Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Life (Basel) ; 11(12)2021 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-34947926

RESUMO

Thoracic aortic aneurysm (TAA) is a life-threatening condition associated with high mortality, in which the aortic wall is deformed due to congenital or age-associated pathological changes. The mechanisms of TAA development remain to be studied in detail, and are the subject of active research. In this review, we describe the morphological changes of the aortic wall in TAA. We outline the genetic disorders associated with aortic enlargement and discuss the potential role of mitochondrial pathology, in particular mitochondrial DNA heteroplasmy, in the disease pathogenesis.

2.
J Thromb Haemost ; 7(8): 1401-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19426286

RESUMO

BACKGROUND: Heme oxygenase-1 (HO-1), a heme degradation enzyme with multiple vasoprotective functions, is systemically induced in pathophysiological states associated with oxidative stress. OBJECTIVES: To evaluate the impact of systemic HO-1 expression on circulating endothelial progenitor cells (EPCs) and re-endothelialization after vascular injury in an animal model. METHODS: Mice received an intravenous (i.v.) injection of the adenovirus-bearing HO-1 gene (Adv-HO-1). The serum levels of vascular endothelial growth factor (VEGF) and stromal cell-derived factor-1 (SDF-1) were determined by ELISA and gene expression examined by quantitative real-time PCR. Circulating EPCs were characterized by flow cytometry and in vitro culture. EPC recruitment and re-endothelialization in injured arteries were assessed in mice receiving GFP+-bone marrow transplantation and guide wire-induced carotid injury. The effect of carbon monoxide (CO), a byproduct from heme degradation by HO-1, was assessed by exposing mice to 250 p.p.m. CO for 2 h day(-1). RESULTS: Systemic HO-1 induction led to elevated serum levels of VEGF and SDF-1 and an increase in circulating EPCs. The re-endothelialization of denuded vessels was accelerated in mice with systemic HO-1 overexpression. A further experiment demonstrated that both EPC mobilization and re-endothelialization were significantly attenuated in mice with HO-1 deficiency. The increase in EPC mobilization and enhanced re-endothelialization was also observed in mice exposed to CO prior to carotid injury. The CO-mediated effect was associated with an increase in circulating SDF-1 but not VEGF. CONCLUSION: These findings support a vital role of HO-1 and its reaction byproduct, CO, in vascular repair through enhancing EPC mobilization.


Assuntos
Vasos Sanguíneos/lesões , Monóxido de Carbono/farmacologia , Células Endoteliais/citologia , Endotélio Vascular/efeitos dos fármacos , Heme Oxigenase-1/farmacologia , Animais , Movimento Celular , Quimiocina CXCL12/sangue , Quimiocina CXCL12/genética , Endotélio Vascular/citologia , Heme Oxigenase-1/administração & dosagem , Camundongos , Regeneração/efeitos dos fármacos , Células-Tronco/fisiologia , Fator A de Crescimento do Endotélio Vascular/sangue , Fator A de Crescimento do Endotélio Vascular/genética
3.
FASEB J ; 15(14): 2733-5, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11606480

RESUMO

Atherosclerosis is an inflammatory disease process associated with elevated levels of plasma cholesterol, especially low-density lipoproteins. The latter become trapped within the arterial wall and are oxidized and taken up by macrophages to form foam cells. This process is an initiating event for atherosclerosis. Fatty acid binding proteins (FABP) are involved in fatty acid metabolism and cellular lipid transport, and adipocyte FABP (aP2) is also expressed in macrophages. We recently generated mice lacking both apolipoprotein (Apo)E and aP2 (ApoE-/-aP2-/-) and found that these mice, compared with ApoE-/- mice, developed markedly smaller atherosclerotic lesions that contained fewer macrophages. Here we investigated the mechanism(s) responsible for this prevention of atherosclerotic lesion formation. Bone marrow transplantations were performed in ApoE-/- mice, receiving cells from either ApoE-/- or ApoE-/-aP2-/- mice. The lack of aP2 in donor marrow cells led to the development of smaller (5.5-fold) atherosclerotic lesions in the recipient mice. No differences were found in plasma cholesterol, glucose, or insulin levels between recipients of bone marrow cells from ApoE-/- or ApoE-/-aP2-/- mice. However, the expression of chemoattractant and inflammatory cytokines was decreased in macrophages from ApoE-/-aP2-/- mice compared with ApoE-/- mice, which may contribute to the decrease in atherosclerotic lesion formation. Taken together, we demonstrate the importance of macrophage aP2 in the development of atherosclerotic lesions.


Assuntos
Arteriosclerose/patologia , Proteínas de Transporte/fisiologia , Hipercolesterolemia/complicações , Macrófagos/metabolismo , Proteínas de Neoplasias , Proteínas do Tecido Nervoso , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Arteriosclerose/sangue , Arteriosclerose/complicações , Glicemia/metabolismo , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Proteínas de Transporte/genética , Colesterol/sangue , Citocinas/sangue , Proteína 7 de Ligação a Ácidos Graxos , Proteínas de Ligação a Ácido Graxo , Insulina/sangue , Camundongos , Camundongos Knockout , Óxido Nítrico/sangue
4.
J Biol Chem ; 276(50): 47658-63, 2001 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-11606591

RESUMO

The phenotypic modulation of vascular smooth muscle cells (VSMC) plays a central role in the pathogenesis of arteriosclerosis. Aortic preferentially expressed gene-1 (APEG-1), a VSMC-specific gene, is expressed highly in differentiated but not in dedifferentiated VSMC. Previously, we identified an E-box element in the mouse APEG-1 proximal promoter, which is essential for VSMC reporter activity. In this study, we investigated the role of upstream stimulatory factors (USF) in the regulation of APEG-1 transcription via this E-box element. By electrophoretic mobility shift assays, recombinant USF1 and USF2 homo- and heterodimers bound specifically to the APEG-1 E-box. Nuclear extracts prepared from primary cultures of rat aortic smooth muscle cells exhibited specific USF1 and USF2 binding to the APEG-1 E-box. To investigate the binding properties of USF during VSMC differentiation, nuclear extracts were prepared from the neural crest cell line, MONC-1, which differentiates into VSMC in culture. Maximal USF1 and USF2 protein levels and binding to the APEG-1 E-box occurred 3 h after the differentiation of MONC-1 cells was initiated. Co-transfection experiments demonstrated that dominant negative USF repressed APEG-1 promoter activity, and USF1, but not USF2, transactivated the APEG-1 promoter. Our studies demonstrate that USF factors contribute to the regulation of APEG-1 expression and may influence the differentiation of VSMC.


Assuntos
Aorta/metabolismo , Proteínas de Ligação a DNA , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Aorta Torácica/citologia , Aorta Torácica/metabolismo , Northern Blotting , Western Blotting , Diferenciação Celular , Núcleo Celular/metabolismo , Células Cultivadas , Dimerização , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Genes Dominantes , Luciferases/metabolismo , Masculino , Músculo Liso/citologia , Fenótipo , Regiões Promotoras Genéticas , Ligação Proteica , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Transcrição Gênica , Transfecção , Fatores Estimuladores Upstream
5.
Mol Cell Biol ; 21(15): 5256-61, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11438679

RESUMO

Aortic carboxypeptidase-like protein (ACLP) is a member of a diverse group of proteins that contain a domain with similarity to that of the Dictyostelium discoideum protein discoidin I. The discoidin domain has been identified in mammalian milk fat globule membrane proteins, blood coagulation factors, and receptor tyrosine kinases, where it may facilitate cell aggregation, adhesion, or cell-cell recognition. Here we show that ACLP is a secreted protein that associates with the extracellular matrix (ECM). During mouse embryogenesis, ACLP is abundantly expressed in the ECM of collagen-rich tissues, including the vasculature, dermis, and the developing skeleton. We deleted the ACLP gene in mice by homologous recombination. The majority of ACLP(-/-) mice die perinatally due to gastroschisis, a severe disruption of the anterior abdominal wall and herniation of the abdominal organs. ACLP(-/-) mice that survived to adulthood developed nonhealing skin wounds. Following injury by a dermal punch biopsy, ACLP(-/-) mice exhibited deficient wound healing compared with controls. In addition, dermal fibroblasts isolated from ACLP(-/-) 18.5-day-postconception embryos exhibited a reduced proliferative capacity compared with wild-type cells. These results indicate that ACLP is an ECM protein that is essential for embryonic development and dermal wound healing processes.


Assuntos
Músculos Abdominais/embriologia , Proteínas/genética , Proteínas/fisiologia , Cicatrização , Animais , Desenvolvimento Ósseo , Carboxipeptidases , Adesão Celular , Agregação Celular , Divisão Celular , Células Cultivadas , Clonagem Molecular , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Imuno-Histoquímica , Camundongos , Microscopia de Fluorescência , Modelos Genéticos , Músculo Liso/citologia , Mutagênese Sítio-Dirigida , Fenótipo , Estrutura Terciária de Proteína , Recombinação Genética , Proteínas Repressoras , Pele/metabolismo , Pele/patologia , Frações Subcelulares , Fatores de Tempo
6.
Circ Res ; 89(2): 168-73, 2001 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-11463724

RESUMO

Heme oxygenase (HO)-1 degrades the pro-oxidant heme and generates carbon monoxide and antioxidant bilirubin. We have previously shown that in response to hypoxia, HO-1-null mice develop infarcts in the right ventricle of their hearts and that their cardiomyocytes are damaged by oxidative stress. To test whether HO-1 protects against oxidative injury in the heart, we generated cardiac-specific transgenic mice overexpressing different levels of HO-1. By use of a Langendorff preparation, hearts from transgenic mice showed improved recovery of contractile performance during reperfusion after ischemia in an HO-1 dose-dependent manner. In vivo, myocardial ischemia and reperfusion experiments showed that infarct size was only 14.7% of the area at risk in transgenic mice compared with 56.5% in wild-type mice. Hearts from these transgenic animals had reduced inflammatory cell infiltration and oxidative damage. Our data demonstrate that overexpression of HO-1 in the cardiomyocyte protects against ischemia and reperfusion injury, thus improving the recovery of cardiac function.


