Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Proc Natl Acad Sci U S A ; 109(17): 6727-32, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22493243

RESUMO

Suppression of cyclooxygenase 2 (COX-2)-derived prostacyclin (PGI(2)) is sufficient to explain most elements of the cardiovascular hazard from nonsteroidal antinflammatory drugs (NSAIDs). However, randomized trials are consistent with the emergence of cardiovascular risk during chronic dosing with NSAIDs. Although deletion of the PGI(2) receptor fosters atherogenesis, the importance of COX-2 during development has constrained the use of conventional knockout (KO) mice to address this question. We developed mice in which COX-2 was deleted postnatally, bypassing cardiorenal defects exhibited by conventional KOs. When crossed into ApoE-deficient hyperlipidemic mice, COX-2 deletion accelerated atherogenesis in both genders, with lesions exhibiting leukocyte infiltration and phenotypic modulation of vascular smooth muscle cells, as reflected by loss of α-smooth muscle cell actin and up-regulation of vascular cell adhesion molecule-1. Stimulated peritoneal macrophages revealed suppression of COX-2-derived prostanoids and augmented 5-lipoxygenase product formation, consistent with COX-2 substrate rediversion. Although deletion of the 5-lipoxygenase activating protein (FLAP) did not influence atherogenesis, it attenuated the proatherogeneic impact of COX-2 deletion in hyperlipidemic mice. Chronic administration of NSAIDs may increasingly confer a cardiovascular hazard on patients at low initial risk. Promotion of atherogenesis by postnatal COX-2 deletion affords a mechanistic explanation for this observation. Coincident inhibition of FLAP may offer an approach to attenuating such a risk from NSAIDs.


Assuntos
Araquidonato 5-Lipoxigenase/metabolismo , Aterosclerose/metabolismo , Ciclo-Oxigenase 2/metabolismo , Animais , Aterosclerose/enzimologia , Ciclo-Oxigenase 2/genética , Camundongos , Camundongos Knockout , Especificidade por Substrato
2.
Circulation ; 121(24): 2654-60, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20530000

RESUMO

BACKGROUND: Although the dominant product of vascular Cyclooxygenase-2 (COX-2), prostacyclin (PGI(2)), restrains atherogenesis, inhibition and deletion of COX-2 have yielded conflicting results in mouse models of atherosclerosis. Floxed mice were used to parse distinct cellular contributions of COX-2 in macrophages and T cells (TCs) to atherogenesis. METHODS AND RESULTS: Deletion of macrophage-COX-2 (Mac-COX-2KOs) was attained with LysMCre mice and completely suppressed lipopolysaccharide-stimulated macrophage prostaglandin (PG) formation and lipopolysaccharide-evoked systemic PG biosynthesis by approximately 30%. Lipopolysaccharide-stimulated COX-2 expression was suppressed in polymorphonuclear leukocytes isolated from MacKOs, but PG formation was not even detected in polymorphonuclear leukocyte supernatants from control mice. Atherogenesis was attenuated when MacKOs were crossed into hyperlipidemic low-density lipoprotein receptor knockouts. Deletion of Mac-COX-2 appeared to remove a restraint on COX-2 expression in lesional nonleukocyte (CD45- and CD11b-negative) vascular cells that express vascular cell adhesion molecule and variably alpha-smooth muscle actin and vimentin, portending a shift in PG profile and consequent atheroprotection. Basal expression of COX-2 was minimal in TCs, but use of CD4Cre to generate TC knockouts depressed its modest upregulation by anti-CD3epsilon. However, biosynthesis of PGs, TC composition in lymphatic organs, and atherogenesis in low-density lipoprotein receptor knockouts were unaltered in TC knockouts. CONCLUSIONS: Macrophage-COX-2, primarily a source of thromboxane A(2) and prostaglandin (PG)E(2), promotes atherogenesis and exerts a restraint on enzyme expression by lesional cells suggestive of vascular smooth muscle cells, a prominent source of atheroprotective prostacyclin. TC COX-2 does not detectably influence TC development or function or atherogenesis in mice.


