Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Gastrointest Liver Physiol ; 281(5): G1271-8, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11668036

RESUMO

Early growth response-1 (Egr-1) is a transcription factor that couples short-term changes in the extracellular milieu to long-term changes in gene expression. Under in vitro conditions, the Egr-1 gene has been shown to respond to many extracellular signals. In most cases, these findings have not been extended to the in vivo setting. The goal of the present study was to explore the role of epidermal growth factor (EGF) in mediating Egr-1 expression in hepatocytes under both in vitro and in vivo conditions. In HepG2 cells, Egr-1 protein and mRNA were upregulated in the presence of EGF. In stable transfections of HepG2 cells, a 1,200-bp Egr-1 promoter contained information for EGF response via a protein kinase C-independent, mitogen-activated protein kinase-dependent signaling pathway. A promoter region containing the two most proximal serum response elements was sufficient to transduce the EGF signal. In transgenic mice that carry the Egr-1 promoter coupled to the LacZ reporter gene, systemic delivery of EGF by intraperitoneal injection resulted in an induction of the endogenous Egr-1 gene and the Egr-1-lacZ transgene in hepatocytes. Together, these results suggest that the 1,200-bp promoter contains information for EGF response in hepatocytes both in vitro and in intact animals.


Assuntos
Proteínas de Ligação a DNA/genética , Fator de Crescimento Epidérmico/farmacologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/fisiologia , Proteínas Imediatamente Precoces , Regiões Promotoras Genéticas/fisiologia , Fatores de Transcrição/genética , Animais , Células Cultivadas , Proteína 1 de Resposta de Crescimento Precoce , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos/genética , Transgenes/efeitos dos fármacos , Transgenes/fisiologia
2.
Proc Natl Acad Sci U S A ; 98(16): 9336-41, 2001 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-11481491

RESUMO

Therapy for ischemic heart disease has been directed traditionally at limiting cell necrosis. We determined by genome profiling whether ischemic myocardium can trigger a genetic program promoting cardiac cell survival, which would be a novel and potentially equally important mechanism of salvage. Although cardiac genomics is usually performed in rodents, we used a swine model of ischemia/reperfusion followed by ventricular dysfunction (stunning), which more closely resembles clinical conditions. Gene expression profiles were compared by subtractive hybridization between ischemic and normal tissue of the same hearts. About one-third (23/74) of the nuclear-encoded genes that were up-regulated in ischemic myocardium participate in survival mechanisms (inhibition of apoptosis, cytoprotection, cell growth, and stimulation of translation). The specificity of this response was confirmed by Northern blot and quantitative PCR. Unexpectedly, this program also included genes not previously described in cardiomyocytes. Up-regulation of survival genes was more profound in subendocardium over subepicardium, reflecting that this response in stunned myocardium was proportional to the severity of the ischemic insult. Thus, in a swine model that recapitulates human heart disease, nonlethal ischemia activates a genomic program of cell survival that relates to the time course of myocardial stunning and differs transmurally in relation to ischemic stress, which induced the stunning. Understanding the genes up-regulated during myocardial stunning, including those not previously described in the heart, and developing strategies that activate this program may open new avenues for therapy in ischemic heart disease.


Assuntos
Sobrevivência Celular/genética , Isquemia Miocárdica/patologia , Miocárdio/patologia , Animais , Apoptose , DNA Complementar , Feminino , Perfilação da Expressão Gênica , Hibridização In Situ , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos
4.
FASEB J ; 14(13): 1870-2, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11023970

RESUMO

Egr-1 is an immediate early gene that couples short-term changes in the extracellular milieu to long-term changes in gene expression. Under in vitro conditions, the Egr-1 gene is expressed in many cell types and is induced by a wide variety of extracellular signals. The mechanisms by which the Egr-1 gene is regulated in vivo remain poorly understood. In this study, we have generated transgenic mice with a construct containing 1200 bp of the mouse Egr-1 promoter coupled to nuclear localized LacZ. In multiple independent lines of mice, reporter gene expression was detected in subsets of endothelial cells, vascular smooth-muscle cells, cardiomyocytes, neurons, and hepatocytes. This pattern closely resembled that of the endogenous gene. After partial hepatectomy, reporter gene activity was upregulated between two- and fivefold in regenerating livers. Taken together, these findings suggest that the Egr-1 promoter contains information for appropriate spatial and temporal expression in vivo.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas Imediatamente Precoces/genética , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Animais , Proteína 1 de Resposta de Crescimento Precoce , Regulação da Expressão Gênica , Genes Reporter , Hepatectomia , Óperon Lac , Camundongos , Camundongos Transgênicos , Distribuição Tecidual
6.
J Biol Chem ; 275(47): 36653-8, 2000 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-10973958

RESUMO

Activated macrophages are critical cellular participants in inflammatory disease states. Transforming growth factor (TGF)-beta1 is a growth factor with pleiotropic effects including inhibition of immune cell activation. Although the pathway of gene activation by TGF-beta1 via Smad proteins has recently been elucidated, suppression of gene expression by TGF-beta1 remains poorly understood. We found that of Smad1-Smad7, Smad3 alone was able to inhibit expression of markers of macrophage activation (inducible nitric-oxide synthase and matrix metalloproteinase-12) following lipopolysaccharide treatment in gene reporter assays. Transient and constitutive overexpression of a dominant negative Smad3 opposed the inhibitory effect of TGF-beta1. Domain swapping experiments suggest that both the Smad MH-1 and MH-2 domains are required for inhibition. Mutation of a critical amino acid residue required for DNA binding in the MH-1 of Smad3 (R74A) resulted in the loss of inhibition. Transient overexpression of p300, an interactor of the Smad MH-2 domain, partially alleviated the inhibition by TGF-beta1/Smad3, suggesting that inhibition of gene expression may be due to increased competition for limiting amounts of this coactivator. Our results have implications for the understanding of gene suppression by TGF-beta1 and for the regulation of activated macrophages by TGF-beta1.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Transativadores/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Biomarcadores , Linhagem Celular , Lipopolissacarídeos/farmacologia , Metaloproteinase 12 da Matriz , Metaloendopeptidases/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Proteína Smad3 , Relação Estrutura-Atividade , Transcrição Gênica/efeitos dos fármacos , Regulação para Cima
7.
J Exp Med ; 192(5): 695-704, 2000 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-10974035

RESUMO

Transforming growth factor (TGF)-beta(1) is a pleiotropic cytokine/growth factor that is thought to play a critical role in the modulation of inflammatory events. We demonstrate that exogenous TGF-beta(1) can inhibit the expression of the proinflammatory adhesion molecule, E-selectin, in vascular endothelium exposed to inflammatory stimuli both in vitro and in vivo. This inhibitory effect occurs at the level of transcription of the E-selectin gene and is dependent on the action of Smad proteins, a class of intracellular signaling proteins involved in mediating the cellular effects of TGF-beta(1). Furthermore, we demonstrate that these Smad-mediated effects in endothelial cells result from a novel competitive interaction between Smad proteins activated by TGF-beta(1) and nuclear factor kappaB (NFkappaB) proteins activated by inflammatory stimuli (such as cytokines or bacterial lipopolysaccharide) that is mediated by the transcriptional coactivator cyclic AMP response element-binding protein (CREB)-binding protein (CBP). Augmentation of the limited amount of CBP present in endothelial cells (via overexpression) or selective disruption of Smad-CBP interactions (via a dominant negative strategy) effectively antagonizes the ability of TGF-beta(1) to block proinflammatory E-selectin expression. These data thus demonstrate a novel mechanism of interaction between TGF-beta(1)-regulated Smad proteins and NFkappaB proteins regulated by inflammatory stimuli in vascular endothelial cells. This type of signaling mechanism may play an important role in the immunomodulatory actions of this cytokine/growth factor in the cardiovascular system.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Selectina E/genética , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , NF-kappa B/fisiologia , Transativadores/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Endotélio Vascular/citologia , Humanos , Interleucina-1/farmacologia , Ratos , Proteína Smad2 , Proteína Smad3
8.
Ann N Y Acad Sci ; 902: 230-9; discussion 239-40, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10865843

RESUMO

Phenotypic modulation of endothelium to a dysfunctional state contributes to the pathogenesis of cardiovascular diseases such as atherosclerosis. The localization of atherosclerotic lesions to arterial geometries associated with disturbed flow patterns suggests an important role for local hemodynamic forces in atherogenesis. There is increasing evidence that the vascular endothelium, which is directly exposed to various fluid mechanical forces generated by pulsatile blood flow, can discriminate among these stimuli and transduce them into genetic regulatory events. At the level of individual genes, this regulation is accomplished via the binding of certain transcription factors, such as NF kappa B and Egr-1, to shear-stress response elements (SSREs) that are present in the promoters of biomechanically inducible genes. At the level of multiple genes, distinct patterns of up- and downregulation appear to be elicited by exposure to steady laminar shear stresses versus comparable levels of non-laminar (e.g., turbulent) shear stresses or cytokine stimulation (e.g., IL-1 beta). Certain genes upregulated by steady laminar shear stress stimulation (such as eNOS, COX-2, and Mn-SOD) support vasoprotective (i.e., anti-inflammatory, anti-thrombotic, anti-oxidant) functions in the endothelium. We hypothesize that the selective and sustained expression of these and related "atheroprotective genes" in the endothelial lining of lesion-protected areas represents a mechanism whereby hemodynamic forces can influence lesion formation and progression.


Assuntos
Artérias/fisiopatologia , Arteriosclerose/fisiopatologia , Endotélio Vascular/fisiopatologia , Hemodinâmica , Animais , Arteriosclerose/genética , Fenômenos Biomecânicos , Regulação da Expressão Gênica , Humanos , Estresse Mecânico
9.
J Biol Chem ; 275(33): 25766-73, 2000 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-10825169

RESUMO

Matrix metalloproteinases (MMP) have been identified in vulnerable areas of atherosclerotic plaques and may contribute to plaque instability through extracellular matrix degradation. Human metalloelastase (MMP-12) is a macrophage-specific MMP with broad substrate specificity and is capable of degrading proteins found in the extracellular matrix of atheromas. Despite its potential importance, little is known about the regulation of MMP-12 expression in the context of atherosclerosis. In this study, we report that in human peripheral blood-derived macrophages, MMP-12 mRNA was markedly up-regulated by several pro-atherosclerotic cytokines and growth factors including interleukin-1beta, tumor necrosis factor-alpha, macrophage colony-stimulating factor, vascular endothelial growth factor, and platelet-derived growth factor-BB. In contrast, the pleiotropic anti-inflammatory growth factor transforming growth factor-beta1 (TGF-beta1) inhibited cytokine-mediated induction of MMP-12 mRNA, protein, and enzymatic activity. Analyses of MMP-12 promoter through transient transfections and electrophoretic mobility shift assays indicated that both its induction by cytokines and its inhibition by TGF-beta1 depended on signaling through an AP-1 site at -81 base pairs. Moreover, the inhibitory effect of TGF-beta1 on MMP-12 was dependent on Smad3. Taken together, MMP-12 is induced by several factors implicated in atherosclerosis. The inhibition of MMP-12 expression by TGF-beta1 suggests that TGF-beta1, acting via Smad3, may promote plaque stability.


Assuntos
Citocinas/metabolismo , Macrófagos/enzimologia , Metaloendopeptidases/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Arteriosclerose/metabolismo , Sítios de Ligação , Western Blotting , Linhagem Celular , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Indução Enzimática , Genes Dominantes , Genes Reporter , Humanos , Metaloproteinase 12 da Matriz , Metaloendopeptidases/sangue , Metaloendopeptidases/genética , Camundongos , Monócitos/enzimologia , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Proteína Smad3 , Fatores de Tempo , Transativadores/metabolismo , Fator de Transcrição AP-1/metabolismo , Transfecção , Regulação para Cima
10.
Trends Cardiovasc Med ; 10(3): 132-7, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11428000

RESUMO

Transforming growth factor beta-1 is the prototypical member of a class of growth factors whose actions have been strongly implicated in a number of pathophysiologic processes including chronic vascular diseases such as atherosclerosis and hypertension. One of the hall-marks of this class of growth factors is the diverse nature of their actions; a characteristic that is thought to arise from the fact that the effects of these factors are very dependent upon the particular cellular context in which they operate. There has been substantial progress in understanding the molecular signaling mechanisms utilized by these factors. These findings are beginning to provide a mechanistic framework with which to understand the complex and pleiotropic actions of these factors on cells and tissues of the cardiovascular system.


Assuntos
Doenças Cardiovasculares/fisiopatologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Humanos , Transdução de Sinais
11.
J Biol Chem ; 274(13): 8797-805, 1999 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-10085121

RESUMO

Smad proteins are essential components of the intracellular signaling pathways utilized by members of the transforming growth factor-beta (TGF-beta) superfamily of growth factors. Certain Smad proteins (e.g. Smad1, -2, and -3) can act as regulated transcriptional activators, a process that involves phosphorylation of these proteins by activated TGF-beta superfamily receptors. We demonstrate that the intracellular kinase mitogen-activated protein kinase kinase kinase-1 (MEKK-1), an upstream activator of the stress-activated protein kinase/c-Jun N-terminal kinase pathway, can participate in Smad2-dependent transcriptional events in cultured endothelial cells. A constitutively active form of MEKK-1 but not mitogen-activated protein kinase kinase-1 (MEK-1) or TGF-beta-activated kinase-1, two distinct intracellular kinases, can specifically activate a Gal4-Smad2 fusion protein, and this effect correlates with an increase in the phosphorylation state of the Smad2 protein. These effects do not require the presence of the C-terminal SSXS motif of Smad2 that is the site of TGF-beta type 1 receptor-mediated phosphorylation. Activation of Smad2 by active MEKK-1 results in enhanced Smad2-Smad4 interactions, nuclear localization of Smad2 and Smad4, and the stimulation of Smad protein-transcriptional coactivator interactions in endothelial cells. Overexpression of Smad7 can inhibit the MEKK-1-mediated stimulation of Smad2 transcriptional activity. A physiological level of fluid shear stress, a known activator of endogenous MEKK-1 activity in endothelial cells, can stimulate Smad2-mediated transcriptional activity. These data demonstrate a novel mechanism for activation of Smad protein-mediated signaling in endothelial cells and suggest that Smad2 may act as an integrator of diverse stimuli in these cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transativadores/metabolismo , Ativação Transcricional/genética , Animais , Bovinos , Células Cultivadas , Endotélio Vascular/metabolismo , Genes Reporter/genética , Imuno-Histoquímica , MAP Quinase Quinase 1 , MAP Quinase Quinase 4 , Fosforilação , Regiões Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Transdução de Sinais , Proteína Smad2 , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta/genética
12.
Mol Med Today ; 5(1): 40-6, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10088131

RESUMO

Vascular endothelium, the cellular monolayer lining the entire cardiovascular system, is exposed to a variety of biochemical and biomechanical stimuli. Fluid shear stresses generated by blood flow in the vasculature can profoundly influence the phenotype of the endothelium by regulating the activity of certain flow-sensitive proteins (for example, enzymes), as well as by modulating gene expression. The finding that specific fluid mechanical forces can alter endothelial structure and function has provided a framework for a mechanistic understanding of flow-dependent processes, ranging from vascular remodeling in response to hemodynamic changes, to the initiation and localization of chronic vascular diseases such as atherosclerosis.


Assuntos
Arteriosclerose/genética , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica/fisiologia , Animais , Arteriosclerose/fisiopatologia , Velocidade do Fluxo Sanguíneo/genética , Endotélio Vascular/fisiologia , Hemodinâmica/genética , Humanos , Fenótipo , Estresse Mecânico
13.
Circulation ; 98(22): 2396-403, 1998 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9832484

RESUMO

BACKGROUND: biomechanical forces generated by blood flow within the cardiovascular system have been proposed as important modulators of regional endothelial phenotype and function. This process is thought to involve the regulation of vascular gene expression by physiological fluid mechanical stimuli such as fluid shear stresses. METHODS AND RESULTS: We demonstrate sustained upregulation of a recently identified gene encoding a human prostaglandin transporter (hPGT) in cultured human vascular endothelium exposed to a physiological fluid mechanical stimulus in vitro. This biomechanical induction is selective in that steady laminar shear stress is sufficient to upregulate the hPGT gene at the level of transcriptional activation, whereas a comparable level of turbulent shear stress (a nonphysiological stimulus) is not. Various biochemical stimuli, such as bacterial endotoxin and the inflammatory cytokines recombinant human interleukin 1beta cytokines (rhIL-1beta) and tumor necrosis factor-alpha (TNF-alpha), did not significantly induce hPGT. Using a specific antiserum to hPGT, we demonstrate endothelial expression within the arterial vasculature and the microcirculation of highly vascularized tissues such as the heart. CONCLUSIONS: Our results identify hPGT as an inducible gene in vascular endothelium and suggest that biomechanical stimuli generated by blood flow in vivo may be important determinants of hPGT expression. Furthermore, this demonstration of regulated endothelial expression of hPGT implicates this molecule in the regional metabolism of prostanoids within the cardiovascular system.


Assuntos
Antiporters/genética , Antiporters/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Antiporters/biossíntese , Células Cultivadas , DNA Complementar/análise , Proteínas de Ligação a DNA/biossíntese , Endotélio Vascular/química , Regulação da Expressão Gênica , Biblioteca Gênica , Humanos , Recém-Nascido , Transportadores de Ânions Orgânicos , Peptídeos/análise , RNA Mensageiro/análise , Estresse Mecânico , Regulação para Cima
14.
Proc Natl Acad Sci U S A ; 95(16): 9506-11, 1998 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-9689110

RESUMO

The transforming growth factor-beta (TGF-beta) superfamily of growth factors and cytokines has been implicated in a variety of physiological and developmental processes within the cardiovascular system. Smad proteins are a recently described family of intracellular signaling proteins that transduce signals in response to TGF-beta superfamily ligands. We demonstrate by both a mammalian two-hybrid and a biochemical approach that human Smad2 and Smad4, two essential Smad proteins involved in mediating TGF-beta transcriptional responses in endothelial and other cell types, can functionally interact with the transcriptional coactivator CREB binding protein (CBP). This interaction is specific in that it requires ligand (TGF-beta) activation and is mediated by the transcriptional activation domains of the Smad proteins. A closely related, but distinct endothelial-expressed Smad protein, Smad7, which does not activate transcription in endothelial cells, does not interact with CBP. Furthermore, Smad2,4-CBP interactions involve the COOH terminus of CBP, a region that interacts with other regulated transcription factors such as certain signal transduction and transcription proteins and nuclear receptors. Smad-CBP interactions are required for Smad-dependent TGF-beta-induced transcriptional responses in endothelial cells, as evidenced by inhibition with overexpressed 12S E1A protein and reversal of this inhibition with exogenous CBP. This report demonstrates a functional interaction between Smad proteins and an essential component of the mammalian transcriptional apparatus (CBP) and extends our insight into how Smad proteins may regulate transcriptional responses in many cell types. Thus, functional Smad-coactivator interactions may be an important locus of signal integration in endothelial cells.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Endotélio Vascular/metabolismo , Transativadores/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo , Animais , Sequência de Bases , Bovinos , Células Cultivadas , Primers do DNA , Endotélio Vascular/citologia , Humanos , Proteína Smad2 , Proteína Smad4
17.
Proc Natl Acad Sci U S A ; 94(17): 9314-9, 1997 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-9256479

RESUMO

Vascular endothelium is an important transducer and integrator of both humoral and biomechanical stimuli within the cardiovascular system. Utilizing a differential display approach, we have identified two genes, Smad6 and Smad7, encoding members of the MAD-related family of molecules, selectively induced in cultured human vascular endothelial cells by steady laminar shear stress, a physiologic fluid mechanical stimulus. MAD-related proteins are a recently identified family of intracellular proteins that are thought to be essential components in the signaling pathways of the serine/threonine kinase receptors of the transforming growth factor beta superfamily. Smad6 and Smad7 possess unique structural features (compared with previously described MADs), and they can physically interact with each other, and, in the case of Smad6, with other known human MAD species, in endothelial cells. Transient expression of Smad6 or Smad7 in vascular endothelial cells inhibits the activation of a transfected reporter gene in response to both TGF-beta and fluid mechanical stimulation. Both Smad6 and Smad7 exhibit a selective pattern of expression in human vascular endothelium in vivo as detected by immunohistochemistry and in situ hybridization. Thus, Smad6 and Smad7 constitute a novel class of MAD-related proteins, termed vascular MADs, that are induced by fluid mechanical forces and can modulate gene expression in response to both humoral and biomechanical stimulation in vascular endothelium.


Assuntos
Proteínas de Ligação a DNA/genética , Endotélio Vascular/fisiologia , Expressão Gênica , Transativadores , Sequência de Aminoácidos , Células Cultivadas , Proteínas de Ligação a DNA/biossíntese , Humanos , Imuno-Histoquímica , Hibridização In Situ , Dados de Sequência Molecular , Alinhamento de Sequência , Transdução de Sinais/genética , Proteína Smad6 , Proteína Smad7 , Estresse Mecânico
18.
J Clin Invest ; 99(12): 2941-9, 1997 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9185518

RESUMO

In vascular endothelium, the electroneutral Na-K-Cl cotransport system is thought to function in the maintenance of a selective permeability barrier in certain vascular beds (e.g., brain), as well as in the preservation of endothelial homeostasis in the face of fluctuating osmotic conditions that may accompany certain pathophysiological conditions (e.g., diabetes mellitus). Here we demonstrate that the gene encoding the bumetanide-sensitive cotransporter BSC2, one of the two major isoforms of Na-K-Cl cotransporters present in mammalian cells, can be differentially regulated by inflammatory cytokines and fluid mechanical forces in cultured endothelium. Interleukin-1beta and tumor necrosis factor-alpha significantly upregulate expression of BSC2 mRNA and protein in human umbilical vein endothelial cells, a response that is inhibited by pretreatment with interferon-gamma. Steady laminar fluid shear stress, at a physiologic magnitude (10 dyn/cm2), is also able to induce and maintain elevated expression of BSC2 in cultured human umbilical vein endothelial cells, while a comparable time-averaged magnitude of turbulent fluid shear stress is not. In vivo, BSC2 mRNA is upregulated after intraperitoneal administration of bacterial endotoxin (LPS) in murine lung and kidney, but not in cardiac tissue. These results provide the first experimental evidence that the BSC2 gene can be selectively regulated by different inflammatory cytokine and fluid mechanical stimuli in endothelium, and support a role for BSC2 in vascular homeostasis and inflammation.


Assuntos
Bumetanida/farmacologia , Proteínas de Transporte/genética , Citocinas/farmacologia , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica , Animais , Fenômenos Biomecânicos , Linhagem Celular , Humanos , Interferon gama/farmacologia , Interleucina-1/farmacologia , Rim/metabolismo , Lipopolissacarídeos/farmacologia , Pulmão/metabolismo , Camundongos , RNA Mensageiro/metabolismo , Reologia , Simportadores de Cloreto de Sódio-Potássio , Fator de Necrose Tumoral alfa/farmacologia , Veias Umbilicais
19.
Cell ; 89(7): 1165-73, 1997 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-9215638

RESUMO

TGFbeta signaling is initiated when the type I receptor phosphorylates the MAD-related protein, Smad2, on C-terminal serine residues. This leads to Smad2 association with Smad4, translocation to the nucleus, and regulation of transcriptional responses. Here we demonstrate that Smad7 is an inhibitor of TGFbeta signaling. Smad7 prevents TGFbeta-dependent formation of Smad2/Smad4 complexes and inhibits the nuclear accumulation of Smad2. Smad7 interacts stably with the activated TGFbeta type I receptor, thereby blocking the association, phosphorylation, and activation of Smad2. Furthermore, mutations in Smad7 that interfere with receptor binding disrupt its inhibitory activity. These studies thus define a novel function for MAD-related proteins as intracellular antagonists of the type I kinase domain of TGFbeta family receptors.


Assuntos
Proteínas de Transporte/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/fisiologia , Transativadores , Fator de Crescimento Transformador beta/fisiologia , Sequência de Aminoácidos , Animais , Células COS , Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endotélio Vascular/química , Endotélio Vascular/citologia , Humanos , Neoplasias Hepáticas , Dados de Sequência Molecular , Fosforilação , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2 , Células Tumorais Cultivadas , Veias Umbilicais/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...