Assuntos
Heme Oxigenase (Desciclizante)/genética , Isquemia Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/enzimologia , Animais , Regulação Enzimológica da Expressão Gênica/fisiologia , Genótipo , Coração/fisiopatologia , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1 , Humanos , Proteínas de Membrana , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle , Isquemia Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/patologia
7.
Nat Med ; 7(6): 693-8, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11385506

RESUMO

Heme oxygenase (HO-1, encoded by Hmox1) is an inducible protein activated in systemic inflammatory conditions by oxidant stress. Vascular injury is characterized by a local reparative process with inflammatory components, indicating a potential protective role for HO-1 in arterial wound repair. Here we report that HO-1 directly reduces vasoconstriction and inhibits cell proliferation during vascular injury. Expression of HO-1 in arteries stimulated vascular relaxation, mediated by guanylate cyclase and cGMP, independent of nitric oxide. The unexpected effects of HO-1 on vascular smooth muscle cell growth were mediated by cell-cycle arrest involving p21Cip1. HO-1 reduced the proliferative response to vascular injury in vivo; expression of HO-1 in pig arteries inhibited lesion formation and Hmox1-/- mice produced hyperplastic arteries compared with controls. Induction of the HO-1 pathway moderates the severity of vascular injury by at least two adaptive mechanisms independent of nitric oxide, and is a potential therapeutic target for diseases of the vasculature.


Assuntos
Artérias/fisiologia , Heme Oxigenase (Desciclizante)/metabolismo , Músculo Liso Vascular/citologia , Vasoconstrição , Animais , Artérias/enzimologia , Artérias/lesões , Ciclo Celular/fisiologia , Células Cultivadas , Meios de Cultura Livres de Soro , GMP Cíclico/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ciclinas/metabolismo , Indução Enzimática , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Deleção de Genes , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Heme Oxigenase-1 , Proteínas de Membrana , Camundongos , Músculo Liso Vascular/fisiologia , Protoporfirinas/farmacologia , Suínos , Transfecção , Vasoconstrição/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
8.
Circ Res ; 88(10): 1088-94, 2001 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-11375280

RESUMO

Heme oxygenase (HO) is a cytoprotective enzyme that degrades heme (a potent oxidant) to generate carbon monoxide (a vasodilatory gas that has anti-inflammatory properties), bilirubin (an antioxidant derived from biliverdin), and iron (sequestered by ferritin). Because of properties of HO and its products, we hypothesized that HO would be important for the regulation of blood pressure and ischemic injury. We studied chronic renovascular hypertension in mice deficient in the inducible isoform of HO (HO-1) using a one kidney-one clip (1K1C) model of disease. Systolic blood pressure was not different between wild-type (HO-1(+/+)), heterozygous (HO-1(+/-)), and homozygous null (HO-1(-/-)) mice at baseline. After 1K1C surgery, HO-1(+/+) mice developed hypertension (140+/-2 mm Hg) and cardiac hypertrophy (cardiac weight index of 5.0+/-0.2 mg/g) compared with sham-operated HO-1(+/+) mice (108+/-5 mm Hg and 4.1+/-0.1 mg/g, respectively). However, 1K1C produced more severe hypertension (164+/-2 mm Hg) and cardiac hypertrophy (6.9+/-0.6 mg/g) in HO-1(-/-) mice. HO-1(-/-) mice also experienced a high rate of death (56%) within 72 hours after 1K1C surgery compared with HO-1(+/+) (25%) and HO-1(+/-) (28%) mice. Assessment of renal function showed a significantly higher plasma creatinine in HO-1(-/-) mice compared with HO-1(+/-) mice. Histological analysis of kidneys from 1K1C HO-1(-/-) mice revealed extensive ischemic injury at the corticomedullary junction, whereas kidneys from sham HO-1(-/-) and 1K1C HO-1(+/-) mice appeared normal. Taken together, these data suggest that chronic deficiency of HO-1 does not alter basal blood pressure; however, in the 1K1C model an absence of HO-1 leads to more severe renovascular hypertension and cardiac hypertrophy. Moreover, renal artery clipping leads to an acute increase in ischemic damage and death in the absence of HO-1.


Assuntos
Injúria Renal Aguda/patologia , Heme Oxigenase (Desciclizante)/deficiência , Hipertensão Renovascular/genética , Injúria Renal Aguda/sangue , Injúria Renal Aguda/complicações , Animais , Pressão Sanguínea/genética , Cardiomegalia/etiologia , Cardiomegalia/patologia , Doença Crônica , Creatinina/sangue , Modelos Animais de Doenças , Antagonistas dos Receptores de Endotelina , Endotelina-1/genética , Endotelina-1/metabolismo , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1 , Heterozigoto , Homozigoto , Hipertensão Renovascular/sangue , Hipertensão Renovascular/complicações , Imuno-Histoquímica , Rim/patologia , Proteínas de Membrana , Camundongos , Camundongos Knockout , Nefrectomia , Tamanho do Órgão , RNA Mensageiro/metabolismo , Receptor de Endotelina A , Obstrução da Artéria Renal/complicações , Índice de Gravidade de Doença , Taxa de Sobrevida
9.
Nat Med ; 7(5): 598-604, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11329062

RESUMO

Carbon monoxide (CO) can arrest cellular respiration, but paradoxically, it is synthesized endogenously by heme oxygenase type 1 (Ho-1) in response to ischemic stress. Ho-1-deficient (Hmox1-/-) mice exhibited lethal ischemic lung injury, but were rescued from death by inhaled CO. CO drove ischemic protection by activating soluble guanylate cyclase and thereby suppressed hypoxic induction of the gene encoding plasminogen activator inhibitor-1 (PAI-1) in mononuclear phagocytes, which reduced accrual of microvascular fibrin. CO-mediated ischemic protection observed in wild-type mice was lost in mice null for the gene encoding PAI-1 (Serpine1). These data establish a fundamental link between CO and prevention of ischemic injury based on the ability of CO to derepress the fibrinolytic axis. These data also point to a potential therapeutic use for inhaled CO.


Assuntos
Monóxido de Carbono/administração & dosagem , Traumatismo por Reperfusão/prevenção & controle , Animais , Sequência de Bases , Monóxido de Carbono/uso terapêutico , Linhagem Celular , Primers do DNA , Feminino , Fibrinólise , Heme Oxigenase (Desciclizante)/genética , Imuno-Histoquímica , Lipopolissacarídeos/administração & dosagem , Pulmão/irrigação sanguínea , Masculino , Camundongos , Inibidor 1 de Ativador de Plasminogênio/biossíntese
10.
J Biol Chem ; 276(20): 17479-83, 2001 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-11278246

RESUMO

Inactivation of glycogen synthase kinase 3beta (GSK3beta) and the resulting stabilization of free beta-catenin are critical steps in the activation of Wnt target genes. While Akt regulates GSK3alpha/beta in the phosphatidylinositide 3-OH kinase signaling pathway, its role in Wnt signaling is unknown. Here we report that expression of Wnt or Dishevelled (Dvl) increased Akt activity. Activated Akt bound to the Axin-GSK3beta complex in the presence of Dvl, phosphorylated GSK3beta and increased free beta-catenin levels. Furthermore, in Wnt-overexpressing PC12 cells, dominant-negative Akt decreased free beta-catenin and derepressed nerve growth factor-induced differentiation. Therefore, Akt acts in association with Dvl as an important regulator of the Wnt signaling pathway.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras , Transdução de Sinais/fisiologia , Transativadores , Proteínas de Peixe-Zebra , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteína Axina , Linhagem Celular , Proteínas do Citoesqueleto/metabolismo , Proteínas Desgrenhadas , Ativação Enzimática , Genes Reporter , Quinase 3 da Glicogênio Sintase , Quinases da Glicogênio Sintase , Humanos , Camundongos , Células PC12 , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Proteínas Wnt , beta Catenina
11.
J Biol Chem ; 275(47): 36847-51, 2000 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-11018023

RESUMO

The onset of myocardial infarction occurs frequently in the early morning, and it may partly result from circadian variation of fibrinolytic activity. Plasminogen activator inhibitor-1 activity shows a circadian oscillation and may account for the morning onset of myocardial infarction. However, the molecular mechanisms regulating this circadian oscillation remain unknown. Recent evidence indicates that basic helix-loop-helix (bHLH)/PAS domain transcription factors play a crucial role in controlling the biological clock that controls circadian rhythm. We isolated a novel bHLH/PAS protein, cycle-like factor (CLIF) from human umbilical vein endothelial cells. CLIF shares high homology with Drosophila CYCLE, one of the essential transcriptional regulators of circadian rhythm. CLIF is expressed in endothelial cells and neurons in the brain, including the suprachiasmatic nucleus, the center of the circadian clock. In endothelial cells, CLIF forms a heterodimer with CLOCK and up-regulates the PAI-1 gene through E-box sites. Furthermore, Period2 and Cryptochrome1, whose expression show a circadian oscillation in peripheral tissues, inhibit the PAI-1 promoter activation by the CLOCK:CLIF heterodimer. These results suggest that CLIF regulates the circadian oscillation of PAI-1 gene expression in endothelial cells. In addition, the results potentially provide a molecular basis for the morning onset of myocardial infarction.


Assuntos
Ritmo Circadiano , Regulação da Expressão Gênica , Sequências Hélice-Alça-Hélice , Inibidor 1 de Ativador de Plasminogênio/genética , Fatores de Transcrição/fisiologia , Fatores de Transcrição ARNTL , Sequência de Aminoácidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Clonagem Molecular , Dimerização , Humanos , Hibridização In Situ , Dados de Sequência Molecular , Filogenia , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Ativação Transcricional
12.
J Biol Chem ; 275(32): 24840-6, 2000 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-10823822

RESUMO

Heme oxygenase (HO)-1 is a stress response protein that is regulated by oxidative stress. HO-1 catalyzes the generation of biliverdin, carbon monoxide, and iron from heme. Lipopolysaccharide (LPS) and interleukin (IL)-1beta induce HO-1 through the binding of nuclear proteins to AP-1 motifs in enhancer regions upstream from the transcription start site. The DNA binding activity of AP-1 proteins depends on the reduction of cysteines in their DNA-binding domains. We found that agents that disrupt free sulfhydryl groups abolish AP-1 binding activity in nuclear proteins obtained from rat aortic smooth muscle cells and macrophages stimulated with IL-1beta or LPS. Thioredoxin (TRX) may regulate the redox status of nuclear transcription factors in response to oxidative stimuli, thus we determined the role of TRX in the physiologic regulation of HO-1. TRX underwent nuclear translocation in cells stimulated with IL-1beta and LPS. We transfected macrophages with a heterologous promoter construct containing two AP-1 sites from an upstream enhancer region in the HO-1 promoter. Recombinant TRX induced promoter activity to a level analogous to that induced by LPS, and this TRX response was abolished by mutation of the AP-1 sites. An inhibitor of TRX reductase, used to prevent TRX translocation in the reduced state, decreased HO-1 induction by IL-1beta and LPS. These data provide the first evidence that TRX contributes to the induction of HO-1 by inflammatory mediators.


Assuntos
DNA Liase (Sítios Apurínicos ou Apirimidínicos) , Heme Oxigenase (Desciclizante)/genética , Interleucina-1/farmacologia , Macrófagos/enzimologia , Músculo Liso Vascular/enzimologia , Tiorredoxinas/metabolismo , Animais , Aorta/citologia , Aorta/enzimologia , Carbono-Oxigênio Liases/genética , Linhagem Celular , Células Cultivadas , Elementos Facilitadores Genéticos , Indução Enzimática , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Heme Oxigenase (Desciclizante)/biossíntese , Heme Oxigenase-1 , Humanos , Lipopolissacarídeos/farmacologia , Masculino , Proteínas de Membrana , Músculo Liso Vascular/citologia , Mutagênese Sítio-Dirigida , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia , Fator de Transcrição AP-1/metabolismo , Transfecção
13.
Circulation ; 102(24): 3015-22, 2000 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-11113055

RESUMO

BACKGROUND: Heme oxygenase (HO)-1 is an enzyme that degrades heme to generate CO (a vasodilatory gas), iron, and the potent antioxidant bilirubin. A disease process characterized by decreases in vascular tone and increases in oxidative stress is endotoxic shock. Moreover, HO-1 is markedly induced in multiple organs after the administration of endotoxin (lipopolysaccharide [LPS]) to mice. METHODS AND RESULTS: To determine the role of HO-1 in endotoxemia, we administered LPS to mice that were wild-type (+/+), heterozygous (+/-), or homozygous null (-/-) for targeted disruption of HO-1. LPS produced a similar induction of HO-1 mRNA and protein in HO-1(+/+) and HO-1(+/-) mice, whereas HO-1(-/-) mice showed no HO-1 expression. Four hours after LPS, systolic blood pressure (SBP) decreased in all the groups. However, SBP was significantly higher in HO-1(-/-) mice (121+/-5 mm Hg) after 24 hours, compared with HO-1(+/+) (96+/-7 mm Hg) and HO-1(+/-) (89+/-13 mm Hg) mice. A sustained increase in endothelin-1 contributed to this SBP response. Even though SBP was higher, mortality was increased in HO-1(-/-) mice, and they exhibited hepatic and renal dysfunction that was not present in HO-1(+/+) and HO-1(+/-) mice. The end-organ damage and death in HO-1(-/-) mice was related to increased oxidative stress. CONCLUSIONS: These data suggest that the increased mortality during endotoxemia in HO-1(-/-) mice is related to increased oxidative stress and end-organ (renal and hepatic) damage, not to refractory hypotension.


Assuntos
Endotoxemia/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Lipopolissacarídeos/toxicidade , Insuficiência de Múltiplos Órgãos/mortalidade , Animais , Endotelina-1/biossíntese , Endotelina-1/genética , Endotoxemia/enzimologia , Endotoxemia/fisiopatologia , Feminino , Heme Oxigenase (Desciclizante)/deficiência , Heme Oxigenase-1 , Hipotensão/induzido quimicamente , Hipotensão/etiologia , Pulmão/patologia , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Mortalidade , Insuficiência de Múltiplos Órgãos/enzimologia , Insuficiência de Múltiplos Órgãos/fisiopatologia , Estresse Oxidativo , RNA Mensageiro/biossíntese
14.
J Biol Chem ; 274(44): 31565-70, 1999 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-10531360

RESUMO

Endothelial PAS domain protein 1 (EPAS1) is a basic helix-loop-helix/PAS domain transcription factor that is preferentially expressed in vascular endothelial cells. EPAS1 shares high homology with hypoxia-inducible factor-1alpha (HIF-1alpha) and, like HIF-1alpha, has been shown to bind to the HIF-1-binding site and to activate its downstream genes such as vascular endothelial growth factor (VEGF) and erythropoietin. In this report, we show that EPAS1 increased VEGF gene expression through the HIF-1-binding site. This transactivation was enhanced further by cotransfection of an aryl hydrocarbon receptor nuclear translocator expression plasmid. Deletion analysis of EPAS1 revealed a potent activation domain (amino acids 486-639) essential for EPAS1 to transactivate the VEGF promoter. We confirmed the ability of this domain to activate transcription using a Gal4 fusion protein system. Because a truncated EPAS1 protein lacking the transactivation domain at amino acids 486-639 eliminated induction of the VEGF promoter by wild-type EPAS1, the truncated protein functions as a dominant-negative mutant. Most important, infection of the cells with an adenoviral construct expressing this mutant inhibited the induction of VEGF mRNA under conditions that mimic hypoxia. Our results suggest that EPAS1 is an important regulator of VEGF gene expression. Since VEGF plays a crucial role in angiogenesis, the ability of dominant-negative EPAS1 to inhibit VEGF promoter activity raises the possibility of a novel approach to inhibiting pathological angiogenesis.


Assuntos
Mutação , Proteínas de Saccharomyces cerevisiae , Transativadores/genética , Ativação Transcricional , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Bovinos , Proteínas de Ligação a DNA/metabolismo , Desferroxamina/farmacologia , Fatores de Crescimento Endotelial/biossíntese , Fatores de Crescimento Endotelial/genética , Endotélio Vascular/citologia , Proteínas Fúngicas , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Linfocinas/biossíntese , Linfocinas/genética , Proteínas Nucleares/metabolismo , Fragmentos de Peptídeos/genética , Regiões Promotoras Genéticas , Ligação Proteica , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Proteínas Recombinantes de Fusão , Deleção de Sequência , Transativadores/metabolismo , Fatores de Transcrição , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
15.
J Biol Chem ; 274(20): 14344-51, 1999 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-10318857

RESUMO

Aortic preferentially expressed gene-1 (APEG-1) was originally identified as a 1.4-kilobase (kb) transcript preferentially expressed in differentiated vascular smooth muscle cells (VSMC). Its expression is markedly down-regulated in de-differentiated VSMC, suggesting a role for APEG-1 in VSMC differentiation. We have now determined that APEG-1 is a single-copy gene in the human, rat, and mouse genomes and have mapped human APEG-1 to chromosome 2q34. To study the molecular mechanisms regulating its expression, we characterized the genomic organization and promoter of mouse APEG-1. APEG-1 spans 4.5 kb in the mouse genome and is composed of five exons. Using reporter gene transfection analysis, we found that a 2. 7-kb APEG-1 5'-flanking sequence directed a high level of promoter activity only in VSMC. Its activity was minimal in five other cell types. A repressor region located within an upstream 685-base pair sequence suppressed the activity of this 2.7-kb promoter. Further deletion and mutation analyses identified an E box motif as a positive regulatory element, which was bound by nuclear protein prepared from VSMC. In conjunction with its flanking sequence, this E box motif confers VSMC-specific enhancer activity to a heterologous SV40 promoter. To our knowledge, this is the first demonstration of an E box motif that mediates gene expression restricted to VSMC.


Assuntos
Proteínas Musculares/genética , Músculo Liso Vascular/metabolismo , Regiões Promotoras Genéticas , Sequência de Aminoácidos , Animais , Sequência de Bases , Biomarcadores , Células Cultivadas , Cromossomos Humanos Par 2 , Clonagem Molecular , Humanos , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Quinase de Cadeia Leve de Miosina , Proteínas Serina-Treonina Quinases , Ratos , Deleção de Sequência
16.
J Clin Invest ; 103(8): R23-9, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10207174

RESUMO

Heme oxygenase (HO) catalyzes the oxidation of heme to generate carbon monoxide (CO) and bilirubin. CO increases cellular levels of cGMP, which regulates vascular tone and smooth muscle development. Bilirubin is a potent antioxidant. Hypoxia increases expression of the inducible HO isoform (HO-1) but not the constitutive isoform (HO-2). To determine whether HO-1 affects cellular adaptation to chronic hypoxia in vivo, we generated HO-1 null (HO-1(-/-)) mice and subjected them to hypoxia (10% oxygen) for five to seven weeks. Hypoxia caused similar increases in right ventricular systolic pressure in wild-type and HO-1(-/-) mice. Although ventricular weight increased in wild-type mice, the increase was greater in HO-1(-/-) mice. Similarly, the right ventricles were more dilated in HO-1(-/-) mice. After seven weeks of hypoxia, only HO-1(-/-) mice developed right ventricular infarcts with organized mural thrombi. No left ventricular infarcts were observed. Lipid peroxidation and oxidative damage occurred in right ventricular cardiomyocytes in HO-1(-/-), but not wild-type, mice. We also detected apoptotic cardiomyocytes surrounding areas of infarcted myocardium by terminal deoxynucleotide transferase-mediated dUTP nick end-labeling (TUNEL) assays. Our data suggest that in the absence of HO-1, cardiomyocytes have a maladaptive response to hypoxia and subsequent pulmonary hypertension. J.Clin. Invest. 103:R23-R29 (1999).


Assuntos
Heme Oxigenase (Desciclizante)/fisiologia , Infarto do Miocárdio/etiologia , Disfunção Ventricular Esquerda/etiologia , Animais , Dilatação Patológica , Feminino , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase-1 , Hipóxia , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Pressão Ventricular
17.
J Biol Chem ; 274(13): 9045-52, 1999 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-10085153

RESUMO

Nitric oxide (NO), a free radical gas whose production is catalyzed by the enzyme NO synthase, participates in the regulation of multiple organ systems. The inducible isoform of NO synthase (iNOS) is transcriptionally up-regulated by inflammatory stimuli; a critical mediator of this process is nuclear factor (NF)-kappaB. Our objective was to determine which regulatory elements other than NF-kappaB binding sites are important for activation of the iNOS promoter/enhancer. We also wanted to identify transcription factors that may be functioning in conjunction with NF-kappaB (subunits p50 and p65) to drive iNOS transcription. Deletion analysis of the iNOS promoter/enhancer revealed that an AT-rich sequence (-61 to -54) downstream of the NF-kappaB site (-85 to -76) in the 5'-flanking sequence was important for iNOS induction by interleukin-1beta and endotoxin in vascular smooth muscle cells. This AT-rich sequence, corresponding to an octamer (Oct) binding site, bound the architectural transcription factor high mobility group (HMG)-I(Y) protein. Electrophoretic mobility shift assays showed that HMG-I(Y) and NF-kappaB subunit p50 bound to the iNOS promoter/enhancer to form a ternary complex. The formation of this complex required HMG-I(Y) binding at the Oct site. The location of an HMG-I(Y) binding site typically overlaps that of a recruited transcription factor. In the iNOS promoter/enhancer, however, HMG-I(Y) formed a complex with p50 while binding downstream of the NF-kappaB site. Furthermore, overexpression of HMG-I(Y) potentiated iNOS promoter/enhancer activity by p50 and p65 in transfection experiments, suggesting that HMG-I(Y) contributes to the transactivation of iNOS by NF-kappaB.


Assuntos
Proteínas de Grupo de Alta Mobilidade/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico Sintase/genética , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação/genética , Células Cultivadas , Análise Mutacional de DNA , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica/genética , Proteína HMGA1a , Interleucina-1/farmacologia , Músculo Liso Vascular/enzimologia , NF-kappa B/genética , Óxido Nítrico Sintase Tipo II , Regiões Promotoras Genéticas/genética , Ratos , Ativação Transcricional/genética
18.
J Biol Chem ; 274(3): 1525-32, 1999 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9880529

RESUMO

Nonhistone chromosomal proteins of the high mobility group (HMG) affect the transcriptional regulation of certain mammalian genes. For example, HMG-I(Y) controls cytokine-mediated promoters that require transcription factors, such as nuclear factor-kappaB, for maximal expression. Even though a great deal is known about how HMG-I(Y) facilitates expression of other genes, less is known about the regulation of HMG-I(Y) itself, especially in cells in primary culture. Therefore we investigated the effect of endotoxin and the cytokine interleukin-1beta on HMG-I(Y) expression in vascular smooth muscle cells. Induction of HMG-I(Y) peaked after 48 h of interleukin-1beta stimulation (6.2-fold) in cells in primary culture, and this increase in mRNA corresponded to an increase in HMG-I(Y) protein. Moreover, immunohistochemical staining revealed a dramatic increase in HMG-I(Y) protein expression in vascular smooth muscle cells after endotoxin stimulation in vivo. This increase in HMG-I(Y) expression (both in vitro and in vivo) mirrored an up-regulation of inducible nitric oxide synthase, a cytokine-responsive gene. The functional significance of this coinduction is underscored by our finding that HMG-I(Y) potentiated the response of inducible nitric oxide synthase to nuclear factor-kappaB transactivation. Taken together, these studies suggest that induction of HMG-I(Y), and subsequent transactivation of iNOS, may contribute to a reduction in vascular tone during endotoxemia and other systemic inflammatory processes.