Assuntos
Aterosclerose/genética , Ciclo-Oxigenase 2/genética , Deleção de Genes , Animais , Aterosclerose/metabolismo , Aterosclerose/patologia , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Feminino , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Prostaglandinas/metabolismo , RNA Mensageiro/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Linfócitos T/metabolismo , Linfócitos T/patologia
3.
Arterioscler Thromb Vasc Biol ; 29(7): 1039-45, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19390055

RESUMO

OBJECTIVE: In dyslipidemic states, platelets become hyperreactive, secreting molecules that promote atherosclerosis. We have shown that the semaphorin family member, sema4D (CD100), is expressed on the surface of platelets and proposed that its role includes promoting thrombus growth by binding to nearby platelets and endothelial cells, both of which express sema4D receptors. Here we tested the hypothesis that deleting sema4D will attenuate the adverse consequences of dyslipidemia on platelets and the vessel wall. METHODS AND RESULTS: Platelet function and atherosclerotic lesion formation were measured in LDLR(-/-) and sema4D(-/-)LDLR(-/-) mice after 6 months on a high-fat diet. All of the mice developed the dyslipidemia expected on this diet in the absence of functional LDL receptors. However, when compared to LDLR(-/-) mice, sema4D(-/-) LDLR(-/-) mice had reduced lipid deposition in the descending aorta, a 6-fold decrease in the frequency of arterial occlusion and a reduction to near wild-type levels in the accumulation of platelets after injury. These differences were retained ex vivo, with a marked decrease in platelet accumulation on collagen under flow and in platelet aggregation. CONCLUSIONS: These results show that loss of sema4D expression reduces the platelet hyperactivity otherwise found in dyslipidemia, and confers protection against the development of atherosclerosis.


Assuntos
Antígenos CD/fisiologia , Aterosclerose/fisiopatologia , Hiperlipidemias/fisiopatologia , Agregação Plaquetária/fisiologia , Semaforinas/fisiologia , Animais , Aorta Torácica/patologia , Aterosclerose/sangue , Aterosclerose/etiologia , Dieta Aterogênica , Hiperlipidemias/sangue , Hiperlipidemias/complicações , Masculino , Camundongos , Camundongos Knockout , Trombose/fisiopatologia
4.
Sci Transl Med ; 4(132): 132ra54, 2012 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-22553252

RESUMO

Prostacyclin (PGI(2)) is a vasodilator and platelet inhibitor, properties consistent with cardioprotection. More than a decade ago, inhibition of cyclooxygenase-2 (COX-2) by the nonsteroidal anti-inflammatory drugs (NSAIDs) rofecoxib and celecoxib was found to reduce the amount of the major metabolite of PGI(2) (PGI-M) in the urine of healthy volunteers. This suggested that NSAIDs might cause adverse cardiovascular events by reducing production of cardioprotective PGI(2). This prediction was based on the assumption that the concentration of PGI-M in urine likely reflected vascular production of PGI(2) and that other cardioprotective mediators, especially nitric oxide (NO), were not able to compensate for the loss of PGI(2). Subsequently, eight placebo-controlled clinical trials showed that NSAIDs that block COX-2 increase adverse cardiovascular events. We connect tissue-specific effects of NSAID action and functional correlates in mice with clinical outcomes in humans by showing that deletion of COX-2 in the mouse vasculature reduces excretion of PGI-M in urine and predisposes the animals to both hypertension and thrombosis. Furthermore, vascular disruption of COX-2 depressed expression of endothelial NO synthase and the consequent release and function of NO. Thus, suppression of PGI(2) formation resulting from deletion of vascular COX-2 is sufficient to explain the cardiovascular hazard from NSAIDs, which is likely to be augmented by secondary mechanisms such as suppression of NO production.


Assuntos
Pressão Sanguínea/fisiologia , Vasos Sanguíneos/enzimologia , Vasos Sanguíneos/fisiopatologia , Ciclo-Oxigenase 2/metabolismo , Trombose/enzimologia , Trombose/fisiopatologia , Animais , Epoprostenol/biossíntese , Deleção de Genes , Homeostase , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Especificidade de Órgãos , Sístole
5.
J Clin Invest ; 122(4): 1459-68, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22406532

RESUMO

The clinical use of niacin to treat dyslipidemic conditions is limited by noxious side effects, most commonly facial flushing. In mice, niacin-induced flushing results from COX-1-dependent formation of PGD2 and PGE2 followed by COX-2-dependent production of PGE2. Consistent with this, niacin-induced flushing in humans is attenuated when niacin is combined with an antagonist of the PGD2 receptor DP1. NSAID-mediated suppression of COX-2-derived PGI2 has negative cardiovascular consequences, yet little is known about the cardiovascular biology of PGD2. Here, we show that PGD2 biosynthesis is augmented during platelet activation in humans and, although vascular expression of DP1 is conserved between humans and mice, platelet DP1 is not present in mice. Despite this, DP1 deletion in mice augmented aneurysm formation and the hypertensive response to Ang II and accelerated atherogenesis and thrombogenesis. Furthermore, COX inhibitors in humans, as well as platelet depletion, COX-1 knockdown, and COX-2 deletion in mice, revealed that niacin evoked platelet COX-1-derived PGD2 biosynthesis. Finally, ADP-induced spreading on fibrinogen was augmented by niacin in washed human platelets, coincident with increased thromboxane (Tx) formation. However, in platelet-rich plasma, where formation of both Tx and PGD2 was increased, spreading was not as pronounced and was inhibited by DP1 activation. Thus, PGD2, like PGI2, may function as a homeostatic response to thrombogenic and hypertensive stimuli and may have particular relevance as a constraint on platelets during niacin therapy.


Assuntos
Plaquetas/enzimologia , Ciclo-Oxigenase 1/fisiologia , Proteínas de Membrana/fisiologia , Prostaglandina D2/biossíntese , 6-Cetoprostaglandina F1 alfa/análogos & derivados , 6-Cetoprostaglandina F1 alfa/biossíntese , 6-Cetoprostaglandina F1 alfa/urina , Difosfato de Adenosina/farmacologia , Angioplastia Coronária com Balão/efeitos adversos , Animais , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/metabolismo , Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Plaquetas/efeitos dos fármacos , Plaquetas/ultraestrutura , Trombose das Artérias Carótidas/etiologia , Trombose das Artérias Carótidas/metabolismo , Trombose das Artérias Carótidas/prevenção & controle , Ciclo-Oxigenase 1/sangue , Ciclo-Oxigenase 1/deficiência , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/deficiência , Ciclo-Oxigenase 2/genética , Inibidores de Ciclo-Oxigenase/farmacologia , Método Duplo-Cego , Endotélio Vascular/lesões , Endotélio Vascular/metabolismo , Feminino , Humanos , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Hipertensão/prevenção & controle , Masculino , Proteínas de Membrana/sangue , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Ativação Plaquetária/fisiologia , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Prostaglandina D2/fisiologia , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/sangue , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/fisiologia , Receptores de LDL/deficiência , Receptores Nicotínicos/biossíntese , Receptores Nicotínicos/sangue , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Prostaglandina/deficiência
6.
J Immunol ; 169(2): 1014-20, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12097409

RESUMO

The hallmarks of chronic, severe asthma include prominent airway inflammation and airway smooth muscle (ASM) hypertrophy and hyperplasia. One of the factors that contribute to the injury and repair process within the airway is activation of proteases and turnover of extracellular matrix components. Mast cells, which are present in increased numbers in the asthmatic airway, are a rich source of the neutral protease chymase, which can degrade several basement membrane components. Recent data suggest that proteases also play a critical role in regulating the expression of CD44, the primary receptor for the matrix glycosaminoglycan hyaluronan. In this study we investigated the effects of chymase treatment on human ASM cell function. We found that chymase degraded the smooth muscle cell pericellular matrix. This was accompanied by an increased release of fibronectin and soluble CD44, but not soluble ICAM-1 or soluble hyaluronan, into the conditioned medium. In addition, chymase inhibited T cell adhesion to ASM and dramatically reduced epidermal growth factor-induced smooth muscle cell proliferation. These data suggest that the local release of mast cell chymase may have profound effects on ASM cell function and airway remodeling.


Assuntos
Comunicação Celular/fisiologia , Matriz Extracelular/metabolismo , Inibidores do Crescimento/fisiologia , Mastócitos/enzimologia , Mitógenos/farmacologia , Músculo Liso/enzimologia , Serina Endopeptidases/fisiologia , Traqueia/enzimologia , Adesão Celular/fisiologia , Células Cultivadas , Quimases , Regulação para Baixo , Fator de Crescimento Epidérmico/farmacologia , Matriz Extracelular/enzimologia , Matriz Extracelular/patologia , Inibidores do Crescimento/farmacologia , Humanos , Receptores de Hialuronatos/metabolismo , Mitógenos/antagonistas & inibidores , Músculo Liso/citologia , Serina Endopeptidases/farmacologia , Transdução de Sinais/fisiologia , Solubilidade , Traqueia/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA