased (72 +/- 14%, 92 +/- 11%) in SM1 and decreased to 57 +/- 1% and 80 +/- 3% in SM2 transgenic mice. SM1 transgenic bladders had faster (1.8 +/- 0.3 s) and SM2 slower (7.1 +/- 0.5 s) rates of force redevelopment following a rapid step shortening. We hypothesize that small changes in the SM1:SM2 ratio could be amplified if they are associated with changes in thick filament assembly and underlie the altered contractility. These data provide evidence indicating an in vivo function for the COOH-terminal isoforms of smooth muscle myosin and suggest that the SM1:SM2 ratio is tightly regulated in smooth muscle tissues.


Assuntos
Aorta/metabolismo , Expressão Gênica , Músculo Liso/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Miosinas de Músculo Liso/metabolismo , Bexiga Urinária/metabolismo , Actinas/genética , Animais , Aorta/efeitos dos fármacos , Relação Dose-Resposta a Droga , Cinética , Camundongos , Camundongos Transgênicos , Contração Muscular , Força Muscular , Músculo Liso/efeitos dos fármacos , Cadeias Pesadas de Miosina/química , Cadeias Pesadas de Miosina/genética , Fenótipo , Cloreto de Potássio/farmacologia , Regiões Promotoras Genéticas , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Ratos , Miosinas de Músculo Liso/química , Miosinas de Músculo Liso/genética , Bexiga Urinária/efeitos dos fármacos
8.
Am J Physiol Heart Circ Physiol ; 292(3): H1541-50, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17114245

RESUMO

Although it has been shown that endothelial nitric oxide synthase (eNOS)-derived nitric oxide downregulates mitochondrial oxygen consumption during early reperfusion, its effects on inducible NOS (iNOS) induction and myocardial injury during late reperfusion are unknown. Wild-type (WT) and eNOS(-/-) mice were subjected to 30 min of coronary ligation followed by reperfusion. Expression of iNOS mRNA and protein levels and peroxynitrite production were lower in postischemic myocardium of eNOS(-/-) mice than levels in WT mice 48 h postreperfusion. Significantly improved hemodynamics (+/-dP/dt, left ventricular systolic pressure, mean arterial pressure), increased rate pressure product, and reduced myocardial infarct size (18 +/- 2.5% vs. 31 +/- 4.6%) were found 48 h after reperfusion in eNOS(-/-) mice compared with WT mice. Myocardial infarct size was also significantly decreased in WT mice treated with the specific iNOS inhibitor 1400W (20.5 +/- 3.4%) compared with WT mice treated with PBS (33.9 +/- 5.3%). A marked reperfusion-induced hyperoxygenation state was observed by electron paramagnetic resonance oximetry in postischemic myocardium, but Po(2) values were significantly lower from 1 to 72 h in eNOS(-/-) than in WT mice. Cytochrome c-oxidase activity and NADH dehydrogenase activity were significantly decreased in postischemic myocardium in WT and eNOS(-/-) mice compared with baseline control, respectively, and NADH dehydrogenase activity was significantly higher in eNOS(-/-) than in WT mice. Thus deficiency of eNOS exerted a sustained beneficial effect on postischemic myocardium 48 h after reperfusion with preserved mitochondrial function, which appears to be due to decreased iNOS induction and decreased iNOS-derived peroxynitrite in postischemic myocardium.


Assuntos
Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Óxido Nítrico Sintase Tipo III/deficiência , Óxido Nítrico Sintase Tipo II/biossíntese , Consumo de Oxigênio , Animais , Genótipo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Cardíacas/enzimologia , Mitocôndrias Cardíacas/fisiologia , Reação em Cadeia da Polimerase
9.
Mol Cell Endocrinol ; 249(1-2): 21-31, 2006 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-16504375

RESUMO

At least four alternatively spliced mRNAs can be synthesized from the human AT(1)R (hAT(1)R) gene that differ only in the inclusion or exclusion of exon 2 and/or 3. RT-PCR experiments demonstrate that splice variants harboring exon 2 accounts for at least 30% of all the hAT(1)R mRNA transcripts expressed in the human tissues investigated. Since exon 2 contains two upstream AUGs or open reading frames (uORFs), we hypothesized that these AUGs would inhibit the translation of the downstream hAT(1)R protein ORF harbored in exon 4. This study demonstrates that the inclusion of exon 2 in hAT(1)R mRNA transcripts dramatically reduces hAT(1)R protein levels (nine-fold) and significantly attenuates Ang II responsiveness ( approximately four-fold). Interestingly, only when both AUGs were mutated in combination were the hAT(1)R density and Ang II signaling levels comparable with those values obtained using mRNA splice variants that did not include exon 2. This observation is consistent with a model where the majority of the ribosomes likely translate uORF#1 and are then unable to reinitiate at the downstream hAT(1)R ORF, in part due to the presence of AUG#2 and to the short intercistronic spacing. Importantly, TGF-beta(1) treatment (4ng/ml for 4h) of fibroblasts up-regulated hAT(1)R mRNA splice variants, which harbored exon 2, six-fold. Since AT(1)R activation is closely associated with cardiovascular disease, the inclusion of exon 2 by alternative splicing represents a novel mechanism to reduce the overall production of the hAT(1)R protein and possibly limit the potential pathological effects of AT(1)R activation.


Assuntos
Processamento Alternativo , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Fator de Crescimento Transformador beta/farmacologia , Regulação para Cima , Sequência de Bases , Códon de Iniciação/fisiologia , Éxons , Humanos , Dados de Sequência Molecular , Receptor Tipo 1 de Angiotensina/biossíntese , Alinhamento de Sequência , Fator de Crescimento Transformador beta/fisiologia
10.
J Biol Chem ; 281(12): 7907-18, 2006 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-16436378

RESUMO

Pur alpha and Pur beta are structurally related single-stranded DNA/RNA-binding proteins implicated in the control of cell growth and differentiation. The goal of this study was to determine whether Pur alpha and Pur beta function in a redundant, distinct, or collaborative manner to suppress smooth muscle alpha-actin gene expression in cell types relevant to wound repair and vascular remodeling. RNA interference-mediated loss-of-function analyses revealed that, although Pur beta was the dominant repressor, the combined action of endogenous Pur alpha and Pur beta was necessary to fully repress the full-length smooth muscle alpha-actin promoter in cultured fibroblasts but to a lesser extent in vascular smooth muscle cells. The activity of a minimal core enhancer containing a truncated 5' Pur repressor binding site was unaffected by knockdown of Pur alpha and/or Pur beta in fibroblasts. Conversely, gain-of-function studies indicated that Pur alpha or Pur beta could each independently repress core smooth muscle alpha-actin enhancer activity albeit in a cell type-dependent fashion. Biochemical analyses indicated that purified recombinant Pur alpha and Pur beta were essentially identical in terms of their binding affinity and specificity for GGN repeat-containing strands of several cis-elements comprising the core enhancer. However, Pur alpha and Pur beta exhibited more distinctive protein interaction profiles when evaluated for binding to enhancer-associated transcription factors in extracts from fibroblasts and vascular smooth muscle cells. These findings support the hypothesis that Pur alpha and Pur beta repress smooth muscle alpha-actin gene transcription by means of DNA strand-selective cis-element binding and cell type-dependent protein-protein interactions.


Assuntos
Actinas/metabolismo , Proteínas de Ligação a DNA/química , Miócitos de Músculo Liso/metabolismo , Proteínas do Tecido Nervoso/química , Nucleoproteínas/química , Regiões Promotoras Genéticas , Animais , Ligação Competitiva , Biotinilação , Western Blotting , DNA/química , DNA de Cadeia Simples/química , Relação Dose-Resposta a Droga , Elementos Facilitadores Genéticos , Ensaio de Imunoadsorção Enzimática , Epitopos/química , Fibroblastos/metabolismo , Genes Reporter , Vetores Genéticos , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/metabolismo , Ligação Proteica , RNA/química , Interferência de RNA , Fatores de Transcrição/química , Transcrição Gênica , Transgenes
11.
Mol Biol Cell ; 16(10): 4931-40, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16093352

RESUMO

Profibrotic regulatory mechanisms for tissue repair after traumatic injury have developed under strong evolutionary pressure to rapidly stanch blood loss and close open wounds. We have examined the roles played by two profibrotic mediators, transforming growth factor beta1 (TGFbeta1) and thrombin, in directing expression of the vascular smooth muscle alpha-actin (SMalphaA) gene, an important determinant of myofibroblast differentiation and early protein marker for stromal cell response to tissue injury. TGFbeta1 is a well known transcriptional activator of the SMalphaA gene in myofibroblasts. In contrast, thrombin independently elevates SMalphaA expression in human pulmonary myofibroblasts at the posttranscriptional level. A common feature of SMalphaA up-regulation mediated by thrombin and TGFbeta1 is the involvement of the cold shock domain protein YB-1, a potent repressor of SMalphaA gene transcription in human fibroblasts that also binds mRNA and regulates translational efficiency. YB-1 dissociates from SMalphaA enhancer DNA in the presence of TGFbeta1 or its Smad 2, 3, and 4 coregulatory mediators. Thrombin does not effect SMalphaA gene transcription but rather displaces YB-1 from SMalphaA exon 3 coding sequences previously shown to be required for mRNA translational silencing. The release of YB-1 from promoter DNA coupled with its ability to bind RNA and shuttle between the nucleus and cytoplasm is suggestive of a regulatory loop for coordinating SMalphaA gene output in human pulmonary myofibroblasts at both the transcriptional and translational levels. This loop may help restrict organ-destructive remodeling due to excessive myofibroblast differentiation.


Assuntos
Actinas/fisiologia , Proteínas de Ligação a DNA/fisiologia , Fibroblastos/citologia , Músculo Liso Vascular/metabolismo , Trombina/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Actinas/genética , Animais , Diferenciação Celular , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Elementos Facilitadores Genéticos , Éxons , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Inativação Gênica , Humanos , Recém-Nascido , Pulmão/citologia , Músculo Liso Vascular/citologia , Proteínas Nucleares , Regiões Promotoras Genéticas , Transporte Proteico , Ativação Transcricional , Fator de Crescimento Transformador beta1 , Proteína 1 de Ligação a Y-Box
12.
Brain Behav Immun ; 19(3): 207-16, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15797309

RESUMO

Previous research has shown that psychological stress delays wound closure by >25%. Gene expression of pro-inflammatory cytokines and the maturation of the epithelium were also impaired by stress (Mercado et al.). Wound contraction contributes to the speed of wound closure (Hunt and Hopf). In the current study, wound contraction was decreased by >45% (p<.01) in restraint stressed mice. Fibroblast migration and differentiation into smooth muscle alpha-actin (SmalphaA) -expressing myofibroblasts were delayed in RST mice through day 7 post-wounding. In addition, there was a 25 (p<.01), 48 (p<.01), and 38% (p<.05) decrease in SmalphaA mRNA levels at days 1, 3, and 5 post-wounding in RST mice, respectively. Cytokines that regulate fibroblast migration and differentiation include transforming growth factors-beta1, -beta2, and -beta3 (TGF-betas). Although expression of TGF-beta1 mRNA was downregulated by >25% (p<.01) in RST mice on day 3 post-wounding, no significant differences were detected in active or total TGF-beta1 protein levels. Stress did not alter the expression of TGF-beta2 or -beta3 through day 5 post-wounding. Thus, these data indicate that stress delays wound contraction and myofibroblast differentiation, which are likely independent of expression of TGF-beta1, -beta2, and -beta3.


Assuntos
Actinas/metabolismo , Fibroblastos/metabolismo , Estresse Psicológico/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Cicatrização/fisiologia , Actinas/genética , Animais , Diferenciação Celular/fisiologia , Feminino , Fibroblastos/citologia , Regulação da Expressão Gênica , Camundongos , RNA Mensageiro/análise , Restrição Física/fisiologia , Estresse Psicológico/genética , Fator de Crescimento Transformador beta/genética
13.
J Biol Chem ; 280(7): 6204-14, 2005 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-15576380

RESUMO

Expression of smooth muscle alpha-actin in growth factor-induced myofibroblasts and in differentiated vascular smooth muscle cells is transcriptionally controlled by multiple positive or negative trans-acting factors interacting with distinct cis-elements in the 5'-flanking region of the gene. Because none of the transcriptional regulators reported to date is smooth muscle cell- or myofibroblast-specific per se, the dynamic interplay among many factors interacting at specific sites along the promoter appears to be a signature feature of smooth muscle alpha-actin gene regulation in these cell types. Herein, the ability of the adenovirus E1A 12 S protein to bind and functionally inactivate specific cell regulatory factors has been exploited to identify several previously unknown coactivators of the mouse smooth muscle alpha-actin promoter in rodent fibroblasts and vascular smooth muscle cells. In transient cotransfection assays, ectopic expression of wild type E1A suppressed promoter activity in a dose- and cis-element-dependent manner. In asynchronous cells, N-terminal E1A mutants defective in CREB-binding protein (CBP) and p300 binding capacity exhibited markedly reduced inhibitory activity toward a smooth muscle alpha-actin promoter driven by a composite TEF-1-, SRF-, and Sp1/3-regulated enhancer. In synchronized cells, however, a more complex mutant E1A inhibitory pattern indicated that collaboration between CBP/p300 and the retinoblastoma family of pocket proteins was required to produce a fully functional enhancer. Cotransfection experiments conducted with Rb(-/-) fibroblasts demonstrated the necessity of pRB in augmenting smooth muscle alpha-actin enhancer/promoter activity. Physical interaction studies with the use of purified wild type and mutant E1A proteins confirmed that CBP, p300, and pRB were targets of E1A binding in nuclear extracts of vascular smooth muscle cells and/or fibroblasts. Collectively, these results suggest that a repertoire of E1A-interacting proteins, namely CBP/p300 and pRB, serve to integrate the activities of multiple trans-acting factors to control smooth muscle alpha-actin gene transcription in a cell type- and cell cycle-dependent manner.


Assuntos
Actinas/genética , Proteínas E1A de Adenovirus/metabolismo , Ciclo Celular/genética , Regulação da Expressão Gênica/genética , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Transcrição Gênica/genética , Actinas/metabolismo , Proteínas E1A de Adenovirus/química , Proteínas E1A de Adenovirus/genética , Animais , Linhagem Celular , Proteína p300 Associada a E1A , Elementos Facilitadores Genéticos/genética , Fibroblastos , Camundongos , Mutação/genética , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Repressoras/química , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reprodutibilidade dos Testes , Elementos de Resposta/genética , Proteína do Retinoblastoma/metabolismo , Transativadores/metabolismo
14.
Mol Biol Cell ; 15(10): 4532-43, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15282343

RESUMO

The mouse vascular smooth muscle alpha-actin (SMA) gene enhancer is activated in fibroblasts by transforming growth factor beta1 (TGFbeta1), a potent mediator of myofibroblast differentiation and wound healing. The SMA enhancer contains tandem sites for the Sp1 transcriptional activator protein and Puralpha and beta repressor proteins. We have examined dynamic interplay between these divergent proteins to identify checkpoints for possible control of myofibroblast differentiation during chronic inflammatory disease. A novel element in the SMA enhancer named SPUR was responsible for both basal and TGFbeta1-dependent transcriptional activation in fibroblasts and capable of binding Sp1 and Pur proteins. A novel Sp1:Pur:SPUR complex was dissociated when SMA enhancer activity was increased by TGFbeta1 or Smad protein overexpression. Physical association of Pur proteins with Smad2/3 was observed as was binding of Smads to an upstream enhancer region that undergoes DNA duplex unwinding in TGFbeta1-activated myofibroblasts. Purbeta repression of the SMA enhancer could not be relieved by TGFbeta1, whereas repression mediated by Puralpha was partially rescued by TGFbeta1 or overexpression of Smad proteins. Interplay between Pur repressor isoforms and Sp1 and Smad coactivators may regulate SMA enhancer output in TGFbeta1-activated myofibroblasts during episodes of wound repair and tissue remodeling.


Assuntos
Actinas , Proteínas de Ligação a DNA/metabolismo , Músculo Liso Vascular/fisiologia , Fator de Transcrição Sp1/metabolismo , Transativadores/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Células Cultivadas , DNA/metabolismo , Fibroblastos/citologia , Fibroblastos/fisiologia , Regulação da Expressão Gênica , Genes Reporter , Camundongos , Proteínas do Tecido Nervoso , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transdução de Sinais/fisiologia , Proteínas Smad , Fator de Crescimento Transformador beta1
16.
J Biol Chem ; 278(40): 38749-57, 2003 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-12874279

RESUMO

Plasticity of smooth muscle alpha-actin gene expression in fibroblasts and vascular smooth muscle cells is mediated by opposing effects of transcriptional activators and repressors. Among these factors, three single-stranded DNA-binding proteins, Puralpha, Purbeta, and MSY1, have been implicated as coregulators of a cryptic 5'-enhancer module. In this study, a molecular analysis of Purbeta, the least well characterized member of this group, was conducted. Southwestern and Northwestern blotting of purified Purbeta deletion mutants using smooth muscle alpha-actin-derived probes mapped the minimal single-stranded DNA/RNA-binding domain to a conserved region spanning amino acids 37-263. Quantitative binding assays indicated that the relative affinity and specificity of Purbeta for single-stranded DNA were influenced by purine/pyrimidine content; by non-conserved regions outside amino acids 37-263; and by cell-derived proteins, specifically MSY1. When overexpressed in A7r5 vascular smooth muscle cells, Purbeta (but not Puralpha) inhibited transcription of a smooth muscle-specific mouse alpha-actin promoter transgene. Structural domains required for Purbeta repressor activity included the minimal DNA-binding region and a C-terminal domain required for stabilizing high affinity protein and nucleic acid interactions. Purbeta inhibitory activity in transfected A7r5 cells was potentiated by MSY1, but antagonized by serum response factor, reinforcing the idea that interplay among activators and repressors may account for phenotypic changes in smooth muscle alpha-actin-expressing cell types.


Assuntos
Actinas/biossíntese , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/fisiologia , Músculo Liso Vascular/metabolismo , Transcrição Gênica , Actinas/genética , Animais , Northern Blotting , Southern Blotting , Western Blotting , Linhagem Celular , DNA/metabolismo , DNA Complementar/metabolismo , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Escherichia coli/metabolismo , Deleção de Genes , Genes Reporter , Immunoblotting , Camundongos , Mutação , Oligonucleotídeos/química , Fenótipo , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/metabolismo , Fator de Resposta Sérica/metabolismo , Relação Estrutura-Atividade , Fatores de Tempo , Transfecção , Transgenes
17.
J Biol Chem ; 277(39): 36433-42, 2002 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-12110667

RESUMO

The conversion of stromal fibroblasts into contractile myofibroblasts is an essential feature of the wound-healing response that is mediated by transforming growth factor beta1 (TGF-beta1) and accompanied by transient activation of the vascular smooth muscle alpha-actin (SmalphaA) gene. Multiple positive-regulatory elements were identified as essential mediators of basal SmalphaA enhancer activity in mouse AKR-2B stromal fibroblasts. Three of these elements bind transcriptional activating proteins of known identity in fibroblasts. A fourth site, shown previously to be susceptible to single-strand modifying agents in myofibroblasts, was additionally required for enhancer response to TGF-beta1. However, TGF-beta1 activation was not accompanied by a stoichiometric increase in protein binding to any known positive element in the SmalphaA enhancer. By using oligonucleotide affinity isolation, DNA-binding site competition, gel mobility shift assays, and protein overexpression in SL2 and COS7 cells, we demonstrate that the transcription factors Sp1 and Sp3 can stimulate SmalphaA enhancer activity. One of the sites that bind Sp1/3 corresponds to the region of the SmalphaA enhancer required for TGF-beta1 amplification. Additionally, the TGF-beta1 receptor-regulated Smad proteins, in particular Smad3, are rate-limiting for SmalphaA enhancer activation. Whereas Smad proteins collaborate with Sp1 in activating several stromal cell-associated promoters, they appear to operate independently from the Sp1/3 proteins in activating the SmalphaA enhancer. The identification of Sp and Smad proteins as essential, independent activators of the SmalphaA enhancer provides new insight into the poorly understood process of myofibroblast differentiation.


Assuntos
Actinas/biossíntese , Actinas/genética , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Endotélio Vascular/metabolismo , Elementos Facilitadores Genéticos , Músculo Liso/metabolismo , Músculos/citologia , Plicamicina/análogos & derivados , Fator de Transcrição Sp1/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Motivos de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Ligação Competitiva , Northern Blotting , Western Blotting , Células COS , Diferenciação Celular , Núcleo Celular/metabolismo , Células Cultivadas , Drosophila , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Genes Reporter , Humanos , Immunoblotting , Camundongos , Dados de Sequência Molecular , Mutação , Oligonucleotídeos/metabolismo , Plasmídeos/metabolismo , Plicamicina/farmacologia , Ligação Proteica , Ratos , Fator de Transcrição Sp3 , Transfecção , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1
18.
Cardiovasc Res ; 54(3): 539-48, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12031699

RESUMO

OBJECTIVE: Chronic rejection in cardiac allografts depletes vascular smooth muscle (VSM) alpha-actin from the coronary arterial smooth muscle bed while promoting its abnormal accumulation in cardiomyocytes and myofibroblasts. The objective was to determine if the newly discovered TEF1, MSY1, Puralpha and Purbeta VSM alpha-actin transcriptional reprogramming proteins (TRPs) were associated with development of chronic rejection histopathology in accepted murine cardiac allografts. METHODS: A mouse heterotopic cardiac transplant model was employed using H2 locus-mismatched mouse strains (DBA/2 or FVB/N to C57BL/6). Recipients were immunosuppressed to promote long-term allograft acceptance and emergence of chronic rejection. Explanted grafts and isolated heart cells were evaluated for changes in the DNA-binding activity and subcellular distribution of VSM alpha-actin transcriptional regulatory proteins. RESULTS: The DNA-binding activity of all four TRPs was high in the developing mouse ventricle, minimal in adult donor hearts and increased substantially within 30 days after transplantation. Immunohistologic analysis revealed nuclear localization of Purbeta and MSY1 particularly in fibrotic areas of the allograft myocardium demonstrating extravascular accumulation of VSM alpha-actin. Cardiomyocytes isolated from adult, non-transplanted mouse hearts not only exhibited less VSM alpha-actin expression and lower levels of TRPs compared to isolated cardiac fibroblasts or neonatal cardiomyocytes, but also contained a novel size variant of the MSY1 protein. CONCLUSION: Accumulation of TRPs in cardiac allografts, particularly within the fibroblast-enriched myocardial interstitium, was consistent with their potential role in VSM alpha-actin gene reprogramming, fibrosis and dysfunctional remodeling following transplant. These nuclear protein markers could help stage peri-transplant cellular events that precede formation of graft-destructive fibrosis and coronary vasculopathy during chronic rejection.


Assuntos
Actinas/genética , Genes Reguladores , Transplante de Coração , Músculo Liso Vascular/metabolismo , Miocárdio/metabolismo , Animais , Doença Crônica , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/análise , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , DNA/metabolismo , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/metabolismo , Feminino , Fibrose , Expressão Gênica , Marcadores Genéticos , Rejeição de Enxerto/metabolismo , Rejeição de Enxerto/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Modelos Animais , Miocárdio/química , Miocárdio/patologia , Proteínas do Tecido Nervoso , Fator 1 de Elongação de Peptídeos/análise , Fator 1 de Elongação de Peptídeos/metabolismo , Fatores de Tempo , Fatores de Transcrição , Transcrição Gênica , Transplante Homólogo
19.
J Biol Chem ; 277(10): 8682-92, 2002 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-11751932

RESUMO

An asymmetric polypurine-polypyrimidine cis-element located in the 5' region of the mouse vascular smooth muscle alpha-actin gene serves as a binding site for multiple proteins with specific affinity for either single- or double-stranded DNA. Here, we test the hypothesis that single-stranded DNA-binding proteins are responsible for preventing a cryptic MCAT enhancer centered within this element from cooperating with a nearby serum response factor-interacting CArG motif to trans-activate the minimal promoter in fibroblasts and smooth muscle cells. DNA binding studies revealed that the core MCAT sequence mediates binding of transcription enhancer factor-1 to the double-stranded polypurine-polypyrimidine element while flanking nucleotides account for interaction of Pur alpha and Pur beta with the purine-rich strand and MSY1 with the complementary pyrimidine-rich strand. Mutations that selectively impaired high affinity single-stranded DNA binding by fibroblast or smooth muscle cell-derived Pur alpha, Pur beta, and MSY1 in vitro, released the cryptic MCAT enhancer from repression in transfected cells. Additional experiments indicated that Pur alpha, Pur beta, and MSY1 also interact specifically, albeit weakly, with double-stranded DNA and with transcription enhancer factor-1. These results are consistent with two plausible models of cryptic MCAT enhancer regulation by Pur alpha, Pur beta, and MSY1 involving either competitive single-stranded DNA binding or masking of MCAT-bound transcription enhancer factor-1.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/metabolismo , Músculo Liso/citologia , Proteínas Nucleares , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Animais , Sequência de Bases , Biotinilação , Linhagem Celular , Células Cultivadas , DNA/metabolismo , DNA de Cadeia Simples/metabolismo , Elementos Facilitadores Genéticos , Epitopos , Genes Reporter , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Músculo Liso/metabolismo , Mutação , Proteínas do Tecido Nervoso , Testes de Precipitina , Ligação Proteica , Coelhos , Ratos , Fatores de Transcrição de Domínio TEA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...