Assuntos
Proteínas de Grupo de Alta Mobilidade/biossíntese , Interleucina-1/farmacologia , Lipopolissacarídeos/farmacologia , Músculo Liso Vascular/metabolismo , Proteínas de Neoplasias/biossíntese , Óxido Nítrico Sintase/metabolismo , Fatores de Transcrição/biossíntese , Animais , Células Cultivadas , Distamicinas/farmacologia , Drosophila , Ativação Enzimática , Indução Enzimática , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Proteína HMGA1a , Macrófagos Alveolares/metabolismo , Masculino , Músculo Liso Vascular/efeitos dos fármacos , NF-kappa B/farmacologia , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Ativação Transcricional/efeitos dos fármacos
19.
Circ Res ; 83(10): 980-5, 1998 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-9815145

RESUMO

Proteins of the LIM family are critical regulators of development and differentiation in various cell types. We have described the cloning of cysteine-rich protein 2/smooth muscle LIM protein (CRP2/SmLIM), a LIM-only protein expressed in differentiated vascular smooth muscle cells. As a first step toward understanding the potential functions of CRP2/SmLIM, we analyzed its expression after gastrulation in developing mice and compared the expression of CRP2/SmLIM with that of the other 2 members of the CRP subclass, CRP1 and CRP3/MLP. In situ hybridization in whole-mount and sectioned embryos showed that CRP2/SmLIM was expressed in the sinus venosus and the 2 cardiac chambers at embryonic day 9. Vascular expression of CRP2/SmLIM was first seen at embryonic day 10. At subsequent time points, CRP2/SmLIM expression decreased in the heart but remained high in the vasculature. CRP1 was expressed both in vascular and nonvascular tissues containing smooth muscle cells, whereas CRP3/MLP was expressed only in tissues containing striated muscle. These patterns of expression were maintained in the adult animal and suggest an important role for this gene family in the development of smooth and striated muscle.


Assuntos
Sistema Cardiovascular/embriologia , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Proteínas Musculares/genética , Proteínas Nucleares/genética , Fatores Etários , Animais , Proteína delta de Ligação ao Facilitador CCAAT , Proteínas Estimuladoras de Ligação a CCAAT , Desenvolvimento Embrionário e Fetal , Hibridização In Situ , Proteínas com Domínio LIM , Zíper de Leucina/genética , Camundongos , Músculo Liso Vascular/embriologia , RNA Mensageiro/análise , Fatores de Transcrição/genética , Dedos de Zinco/genética
20.
Circ Res ; 83(4): 396-403, 1998 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-9721696

RESUMO

Heme oxygenase (HO)-1 generates CO, a gas with vasodilatory properties, during heme metabolism. HO-1 is expressed highly in vascular tissue after endotoxin stimulation, and generation of CO through the HO-1 pathway contributes to the hemodynamic compromise of endotoxic shock. Shock related to endotoxemia is an immune-mediated process that involves the generation of proinflammatory cytokines such as interleukin (IL)-1beta. Because transforming growth factor (TGF)-beta1 is a modulator of immune-mediated inflammatory responses and it blocks the hypotension of endotoxic shock, we determined whether TGF-beta1 could be used to reduce expression of HO-1 in vascular tissue and smooth muscle cells. In a rat model of endotoxic shock, lipopolysaccharide-induced HO-1 mRNA and protein expression was reduced by TGF-beta1 in highly vascularized tissue, such as heart and lung, by Northern and Western analysis. Furthermore, TGF-beta1 downregulated HO-1 mRNA after its induction by IL-1beta in vascular smooth muscle cells in culture. TGF-beta1 also decreased HO-1 but not HO-2 protein expression in these cells. TGF-beta1 decreased HO enzyme activity induced in IL-1beta treated vascular smooth muscle cells to a level not different from that in vehicle-treated cells. These studies suggest that this downregulation of HO-1 mRNA and protein expression and decrease in IL-1beta-induced HO enzyme activity may contribute to the beneficial effect of TGF-beta1 on endotoxic shock.


Assuntos
Endotoxemia/enzimologia , Heme Oxigenase (Desciclizante)/sangue , Salmonella typhi , Fator de Crescimento Transformador beta/farmacologia , Animais , Regulação para Baixo/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Masculino , Músculo Liso Vascular/enzimologia , Ratos , Ratos Sprague-Dawley , Choque Séptico/enzimologia , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA