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










Base de dados
Intervalo de ano de publicação
1.
Circ J ; 81(6): 888-890, 2017 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-28420827

RESUMO

BACKGROUND: This study determined whether relaxin or matrix metalloproteinase (MMP)-9 influences angiotensin II (AngII)-induced abdominal aortic aneurysms (AAA).Methods and Results:Male C57BL/6 or apolipoprotein E-/-mice were infused with AngII with or without relaxin. Relaxin did not influence AngII-induced AAA in either mouse strain. Infusion of AngII reduced, but relaxin increased, MMP-9 mRNA in macrophages. We then determined the effects of MMP-9 deficiency on AAA in apolipoprotein E-/-mice. MMP-9 deficiency led to AAA formation in the absence of AngII, and augmented AngII-induced aortic rupture and AAA incidence. CONCLUSIONS: MMP-9 deficiency augmented AngII-induced AAA.


Assuntos
Angiotensina II/efeitos adversos , Aneurisma da Aorta Abdominal/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Relaxina/biossíntese , Angiotensina II/farmacologia , Animais , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/patologia , Apolipoproteínas E/deficiência , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Knockout , Relaxina/genética
2.
Cardiovasc Pathol ; 16(4): 231-6, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17637431

RESUMO

BACKGROUND: Angiotensin II (Ang II) promotes atherosclerotic vascular diseases, in which proinflammatory and proliferative effects play a major pathogenic role. Ang II up-regulates chemokines, such as monocyte chemoattractant protein (MCP)-1 and macrophage inflammatory protein (MIP)-1alpha, which are important pro-inflammatory factors mediating infiltration of inflammatory cells into atherosclerotic lesion. The aim of the present study was to determine whether the presence of MCP-1 or MIP-1alpha is essential in Ang II-induced intimal hyperplasia in the carotid artery ligation model. METHODS: Six-month-old male C57BL/6-, MCP-1-, or MIP-1alpha-deficient mice underwent ligation of the common left carotid artery and were randomly assigned to receive either vehicle or Ang II (1.4 mg kg(-1) day(-1)) via a subcutaneously implanted osmotic infusion pump (model 2004, Alzet) for 4 weeks. RESULTS: Ang II not only increased MCP-1 and MIP-1alpha production but also enhanced neo-intimal formation, media thickness, and adventitia development in the ligated carotid arteries in C57BL/6 mice. However, MCP-1 or MIP-1alpha deficiency failed to affect intimal hyperplasia in vascular remodeling. CONCLUSION: These results indicate that MCP-1 or MIP-1alpha may not be essential in mediating the proliferative effects of Ang II, a major pathological changes in intimal hyperplasia in the carotid artery ligation model.


Assuntos
Angiotensina II/metabolismo , Artérias Carótidas/metabolismo , Quimiocina CCL2/metabolismo , Proteínas Inflamatórias de Macrófagos/metabolismo , Túnica Íntima/patologia , Animais , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Quimiocina CCL3 , Quimiocina CCL4 , Hiperplasia , Imuno-Histoquímica , Ligadura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Túnica Íntima/metabolismo
3.
Thromb Haemost ; 97(5): 847-55, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17479197

RESUMO

Irreversible platelet inhibitors, such as aspirin and clopidogrel, have limited anti-thrombotic efficacy in the clinic due to their bleeding risk. We have developed an orally active reversible P2Y(12) receptor antagonist, BX 667. The aim of this study was to determine if the reversible antagonist BX 667 had a greater therapeutic index than the irreversible P2Y(12) receptor antagonist clopidogrel. Since BX 667 is rapidly converted to its active metabolite BX 048 in rats, we first injected BX 048 intravenously (iv) in a rat arterial venous (A-V) shunt model of thrombosis. BX 048 dose- and concentration-dependently attenuated thrombosis. When administered orally, BX 667 and clopidogrel had similar efficacy, but BX 667 caused less bleeding than clopidogrel. In a rat model of a platelet-rich thrombus induced by vessel injury with FeCl(2), both BX 667 and clopidogrel exhibited higher levels of thrombus inhibition after oral administration compared to their potency in the A-V shunt model. Again, BX 667 caused less bleeding than clopidogrel. In a dog cyclic flow model, iv injection of either BX 667 or clopidogrel dose-dependently reduced thrombus formation with lower bleeding for BX 667 than clopidogrel. Inhibition of thrombosis was highly correlated with inhibition of ADP-induced platelet aggregation in these animal models. In dogs pre-treated with aspirin, BX 667 maintained its wider therapeutic index, measured by inhibition of platelet aggregation over bleeding, compared to the aspirin-clopidogrel combination. These data demonstrate that the reversible P2Y(12) receptor antagonist, BX 667, has a wider therapeutic index than clopidogrel in experimental models of thrombosis.


Assuntos
Proteínas de Membrana/antagonistas & inibidores , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Antagonistas do Receptor Purinérgico P2 , Trombose/prevenção & controle , Animais , Derivação Arteriovenosa Cirúrgica , Lesões das Artérias Carótidas/tratamento farmacológico , Clopidogrel , Modelos Animais de Doenças , Cães , Técnicas In Vitro , Masculino , Estrutura Molecular , Inibidores da Agregação Plaquetária/sangue , Inibidores da Agregação Plaquetária/química , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2 , Receptores Purinérgicos P2Y12 , Ticlopidina/análogos & derivados , Ticlopidina/farmacologia
4.
Am J Physiol Heart Circ Physiol ; 293(1): H69-76, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17434974

RESUMO

The present study tested the hypothesis that murine (m)IFN-beta or mIFN-alpha(2) can eliminate cardiac viral load and protect cardiomyocytes from injury in animals infected with coxsackievirus B3 (CVB3). CVB3-inoculated male Balb/c mice exhibited signs of illness, including lethargy, progressive weight loss, and death (10% on day 3 and 100% on day 8). Cardiac viral load was high [4,277 +/- 1,009 plaque-forming units and 25 +/- 5 copies CVB3/hypoxanthine guanine phosphoribosyl transferase 1 mRNA] on day 4. The cardiac tissue exhibited severe inflammatory infiltration and myocyte damage with an average myocarditis integrated pathology score of 2.1 +/- 0.2 on day 7. Most of the mice infected with CVB3 also developed epicarditis, and 55% had intraventricular thrombi present. Treatment with mIFN-beta [2.5 to 10 million international units (MIU)/kg] dose-dependently improved the general health status in CVB3-inoculated mice, as evidenced by reduction in weight loss, prevention of death, elimination of cardiac viral load, protection of myocytes from injury, decrease in inflammatory cell infiltration, and attenuation of intraventricular thrombus formation. Treatment with 10 MIU/kg mIFN-alpha(2) resulted in a similar level of efficacy as that induced by 5 MIU/kg mIFN-beta, with the exception that mIFN-alpha(2) did not reduce cardiac CVB3 mRNA. However, mIFN-alpha(2) , but not any dose group of mIFN-beta, significantly attenuated CVB3-induced epicarditis. These data demonstrate antiviral effects for both mIFN-beta and mIFN-alpha(2), which lead to protection of the mice from CVB3-induced myocarditis. However, the potential mechanisms leading to a differential host response for the two isoforms of mIFN remain to be elucidated.


Assuntos
Enterovirus/efeitos dos fármacos , Interferon-alfa/administração & dosagem , Interferon beta/administração & dosagem , Miocardite/tratamento farmacológico , Miocardite/virologia , Pericárdio/efeitos dos fármacos , Pericárdio/virologia , Animais , Antivirais/administração & dosagem , Cardiotônicos/administração & dosagem , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células Musculares/efeitos dos fármacos , Células Musculares/virologia , Resultado do Tratamento
5.
Thromb Haemost ; 97(1): 54-61, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17200771

RESUMO

We have discovered a novel small-molecule TAFIa inhibitor, BX 528, which is potent, highly selective against other carboxypeptidases and safe. The present study was to determine if BX 528 can enhance exogenous and endogenous thrombolysis in four different animal models. In the first three models, a thrombus was induced by FeCl (2) (dogs) or laser (rats) injury of the femoral artery, or formed ex vivo and implanted in the jugular vein in rabbits. A low dose of exogenous t-PA was given to induce a low-level thrombolysis on an established thrombus. Co-treatment with BX 528 further enhanced the thrombolytic effects induced by the exogenous t-PA and, thus, reduced thrombosis in all three animal models. In a second rat model, fibrin deposition in the lungs was induced by batroxobin, which was spontaneously resolved in 30 minutes due to the activation of endogenous fibrinolysis. Pre-treatment with lipopolysaccharide (LPS) attenuated this spontaneous fibrinolysis. Co-treatment with 10 mg/kg BX 528 prevented the LPS-induced attenuation of endogenous fibrinolysis. Thus, these studies demonstrated that inhibition of TAFIa by BX 528, our newly discovered small-molecule TAFIa inhibitor, enhanced both the exogenous (induced by a low dose of t-PA) and endogenous (LPS-induced resistance) thrombolysis without increasing the bleeding risk in four different animal models of thrombosis in different species (rat, dog and rabbit) employing different thrombogenic stimuli (FeCl (2) , laser, ex vivo and batroxobin) to induce thrombus formation in different tissues (artery, vein and lung microcirculation).


Assuntos
Carboxipeptidase B2/antagonistas & inibidores , Fibrinólise/efeitos dos fármacos , Fibrinolíticos/farmacologia , Trombose/tratamento farmacológico , Animais , Modelos Animais de Doenças , Cães , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Lipopolissacarídeos/farmacologia , Propionatos/farmacologia , Coelhos , Ratos , Ativador de Plasminogênio Tecidual/farmacologia
6.
Transl Res ; 149(2): 70-5, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17240317

RESUMO

It has been demonstrated that urokinase-type plasminogen activator (uPA) plays an important role in vascular remodeling. This study was designed to determine whether uPA deficiency (KO) affects carotid artery ligation-induced vessel remodeling and the interaction with angiotensin II (Ang II). Ligation of the left common carotid artery in 6-month-old wild-type (C57 black/6J) mice for 4 weeks induced a concentric remodeling with vessel wall thickening, characterized by cell proliferation in neointima, media, and adventitia, and with lumen narrowing without a significant enlargement of overall vessel dimension. Intima lesions were characterized by alpha-actin positive smooth muscle cell (SMC) proliferation in a matrix background. No detectable presence of MAC-3 positive macrophages existed in the vascular wall. The ligation-induced vascular neointimal formation and adventitial proliferation, but not lumen narrowing and media expansion, were completely prevented in age-matched uPA-KO mice. Chronic infusion of Ang II (1.44 mg/kg per day) via a subcutaneously implanted osmotic minipump did not significantly affect the gross morphology of the nonligated carotid artery from both wild-type and uPA-KO mice, but it enhanced ligation-induced vascular remodeling. However, in the presence of Ang II, uPA deficiency had no effects on ligation-induced mophermetric change, but it partially and significantly reduced cell proliferation. These data indicate that uPA may play a critical role in ligation-induced vessel remodeling. Ang II may activate other mechanisms independent of uPA to exacerbate ligation-induced vascular remodeling.


Assuntos
Doenças das Artérias Carótidas/patologia , Doenças das Artérias Carótidas/fisiopatologia , Ativador de Plasminogênio Tipo Uroquinase/genética , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Animais , Bombas de Infusão Implantáveis , Ligadura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ativador de Plasminogênio Tipo Uroquinase/deficiência , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Vasoconstritores/metabolismo , Vasoconstritores/farmacologia
7.
J Vasc Surg ; 44(2): 364-71, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16890870

RESUMO

BACKGROUND: We explored the role of angiotensin II in determining the histomorphometric features of plaque stability in apolipoprotein E-deficient mice submitted to ligation of the carotid artery. METHODS: Six-month-old apolipoprotein E-deficient mice underwent ligation of the common left carotid artery and were immediately assigned to receive either angiotensin II (1.4 mg . kg(-1) . d(-1) subcutaneously) or vehicle (phosphate-buffered saline; control) via a subcutaneous osmotic minipump for 4 weeks. RESULTS: Ligated arteries from control animals developed intimal lesions composed of macrophage foam cell plaques, which accumulated adjacent to the internal elastic lamina and were surrounded by a fibromuscular layer. Angiotensin II-treated mice had a greater intimal area (threefold), which was accompanied by a fivefold increase in the foam cell area. Lesions from angiotensin II-treated mice also displayed complex morphology characterized by intralesional neovasculature and hemorrhage. The content of active matrix metalloproteinase 2, mainly colocalized with macrophage foam cells, and the production of the inflammatory mediators monocyte chemoattractant protein 1 and vascular cell adhesion molecule 1 were also increased by angiotensin II treatment. Although angiotensin II induced vessel expansion and lumen loss to a similar extent, only vessel enlargement correlated with intimal area. CONCLUSIONS: Taken together, this study's results support a role of angiotensin II in plaque vulnerability by promoting intraplaque neovascularization/hemorrhage, inflammation, and expansive remodeling.


Assuntos
Angiotensina II , Aterosclerose/patologia , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/patologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/etiologia , Aterosclerose/metabolismo , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/cirurgia , Doenças das Artérias Carótidas/etiologia , Doenças das Artérias Carótidas/metabolismo , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Células Espumosas/efeitos dos fármacos , Células Espumosas/patologia , Imuno-Histoquímica , Ligadura , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
8.
Arterioscler Thromb Vasc Biol ; 26(4): 765-72, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16456092

RESUMO

BACKGROUND: Endothelial NO deficiency (endothelial NO synthase [eNOS]-knockout [KO]) enhanced smooth muscle cell (SMC)-rich neointimal lesion formation in a mouse model of carotid artery ligation (CAL). Recent evidence indicated that stromal cell-derived factor-1alpha (SDF-1alpha)-mediated recruitment of circulating SMC progenitor cells substantially contributed to the SMC-rich neointimal hyperplasia induced by vascular injury. The goal of this study was to investigate the effects of eNOS deficiency on the expression of SDF-1alpha and mobilization of circulating SMC progenitor cells in CAL model. METHODS AND RESULTS: Two- to 3-month-old C57BL/6J wild-type (WT) and eNOS-KO mice were evaluated 1, 2, or 4 weeks after CAL. CAL-induced expression of SDF-1alpha, as detected by immunohistochemical staining and further quantified by ELISA in the ligated carotid arteries, was moderate and transient with a peak at 1 week in WT mice. SDF-1alpha expression was significantly higher at 1 week and persisted through 2 weeks in eNOS-KO mice. CAL was associated with increased circulating stem cell antigen-1(+) (Sca-1(+))/c-Kit(-)/Lin- cells (interpreted as SMC progenitor cells), which peaked at 1 week in WT mice. This effect was also significantly greater and longer-lasting in eNOS-KO than WT mice. The number of circulating Sca-1(+)/c-Kit(-)/Lin- cells was positively correlated with the expression of SDF-1alpha but not vascular endothelial growth factor in the ligated carotid arteries. Furthermore, immunostaining showed abundant Sca-1-positive cells in the adventitia of the 1-week ligated carotid arteries from eNOS-KO mice but not in WT mice. We also determined that eNOS deficiency enhanced CAL-induced intimal cell proliferation in the ligated arteries as detected by proliferating cell nuclear antigen staining but did not induce cell apoptosis as detected by staining for active caspase-3. CONCLUSIONS: Our results indicate that eNOS deficiency exacerbates CAL-induced expression of SDF-1alpha and its receptor CXCR4. This is correlated with an increase in Sca-1(+) cells in peripheral blood and adventitia, which may contribute to vascular remodeling and SMC-rich neointimal lesion formation. This suggests that constitutive eNOS inhibits SDF-1alpha expression and provides an important vasculoprotective mechanism for intact endothelium to limit SMC proliferation and recruitment in response to vascular injury.


Assuntos
Lesões das Artérias Carótidas/enzimologia , Lesões das Artérias Carótidas/patologia , Quimiocinas CXC/biossíntese , Músculo Liso Vascular/patologia , Óxido Nítrico Sintase Tipo III/deficiência , Túnica Íntima/patologia , Animais , Artérias Carótidas/patologia , Artérias Carótidas/fisiopatologia , Lesões das Artérias Carótidas/fisiopatologia , Diferenciação Celular , Proliferação de Células , Quimiocina CXCL12 , Técnicas In Vitro , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco/patologia , Células-Tronco/fisiologia , Túnica Íntima/fisiopatologia , Regulação para Cima , Fatores de Crescimento do Endotélio Vascular/metabolismo
9.
J Lipid Res ; 47(3): 476-83, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16371646

RESUMO

Two month old C57BL/6 mice were placed on three different diets: 1) normal diet (NC; 0.025% cholesterol), 2) hypercholesterolemic Western-type diet (HC-W; 0.2% cholesterol), and 3) hypercholesterolemic Paigen-type diet (HC-P; 1.25% cholesterol plus 0.5% cholic acid). At 6 months of age, the animals underwent ligation of the left carotid artery and were randomly assigned to vehicle (PBS, subcutaneous) or angiotensin II (Ang II; 1.4 mg/kg/day, subcutaneous) treatment for 4 weeks. Low density lipoprotein-cholesterol levels were similarly increased in both HC diets (NC, 4 +/- 3 mg/dl; HC-W, 123 +/- 17 mg/dl; HC-P, 160 +/- 14 mg/dl). However, the levels of high density lipoprotein-cholesterol (HDL-C) were reduced only in animals fed the HC-P diet (NC, 82 +/- 6 mg/dl; HC-W, 79 +/- 7 mg/dl; HC-P, 58 +/- 7 mg/dl). In Ang II-treated mice, carotid artery ligation induced intimal smooth muscle cell proliferation to a similar extent in NC- and HP-W-fed animals. However, a significantly larger intimal area developed in ligated vessels from Ang II-treated mice fed the HC-P diet (3.6-fold higher than in Ang II-treated NC mice). Together, these results show the accelerating effect of mild hypercholesterolemia, reduced HDL-C levels, and Ang II on intimal hyperplasia after carotid artery ligation in mice.


Assuntos
Angiotensina II/metabolismo , HDL-Colesterol/metabolismo , Hipercolesterolemia/metabolismo , Hiperplasia/metabolismo , Angiotensina II/sangue , Animais , Artérias Carótidas/metabolismo , Artérias Carótidas/cirurgia , HDL-Colesterol/sangue , Dieta , Hipercolesterolemia/dietoterapia , Hiperplasia/fisiopatologia , Imuno-Histoquímica , Metabolismo dos Lipídeos , Camundongos , Camundongos Endogâmicos C57BL
10.
Basic Clin Pharmacol Toxicol ; 97(1): 29-34, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15943756

RESUMO

L-NAME-induced hypertension is characterized by chronic inhibition of nitric oxide synthesis. We have investigated if tempol, an agent mimicking superoxide dismutase might reduce hypertension and the increased vascular reactivity to pressor agents. Rats were divided into: Control, animals receiving L-NAME 50 mg kg(-1)day(-1), tempol 200 mg kg(-1)day(-1) and tempol plus L-NAME. Drugs were administrated in the drinking water for seven days. L-NAME increased mean arterial blood pressure (Control: 108+/-3 mmHg versus L-NAME 181+/-5 mmHg; P<0.05). Tempol did not change arterial pressure and heart rate in L-NAME and Control groups. The reactivity to phenylephrine increased in the L-NAME group (E(max) Control: 2.00+/-0.15 g versus L-NAME: 2.45+/-0.14 g); tempol+L-NAME (E(max): 2.55+/-0.15 g) and in the tempol group (E(max): 2.57+/-0.14 g). Maximal relaxation induced by acetylcholine was reduced in L-NAME group (60.9+/-3%) and tempol+L-NAME (37.4+/-6%) compared to Control (99.1+/-0.12%) and tempol groups (95.6+/-2.12%). All treated groups presented a reduction in the effects of L-NAME administration on basal vascular tone. Our results show that tempol, in the dose used in this study, did not change the effects of L-NAME on blood pressure which suggests that tempol reduces bioavailability of nitric oxide on aortic isolated ring.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Óxidos N-Cíclicos/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/metabolismo , Superóxido Dismutase/farmacologia , Animais , Aorta/efeitos dos fármacos , Aorta/patologia , Disponibilidade Biológica , Endotélio Vascular/fisiologia , Masculino , Fenilefrina/farmacologia , Ratos , Ratos Wistar , Marcadores de Spin
11.
Eur J Pharmacol ; 512(2-3): 215-22, 2005 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-15840407

RESUMO

Recent evidence indicates that the GTPase activated Rho/Rho-kinase pathway contributes angiotensin II-induced cardiac hypertrophy and vascular remodeling. We tested this hypothesis in vivo by determining the effects of fasudil, a Rho-kinase inhibitor, on angiotensin II-induced cardiac hypertrophy, coronary vascular remodeling, and ventricular dysfunction. Six-month-old apolipoprotein E deficient (apoE-KO) mice were subcutaneously infused with angiotensin II (1.44 mg/kg/day) using an osmotic mini-pump. Mice were randomly assigned to either vehicle or fasudil (136 or 213 mg/kg/day in drinking water) group. Infusion of angiotensin II for 4 weeks resulted in cardiac enlargement, myocyte hypertrophy, and myocardial interstitial and coronary artery perivascular fibrosis. These changes were accompanied by reduced aortic flow velocity and acceleration rate. Cardiac gene expression levels of atrial natriuretic peptide (ANP) and collagen type III detected by real-time reverse transcriptase polymerase chain reaction were significantly increased in angiotensin II-infused mice. Treatment with fasudil dose-dependently attenuated angiotensin II-induced cardiac hypertrophy, prevented perivascular fibrosis, blunted the increase in ANP and collagen type III expression, and improved cardiac function, without changing blood pressure. These data are consistent with a role for Rho-kinase activation in angiotensin II-induced cardiac remodeling and vascular wall fibrosis.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Angiotensina II/farmacologia , Apolipoproteínas E/genética , Cardiomegalia/prevenção & controle , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Apolipoproteínas E/metabolismo , Fator Natriurético Atrial/genética , Pressão Sanguínea/efeitos dos fármacos , Cardiomegalia/induzido quimicamente , Cardiomegalia/patologia , Colágeno Tipo III/genética , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/patologia , Relação Dose-Resposta a Droga , Fibrose/prevenção & controle , Expressão Gênica/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Miocárdio/patologia , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Quinases Associadas a rho
12.
Circulation ; 111(17): 2219-26, 2005 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-15851596

RESUMO

BACKGROUND: Angiotensin II (Ang II) accelerates atherosclerosis and induces abdominal aortic aneurysm (AAA) in an experimental mouse model. Agonism of a G protein-coupled receptor by Ang II activates Rho-kinase and other signaling pathways and results in activation of proteolysis and apoptosis. Enhanced proteolysis and smooth muscle cell apoptosis are important mechanisms associated with AAA. In this study, we tested the hypothesis that fasudil, a Rho-kinase inhibitor, could attenuate Ang II-induced AAA formation by inhibiting vascular wall apoptosis and extracellular matrix proteolysis. METHODS AND RESULTS: Six-month-old apolipoprotein E-deficient mice were infused with Ang II (1.44 mg x kg(-1) x d(-1)) for 1 month. Animals were randomly assigned to treatment with fasudil (136 or 213 mg x kg(-1) x d(-1) in drinking water) or tap water. Ang II infusion induced AAA formation in 75% of the mice, which was accompanied by an increase in proteolysis detected by zymographic analysis and quantified by active matrix metalloproteinase-2 activity, as well as apoptosis detected by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling and quantified by both caspase-3 activity and histone-associated DNA fragmentation. The level of DNA fragmentation in the suprarenal aorta correlated with AAA diameter. Ang II also increased atherosclerotic lesion area and blood pressure. Fasudil treatment resulted in a dose-dependent reduction in both the incidence and severity of AAA. At the higher dose, fasudil decreased AAA by 45% while significantly inhibiting both apoptosis and proteolysis, without affecting atherosclerosis or blood pressure. CONCLUSIONS: These data demonstrate that inhibition of Rho-kinase by fasudil attenuated Ang II-induced AAA through inhibition of both apoptosis and proteolysis pathways.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Angiotensina II/farmacologia , Aneurisma da Aorta Abdominal/tratamento farmacológico , Apolipoproteínas E/deficiência , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Apoptose , Endotélio Vascular/citologia , Matriz Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Inibidores de Proteases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Quinases Associadas a rho
13.
Atherosclerosis ; 178(1): 9-17, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15585195

RESUMO

Angiotensin converting enzyme (ACE) inhibitors prevent a wide variety of key events underlying atherogenesis. Whether these actions depend solely on reduction of angiotensin II (Ang II) generation is still to be determined. This study was undertaken to determine whether enalapril, an ACE inhibitor, prevents atherosclerosis and vascular inflammation induced by Ang II in apolipoprotein E-deficient (apoE-KO) mice. Subcutaneous infusion of Ang II (1.44 mg/(kg day)) for 4 weeks increased blood pressure and accelerated atherosclerosis development in the carotid arteries. The expression of the endothelial adhesion molecules E-selectin, intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1), as well as the chemokines monocyte chemotactic protein-1 (MCP-1) and macrophage-colony stimulating factor (M-CSF) was up-regulated in the aortas of Ang II-treated mice. Enalapril co-treatment (25 mg/(kg day), in drinking water) prevented the development of atherosclerosis without affecting blood pressure or circulating cholesterol. In addition to preventing the Ang II-induced over-expression of adhesion molecules and chemokines in the aorta, enalapril up-regulated the expression of peroxisome proliferator-activated receptors (PPARs)-alpha and -gamma, potential anti-inflammatory transcription factors. In the aortic arch, a lesion-prone site, the co-treatment with enalapril reduced the percentage of arterial wall occupied by macrophages and foam cells, medial sclerosis and elastin reduplication. Together, these data suggest an important role for Ang II-independent mechanisms in the antiatherogenic and anti-inflammatory effects of ACE inhibitors.


Assuntos
Angiotensina II , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Arteriosclerose/induzido quimicamente , Arteriosclerose/patologia , Enalapril/farmacologia , Vasculite/induzido quimicamente , Vasculite/patologia , Animais , Aorta/metabolismo , Aneurisma Aórtico/induzido quimicamente , Aneurisma Aórtico/prevenção & controle , Apolipoproteínas E/deficiência , Moléculas de Adesão Celular/genética , Quimiocinas/metabolismo , Endotélio/metabolismo , Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , PPAR alfa/genética , PPAR gama/genética , RNA Mensageiro/metabolismo , Regulação para Cima
14.
Thromb Haemost ; 92(5): 956-65, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15543321

RESUMO

During thrombosis, P-selectin is expressed on the surface of activated endothelial cells and platelets. We hypothesized that targeting a plasminogen activator (PA) to P-selectin would enhance local thrombolysis and reduce bleeding risk. Previously, a urokinase (uPA)/anti-P-selectin antibody (HuSZ51) fusion protein was shown to increase fibrinolysis in a hamster pulmonary embolism model. To explore the therapeutic potential of this targeting strategy, we fused the fibrin-selective Desmodus rotundus salivary PA alpha1 (dsPA alpha 1) to HuSZ51 and compared the fibrinolytic activity of P-selectin-targeted dsPA alpha 1 (HuSZ51-dsPA alpha 1) to unmodified dsPA alpha 1 in vitro and in vivo. HuSZ51-dsPA alpha 1 and dsPA alpha 1 were expressed in CHO cells and purified to homogeneity by affinity chromatography. HuSZ51-dsPA alpha 1 bound to thrombin-activated human and dog platelets with comparable affinities to that of parental antibody SZ51. The fusion protein retained the catalytic activities of dsPA alpha 1 in chromogenic and clot lysis assays, indicating that dsPA alpha 1 is fully functional when fused to HuSZ51. Compared to dsPA alpha 1, HuSZ51-dsPA alpha 1 had similar thrombolytic efficacy in a rat pulmonary embolism model and anti-thrombotic potency in a dog model of femoral artery thrombosis. However, HuSZ51-dsPA alpha 1 was less effective in lysis of preexisting arterial thrombi in the dog model. The reduced arterial thrombolysis was not due to the pharmacokinetic properties of HuSZ51-dsPA alpha 1 because antigen level and amidolytic activity were higher in plasma from HuSZ51-dsPA alpha 1-treated groups than corresponding dsPA alpha 1-treated groups. These data indicate that the thrombolytic efficacy of HuSZ51-dsPA alpha 1 varied dependent on the physical composition of thrombi. The lack of stimulation by fibrin in arterial thrombi may contribute to the attenuated thrombolytic efficacy of HuSZ51-dsPA alpha 1 in the dog model.


Assuntos
Plaquetas/efeitos dos fármacos , Fibrina/efeitos dos fármacos , Imunoconjugados/uso terapêutico , Selectina-P/imunologia , Ativadores de Plasminogênio/administração & dosagem , Terapia Trombolítica/métodos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/uso terapêutico , Plaquetas/química , Plaquetas/fisiologia , Modelos Animais de Doenças , Cães , Fibrinolíticos/administração & dosagem , Fibrinolíticos/farmacocinética , Fibrinolíticos/farmacologia , Engenharia Genética , Humanos , Imunoconjugados/farmacocinética , Imunoconjugados/farmacologia , Ativadores de Plasminogênio/genética , Ativadores de Plasminogênio/uso terapêutico , Agregação Plaquetária , Embolia Pulmonar/tratamento farmacológico , Ratos , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Trombina , Trombose/tratamento farmacológico , Resultado do Tratamento
15.
Can J Physiol Pharmacol ; 82(6): 372-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15381960

RESUMO

The present study was designed to evaluate the role of endothelial NO in the hemodynamics and vascular changes that occur in heart failure following myocardial infarction in rats. Left ventricular systolic pressure (LVSP), mean blood pressure (MBP), aortic morphology (media thickness) and reactivity were evaluated in rats with coronary artery ligation (heart failure, HF) or sham operation (SO) untreated or treated for four weeks with either a low dose of NG-nitro-L-arginine methyl ester (L-NAME, 6 mg.kg(-1).day(-1)) or L-arginine (1.5 g.kg(-1).day(-1)). In rats with HF LVSP (HF = 111 +/- 8 mmHg; SO = 143 +/- 6 mmHg, p < 0.05), MBP (HF = 98 +/- 8 mmHg; SO = 127 +/- 6 mmHg, p < 0.05) and aortic media thickness (HF = 68 +/- 6 microm; SO = 75 +/- 2 microm, p < 0.05) were significantly reduced. The contractile response to phenylephrine and the endothelium-independent relaxation to sodium nitroprusside were similar in HF and SO aortas, but the sensitivity (pD2) to acetylcholine (HF = 7.5 +/- 0.06; SO = 7.1 +/- 0.08, p < 0.05) was significantly increased in HF aortas, indicating an enhanced basal NO release. Treatment with L-NAME (LN) reversed the effects of HF on LVSP (HF-LN = 143 +/- 9 mmHg, p < 0.05 vs. HF), MBP (HF-LN = 128 +/- 8 mmHg, p < 0.05 vs. HF), sensitivity to acetylcholine (HF-LN = 6.9 +/- 0.10, p < 0.05 vs. HF) and aortic media thickness (HF-LN = 79 +/- 2 microm, p < 0.05 vs. HF), without changing these parameters in SO rats. L-NAME also selectively increased the maximal response to phenylephrine in HF aortas (HF-LN = 2.4 +/- 0.20 g; HF = 1.6 +/- 0.17 g, p < 0.05). L-arginine (LA) did not change the effects of HF on LSVP, MBP or aortic media thickness, but it reduced the sensitivity to phenylephrine in aortas from SO rats (SO-LA = 6.5 +/- 0.12; SO = 7.0 +/- 0.09, p < 0.05). Taken together, these results suggest an important role for endothelial NO in mediating the reduced vascular growth, myocardial dysfunction and hypotension in rats with HF.


Assuntos
Pressão Sanguínea/fisiologia , Insuficiência Cardíaca/fisiopatologia , Frequência Cardíaca/fisiologia , Óxido Nítrico/fisiologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiologia , Pressão Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Frequência Cardíaca/efeitos dos fármacos , Técnicas In Vitro , Masculino , Nitroprussiato/farmacologia , Ratos , Ratos Wistar , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
16.
Thromb Res ; 111(6): 381-7, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14698657

RESUMO

In this study, we investigated if elevation of endogenous plasminogen activator inhibitor type 1 (PAI-1) by lipopolysaccharide (LPS) can retard thrombolysis in both a rat model of lung vasculature fibrin deposition and a platelet-rich thrombus model induced by endothelial injury. By 3 h following an intravenous bolus injection of 0.5 mg/kg LPS, the plasma PAI-1 level had increased to approximately 8 ng/ml. 125I-labeled fibrinogen was injected intravenously followed by an injection of batroxobin. Batroxobin converts fibrinogen into insoluble fibrin, which was then deposited in the lungs within 5 min, followed by spontaneous fibrinolysis that completely cleared fibrin deposition in the lungs by 30 min. In rats pre-treated with LPS, spontaneous fibrinolysis was significantly retarded. In the endothelial injury model, topical application of FeCl2 on the carotid artery induced an occlusive platelet-rich thrombus, which was not sensitive to endogenous thrombolysis. Exogenous tissue-type plasminogen activator (tPA) was required to recanalize the occlusive thrombus in a dose-dependent manner. Pre-treatment with LPS did not alter the dose-response curve of exogenous tPA-induced thrombolysis. These data indicate that batroxobin-induced lung vasculature fibrin deposition in rats, unlike the FeCl2 model, is sensitive to the impact of endogenous PAI-1 on fibrinolysis.


Assuntos
Trombose das Artérias Carótidas/etiologia , Fibrina/metabolismo , Lipopolissacarídeos/farmacologia , Pneumopatias/etiologia , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Animais , Batroxobina , Artérias Carótidas/patologia , Compostos Ferrosos , Fibrina/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Ativador de Plasminogênio Tecidual/metabolismo , Ativador de Plasminogênio Tecidual/farmacologia
17.
Thromb Haemost ; 90(3): 414-21, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12958609

RESUMO

Studies have shown that inhibition of TAFI by small peptides enhances pharmacological effects of tPA in animal models of thrombosis, suggesting that TAFI modulates the fibrinolytic system. In this study, we investigated the effect of activated human TAFI (TAFIa) on endogenous fibrinolysis in a rat model of intravascular fibrin deposition. (125)I-labeled fibrinogen was injected intravenously followed by a bolus injection of batroxobin, a thrombin-like enzyme. Batroxobin cleaved fibrinogen to form insoluble fibrin that was deposited in tissues, including the lungs. This was shown by a decrease of radioactivity in the blood as a result of consumption of (125)I-labeled fibrinogen and an elevation of radioactivity in the lungs 5 min following batrox-obin administration. Endogenous fibrinolysis was detected by a gradual increase in radioactivity in the blood and a decrease in radioactivity in the lungs at 30 min, an indication of radiolabeled fibrin degradation products (FDPs) being released into the circulation from the tissues. Intravenous administration of human TAFIa dose-dependently attenuated the later phase reduction of radioactivity in the lungs. When the dose of TAFIa was 218 micro g/kg, giving a peak plasma level of TAFIa 0.9 +/- 0.05 micro g/ml, the spontaneous fibrinolysis was completely prevented. These results provide direct evidence that an increase in circulating TAFIa impairs endogenous clot lysis in a rat model of fibrin deposition.


Assuntos
Batroxobina/farmacologia , Carboxipeptidase B2/farmacologia , Fibrina/metabolismo , Fibrinólise/efeitos dos fármacos , Pneumopatias/patologia , Animais , Carboxipeptidase B2/sangue , Carboxipeptidase B2/fisiologia , Relação Dose-Resposta a Droga , Fibrinogênio/farmacocinética , Humanos , Injeções Intravenosas , Radioisótopos do Iodo/farmacocinética , Masculino , Ratos , Trombose/etiologia
18.
Arterioscler Thromb Vasc Biol ; 23(9): 1627-32, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12855485

RESUMO

OBJECTIVE: Angiotensin II (Ang II) promotes vascular inflammation, accelerates atherosclerosis, and induces abdominal aortic aneurysm (AAA). These changes were associated with activation of nuclear factor (NF)-kappaB-mediated induction of proinflammatory genes. The incidence of AAA in this model was higher in male than in female mice, and the vascular effects of estrogen may be associated with anti-inflammatory actions. The present study was undertaken to test the hypothesis that estrogen can attenuate Ang II-induced AAA in apolipoprotein E-deficient mice via its anti-inflammatory mechanism. METHODS AND RESULTS: Infusion of Ang II (1.44 mg/kg per d for 1 month) induced AAA in 90% of the animals (n=20) with an expansion of the suprarenal aorta (diameter 1.9+/-0.14 mm versus <1 mm in normal mice). In mice treated with 17beta-estradiol (E2, 0.25-mg subcutaneous pellets), Ang II induced AAA only in 42% of the animals (n=19) with a significant reduction of average diameters of the suprarenal aorta (1.5+/-0.14 mm). E2 also decreased the expressions of intracellular adhesion molecule-1, vascular cellular adhesion molecule-1, E-selectin, monocyte chemotactic protein-1, and macrophage-colony stimulating factor in the aorta. CONCLUSIONS: These data suggest that attenuation of AAA by E2 is associated with inhibition of proinflammatory gene expression.


Assuntos
Angiotensina II/farmacologia , Aneurisma da Aorta Abdominal/etiologia , Aneurisma da Aorta Abdominal/prevenção & controle , Apolipoproteínas E/deficiência , Estradiol/farmacologia , Animais , Aneurisma da Aorta Abdominal/patologia , Modelos Animais de Doenças , Estradiol/uso terapêutico , Feminino , Inflamação/etiologia , Inflamação/prevenção & controle , Masculino , Camundongos
19.
Am J Physiol Regul Integr Comp Physiol ; 283(6): R1442-9, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12388474

RESUMO

Cardiovascular diseases, such as atherosclerosis and hypertension, are associated with arterial stiffening. Previous studies showed that ANG II exacerbated atherosclerosis and induced hypertension and aneurysm formation in apolipoprotein E-deficient (apoE-KO) mice. The aim of the present study was to examine the effects of chronic treatment of ANG II on the arterial elastic properties in apoE-KO mice. We hypothesized that ANG II will injure the arterial wall resulting in increased arterial stiffening. Male apoE-KO mice were infused with either ANG II (1.44 mg. kg(-1). day(-1)) or vehicle (PBS) for 30 days. ANG II treatment accelerated atherosclerosis in the carotid artery by sixfold (P < 0.001) and increased blood pressure by 30% (P < 0.05). Additionally, our data demonstrated that ANG II increased arterial stiffening using both in vivo and in vitro methods. ANG II significantly increased pulse wave velocity by 36% (P < 0.01) and decreased arterial elasticity as demonstrated by a more than 900% increase in maximal stiffening (high strain Young's modulus) compared with vehicle (P < 0.05). These functional changes were correlated with morphological and biochemical changes as demonstrated by an increase in collagen content (60%), a decrease in elastin content (74%), and breaks in the internal elastic lamina in the aortic wall. In addition, endothelium-independent vasorelaxation to sodium nitroprusside was impaired in the aortic rings of ANG II-treated mice compared with vehicle. Thus, the present data indicate that ANG II injures the artery wall in multiple ways and arterial stiffening may be a common outcome of ANG II-induced arterial damage.


Assuntos
Angiotensina II/toxicidade , Aorta/efeitos dos fármacos , Aorta/patologia , Apolipoproteínas E/deficiência , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/patologia , Acetilcolina/farmacologia , Angiotensina II/administração & dosagem , Animais , Aneurisma da Aorta Abdominal/induzido quimicamente , Apolipoproteínas E/biossíntese , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Arteriosclerose/induzido quimicamente , Pressão Sanguínea/efeitos dos fármacos , Colágeno/análise , Esquema de Medicação , Elasticidade/efeitos dos fármacos , Elastina/análise , Endotélio Vascular/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Nitroprussiato/farmacologia
20.
Atherosclerosis ; 162(1): 23-31, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11947894

RESUMO

Low density lipoprotein receptor deficient (LDLR-KO) and apolipoprotein E deficient (apo E-KO) mice both develop hyperlipidemia and atherosclerosis by different mechanisms. The aim of the present study was to compare the effects of simvastatin on cholesterol levels, endothelial dysfunction, and aortic lesions in these two models of experimental atherosclerosis. Male LDLR-KO mice fed a high cholesterol (HC; 1%) diet developed atherosclerosis at 8 months of age with hypercholesterolemia. The addition of simvastatin (300 mg/kg daily) to the HC diet for 2 more months lowered total cholesterol levels by approximately 57% and reduced aortic plaque area by approximately 15% compared with the LDLR-KO mice continued on HC diet alone, P<0.05. Simvastatin treatment also improved acetylcholine (ACh)-induced endothelium-dependent vasorelaxation in isolated aortic rings, which was associated with an increase in NOS-3 expression by approximately 88% in the aorta measured by real time polymerase chain reaction (PCR), P<0.05. In contrast, in age-matched male apo E-KO mice fed a normal diet, the same treatment of simvastatin elevated serum total cholesterol by approximately 35%, increased aortic plaque area by approximately 15%, and had no effect on endothelial function. These results suggest that the therapeutic effects of simvastatin may depend on the presence of a functional apolipoprotein E.


Assuntos
Apolipoproteínas E/efeitos dos fármacos , Apolipoproteínas E/deficiência , Arteriosclerose/tratamento farmacológico , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Sinvastatina/uso terapêutico , Acetilcolina/farmacologia , Animais , Aorta/patologia , Arteriosclerose/sangue , Arteriosclerose/etiologia , HDL-Colesterol/sangue , HDL-Colesterol/efeitos dos fármacos , LDL-Colesterol/sangue , LDL-Colesterol/efeitos dos fármacos , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Hipercolesterolemia/sangue , Hipercolesterolemia/tratamento farmacológico , Hipercolesterolemia/etiologia , Masculino , Camundongos , Camundongos Knockout , Modelos Cardiovasculares , Relaxamento Muscular/efeitos dos fármacos , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , RNA Mensageiro/biossíntese , RNA Mensageiro/efeitos dos fármacos , Receptores de LDL/deficiência , Receptores de LDL/efeitos dos fármacos , Resultado do Tratamento , Triglicerídeos/sangue , Vasodilatadores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...