Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Mol Cell Cardiol ; 137: 71-81, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31634485

RESUMO

Mutations in the PTPN11 gene, which encodes the protein tyrosine phosphatase Shp2, cause Noonan syndrome and LEOPARD syndrome, inherited multifaceted diseases including cardiac and vascular defects. However, the function of Shp2 in blood vessels, especially in vascular smooth muscle cells (VSMCs), remains largely unknown. We generated mice in which Shp2 was specifically deleted in VSMCs and embryonic cardiomyocytes using the SM22α-Cre transgenic mouse line. Conditional Shp2 knockout resulted in massive hemorrhage, cardiovascular defects and embryonic lethality at the late embryonic developmental stage (embryonic date 16.5). The thinning of artery walls in Shp2-knockout embryos was due to decreased VSMC number and reduced extracellular matrix deposition. Myocyte proliferation was decreased in Shp2-knockout arteries and hearts. Importantly, cardiomyocyte-specific Shp2-knockout did not cause similar vascular defects. Shp2 was required for TGFß1-induced expression of ECM components, including collagens in VSMCs. In addition, collagens were sufficient to promote Shp2-inefficient VSMC proliferation. Finally, Shp2 was deleted in adult mouse VSMCs by using SMMHC-CreERT2 and tamoxifen induction. Shp2 deletion dramatically inhibited the expression of ECM components, proliferation of VSMCs and neointima formation in a carotid artery ligation model. Therefore, Shp2 is required for myocyte proliferation in cardiovascular development and vascular remodeling through TGFß1-regulated collagen synthesis.


Assuntos
Coração/embriologia , Miócitos de Músculo Liso/enzimologia , Neointima/embriologia , Neointima/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Animais , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/embriologia , Artérias Carótidas/patologia , Proliferação de Células/efeitos dos fármacos , Colágeno/farmacologia , Ciclina D1/metabolismo , Embrião de Mamíferos/patologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Feminino , Hemorragia/patologia , Integrases/metabolismo , Masculino , Camundongos Knockout , Músculo Liso Vascular/patologia , Miocárdio/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/deficiência , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo
2.
Exp Mol Med ; 51(7): 1-13, 2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31292433

RESUMO

We hypothesized that NADPH oxidase 4 (Nox4) is involved in the formation of neointimal atherosclerotic plaques through the migration of smooth muscle cells (SMCs) in response to flagellin. Here, we demonstrate that TLR5-mediated Nox4 activation regulates the migration of SMCs, leading to neointimal plaque formation in atherosclerosis. To investigate the molecular mechanism by which the TLR5-Nox4 cascade mediates SMC migration, we analyzed the signaling cascade in primary vascular SMCs (VSMCs) from wild-type (WT) or Nox4 KO mice. Stimulation of VSMCs from Nox4 KO mice with flagellin failed to induce H2O2 production and Rac activation compared with stimulation of VSMCs from WT mice. Moreover, the migration of Nox4-deficient VSMCs was attenuated in response to flagellin in transwell migration and wound healing assays. Finally, we performed partial carotid artery ligation in ApoE KO and Nox4ApoE DKO mice fed a high-fat diet (HFD) with or without recombinant FliC (rFliC) injection. Injection of rFliC into ApoE KO mice fed a HFD resulted in significantly increased SMC migration into the intimal layer, whereas SMC accumulation was not detected in Nox4ApoE DKO mice. We conclude that activation of the TLR5-Nox4 cascade plays an important role in the formation of neointimal atherosclerotic plaques.


Assuntos
Aterosclerose/enzimologia , Flagelina/administração & dosagem , NADPH Oxidase 4/metabolismo , Placa Aterosclerótica/enzimologia , Transdução de Sinais , Receptor 5 Toll-Like/metabolismo , Animais , Aterosclerose/patologia , Artérias Carótidas/patologia , Movimento Celular , Quimiocinas/análise , Dieta Hiperlipídica/efeitos adversos , Flagelina/genética , Masculino , Camundongos Knockout para ApoE , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , NADPH Oxidase 4/genética , Neointima/enzimologia , Neointima/patologia , Fenótipo , Placa Aterosclerótica/patologia , Salmonella enteritidis/genética , Receptor 5 Toll-Like/genética
3.
Int J Exp Pathol ; 100(2): 94-101, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31058412

RESUMO

Vascular surgical interventions are often burdened with late complications, including thrombosis or restenosis. The latter is generally caused by neointimal hyperplasia. Although extracellular matrix (ECM) remodelling is an important part of neointima formation, this process is not clearly understood. The aim of the study was to assess the content and activity of membrane-type 1 matrix metalloproteinase in human neointima in the late stages of its development. Matrix metalloproteinase-2 and tissue inhibitor of matrix metalloproteinase-2 were also evaluated. The research was performed on neointima samples collected during secondary vascular interventions from patients with chronic limb ischaemia who developed vascular occlusion at 6-18 months after aorto/ilio-femoral bypass grafting. The control material consisted of segments of femoral arteries collected from organ donors. Western blot and/or ELISA were used for the determination of MT1-MMP and TIMP-2 expression. The activity of MT1-MMP was measured by fluorometric assay and that of MMP-2 by zymography. We demonstrated significantly increased MT1-MMP protein content in neointima when compared to normal arteries. However, the activity of MT1-MMP was significantly lower in neointima than in control samples. The decreased MT1-MMP activity was concomitant with reduced activity of MMP-2. The TIMP-2 protein levels in neointima and normal arteries were not significantly different. The results of our study suggest that the reduced activity of MT1-MMP and consequently MMP-2 in human neointima may play a role in decreased degradation of ECM components and thus promote neointimal overgrowth.


Assuntos
Arteriopatias Oclusivas/cirurgia , Implante de Prótese Vascular/efeitos adversos , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Neointima/enzimologia , Neointima/patologia , Aorta/cirurgia , Artéria Femoral/enzimologia , Artéria Femoral/cirurgia , Oclusão de Enxerto Vascular/enzimologia , Oclusão de Enxerto Vascular/cirurgia , Humanos , Hiperplasia/enzimologia , Artéria Ilíaca/cirurgia , Perna (Membro)/irrigação sanguínea , Reoperação , Inibidor Tecidual de Metaloproteinase-2/metabolismo
4.
Mol Med Rep ; 18(1): 1007-1014, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29845301

RESUMO

Resveratrol has been reported to inhibit vascular smooth muscle cell proliferation and neointimal hyperplasia following arterial injury; however, the underlying mechanisms remain unclear. The present study was designed to investigate the effects of resveratrol on angiotensin II (AngII)­induced proliferation of A7r5 cells and explore the molecular mechanisms responsible for the observed effects. Resveratrol inhibited cell proliferation and migration, and decreased the AngII­induced protein expression of α­smooth muscle actin (α­SMA), proliferating cell nuclear antigen (PCNA) and cyclin­dependent kinase 4 (CDK4). Resveratrol inhibited AngII­induced activation of intracellular Ca2+/calmodulin­dependent protein kinase II (CaMKII) and histone deacetylases 4 (HDAC4), as well as blocking AngII­induced cell cycle progression from the G0/G1 to S­phase. In vivo, 4­weeks of resveratrol treatment decreased the neointima area and the neointima/media area ratio in rats following carotid balloon injury. Resveratrol also inhibited the protein expression of total and phosphorylated CaMKII and HDAC4 in the injured arteries. In conclusion, the present study demonstrated that resveratrol attenuated AngII­induced cell proliferation and neointimal hyperplasia by inhibiting the CaMKII­HDAC4 signaling pathway. These findings suggest that resveratrol may potentially prevent arterial restenosis.


Assuntos
Angiotensina II/efeitos adversos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Ciclo Celular/efeitos dos fármacos , Histona Desacetilases/metabolismo , Neointima/enzimologia , Transdução de Sinais/efeitos dos fármacos , Estilbenos/farmacologia , Angiotensina II/farmacologia , Animais , Linhagem Celular , Hiperplasia , Masculino , Neointima/patologia , Ratos , Ratos Sprague-Dawley , Resveratrol
5.
Biosci Rep ; 37(5)2017 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-28851839

RESUMO

Inflammatory cells play key roles in restenosis upon vascular surgical procedures such as bypass grafts, angioplasty and stent deployment but the molecular mechanisms by which these cells affect restenosis remain unclear. The p110δ isoform of phosphoinositide 3-kinase (PI3K) is mainly expressed in white blood cells. Here, we have investigated whether p110δ PI3K is involved in the pathogenesis of restenosis in a mouse model of carotid injury, which mimics the damage following arterial grafts. We used mice in which p110δ kinase activity has been disabled by a knockin (KI) point mutation in its ATP-binding site (p110δD910A/D910A PI3K mice). Wild-type (WT) and p110δD910A/D910A mice were subjected to longitudinal carotid injury. At 14 and 30 days after carotid injury, mice with inactive p110δ showed strongly decreased infiltration of inflammatory cells (including T lymphocytes and macrophages) and vascular smooth muscle cells (VSMCs), compared with WT mice. Likewise, PI-3065, a p110δ-selective PI3K inhibitor, almost completely prevented restenosis after artery injury. Our data showed that p110δ PI3K plays a main role in promoting neointimal thickening and inflammatory processes during vascular stenosis, with its inhibition providing significant reduction in restenosis following carotid injury. p110δ-selective inhibitors, recently approved for the treatment of human B-cell malignancies, therefore, present a new therapeutic opportunity to prevent the restenosis upon artery injury.


Assuntos
Lesões das Artérias Carótidas/enzimologia , Estenose das Carótidas/enzimologia , Classe I de Fosfatidilinositol 3-Quinases/imunologia , Inflamação/enzimologia , Animais , Artérias Carótidas/enzimologia , Artérias Carótidas/imunologia , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/imunologia , Lesões das Artérias Carótidas/patologia , Estenose das Carótidas/genética , Estenose das Carótidas/imunologia , Estenose das Carótidas/patologia , Classe I de Fosfatidilinositol 3-Quinases/genética , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Masculino , Camundongos Endogâmicos C57BL , Neointima/enzimologia , Neointima/genética , Neointima/imunologia , Neointima/patologia , Mutação Puntual
6.
Biomed Pharmacother ; 94: 430-438, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28778046

RESUMO

Salicornia europaea L. (SE) has been used as folk medicine for the treatment of various diseases such as obesity, diabetes, and cancer. However, its effects on atherosclerotic events in vascular smooth muscle cells (VSMCs) remain unknown. The present study explored the effects of the ethyl acetate fraction of desalted SE hot water extract (SEWEAF) on atherosclerotic responses (especially migration and proliferation) in VSMCs and vascular neointima formation. Treatment with the SEWEAF significantly suppressed the platelet-derived growth factor (PDGF)-BB-induced VSMC migration and proliferation as well the phosphorylation of mitogen-activated protein kinases (MAPKs) such as the p38 MAPK and extracellular signal-regulated kinase (ERK) 1/2. Moreover, oral administration of the SEWEAF resulted in the attenuation of neointima formation in balloon-injured rat carotid arteries. Additionally, HPLC analysis showed that the major components in the two subfractions of the SEWEAF were five phenolic acids and four flavonols. In the SEWEAF components, for which atherosclerosis-linked responses in VSMCs have not been known, p-coumaric acid, quercetin-3-ß-d-glucoside, and isorhamnetin-3-ß-d-glucoside inhibited both PDGF-BB-induced migration and proliferation and isorhamnetin attenuated only PDGF-BB-stimulated VSMC proliferation. These results suggest that the SEWEAF may suppress PDGF-BB-induced VSMC migration by downregulating the phosphorylation of p38 MAPK and ERK1/2, thus leading to the reduction of neointimal hyperplasia during vascular remodeling. Therefore, the desalted SE extract, SEWEAF may be a potential ingredient for dietary supplements or nutraceuticals to ameliorate and/or prevent vascular remodeling-related disorders.


Assuntos
Movimento Celular/efeitos dos fármacos , Chenopodiaceae/química , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Neointima/enzimologia , Neointima/patologia , Extratos Vegetais/farmacologia , Acetatos/química , Animais , Becaplermina , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Flavonóis/farmacologia , Hidroxibenzoatos/farmacologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis/farmacologia , Ratos Sprague-Dawley , Água
7.
Sci Rep ; 7(1): 3753, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28623250

RESUMO

Several large-scale genome-wide association studies have identified single-nucleotide polymorphisms in the genomic region of A Disintegrin And Metalloproteinase with ThromboSpondin type 1 repeats (ADAMTS)-7 and associations to coronary artery disease. Experimental studies have provided evidence for a functional role of ADAMTS-7 in both injury-induced vascular neointima formation and development of atherosclerotic lesions. However, whether ADAMTS-7 is associated with a specific plaque phenotype in humans has not been investigated. Carotid plaques (n = 206) from patients with and without cerebrovascular symptoms were analyzed for expression of ADAMTS-7 by immunohistochemistry and correlated to components associated with plaque vulnerability. Plaques from symptomatic patients showed increased levels of ADAMTS-7 compared with lesions from asymptomatic patients. High levels of ADAMTS-7 correlated with high levels of CD68-staining and lipid content, but with low smooth muscle cell and collagen content, which together are characteristics of a vulnerable plaque phenotype. ADAMTS-7 levels above median were associated with increased risk for postoperative cardiovascular events. Our data show that ADAMTS-7 is associated with a vulnerable plaque phenotype in human carotid lesions. These data support previous observations of a potential proatherogenic role of ADAMTS-7.


Assuntos
Doenças das Artérias Carótidas , Estudo de Associação Genômica Ampla , Miócitos de Músculo Liso , Neointima , Placa Aterosclerótica , Proteína ADAMTS7/genética , Proteína ADAMTS7/metabolismo , Idoso , Doenças das Artérias Carótidas/enzimologia , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , Neointima/enzimologia , Neointima/genética , Neointima/patologia , Placa Aterosclerótica/enzimologia , Placa Aterosclerótica/genética , Placa Aterosclerótica/patologia , Fatores de Risco
8.
Biochim Biophys Acta Mol Basis Dis ; 1863(9): 2210-2219, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28645655

RESUMO

Given the importance of leukotrienes in vascular inflammation induced by local tissue injury, this study investigated the role for 5-lipoxygenase (5-LO) in monocytes in the development of intimal hyperplasia. As a mechanistic study, the importance of monocyte 5-LO in monocyte-macrophage differentiation with subsequent infiltration in neointima was evaluated. In a mouse model of wire-injured femoral artery, intimal hyperplasia started as early as 2wks after injury, and luminal area and blood flow were reduced due to increased neointima formation. Time-dependent increases in macrophage infiltration were observed in neointima and showed a positive relationship with neointima volume. In 5-LO-deficient (KO) mice or wild-type (WT) mice treated with an inhibitor of 5-LO activating protein (MK886, 1 and 10mg/kg), intimal hyperplasia and macrophage infiltration into neointima were reduced, but monocyte adhesion to injured luminal surface was not inhibited, which suggested 5-LO participates in monocyte-macrophage differentiation. In an in vitro study, monocyte-macrophage differentiation was found to be increased by high mobility group box 1 protein (HMGB1), but this effect was attenuated in cells isolated from 5-LO-KO mice. Furthermore, macrophage infiltration and intimal hyperplasia were more prominent in 5-LO-KO mice transplanted with monocytes from WT mice than in 5-LO-KO mice transplanted with monocytes from 5-LO-KO mice. Taken together, it was suggested that 5-LO in monocytes played a pivotal role in monocyte-macrophage differentiation and subsequent infiltration of macrophage in neointima, leading to vascular remodeling after vascular injury.


Assuntos
Araquidonato 5-Lipoxigenase/metabolismo , Artéria Femoral , Indóis/farmacologia , Inibidores de Lipoxigenase/farmacologia , Macrófagos/enzimologia , Monócitos/enzimologia , Neointima , Animais , Araquidonato 5-Lipoxigenase/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Artéria Femoral/enzimologia , Artéria Femoral/lesões , Artéria Femoral/patologia , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Hiperplasia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Monócitos/patologia , Neointima/tratamento farmacológico , Neointima/enzimologia , Neointima/patologia , Túnica Íntima/enzimologia , Túnica Íntima/patologia , Remodelação Vascular/efeitos dos fármacos , Remodelação Vascular/genética
9.
J Surg Res ; 202(2): 413-21, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27229117

RESUMO

BACKGROUND: Diabetic patients display aggressive restenosis after vascular interventions, likely because of proproliferative influences of hyperglycemia and hyperinsulinemia. We have shown that nitric oxide (NO) inhibits neointimal hyperplasia in type 2, but not in type 1, diabetic rats. Here, we examined proteasome activator 28 (PA28) after arterial injury in different diabetic environments, with or without NO. We hypothesize that NO differentially affects PA28 levels based on metabolic environment. MATERIALS AND METHODS: Vascular smooth muscle cell (VSMC) lysates from male, nondiabetic Lean Zucker (LZ) and Zucker Diabetic Fatty (ZDF) rats were assayed for 26S proteasome activity with or without PA28 and S-nitroso-N-acetylpenicillamine. LZ and ZDF VSMCs were treated with (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate for 24 h. Balloon-injured carotid arteries from LZ, streptozotocin-injected LZ (STZ, type 1), and ZDF (type 2) rats treated with disodium 1-[2-(carboxylato)pyrrolidin-1-iyl]diazen-1-ium-1,2-diolate were harvested at 3 or 14 d. PA28α was assessed by Western blotting and immunofluorescent staining. RESULTS: S-nitroso-N-acetylpenicillamine reversed PA28-stimulated increases in 26S proteasome activity in LZ and ZDF VSMCs. Increased (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate lowered PA28α in LZ VSMCs but increased PA28α in ZDF VSMCs. At 3 d after injury, disodium 1-[2-(carboxylato)pyrrolidin-1-iyl]diazen-1-ium-1,2-diolate potentiated injury-induced PA28α decreases in LZ, STZ, and ZDF rats, suggesting VSMCs, depleted at this early time point, are major sources of PA28α. At 14 d after injury, total PA28α staining returned to baseline. However, although intimal and medial PA28α staining increased in injured STZ rats, adventitial PA28α staining increased in injured ZDF rats. CONCLUSIONS: PA28 dysregulation may explain the differential ability of NO to inhibit neointimal hyperplasia in type 1 versus type 2 diabetes.


Assuntos
Lesões das Artérias Carótidas/tratamento farmacológico , Diabetes Mellitus Tipo 1/enzimologia , Diabetes Mellitus Tipo 2/enzimologia , Neointima/prevenção & controle , Óxido Nítrico/uso terapêutico , Complexo de Endopeptidases do Proteassoma/metabolismo , Substâncias Protetoras/uso terapêutico , Animais , Biomarcadores/metabolismo , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/enzimologia , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/complicações , Lesões das Artérias Carótidas/enzimologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 2/complicações , Hiperplasia/etiologia , Hiperplasia/prevenção & controle , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , Neointima/enzimologia , Neointima/etiologia , Neointima/patologia , Óxido Nítrico/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Ratos , Ratos Zucker , Resultado do Tratamento
10.
J Cardiovasc Pharmacol Ther ; 21(2): 187-200, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26240073

RESUMO

PURPOSE: Gingerol inhibits growth of cancerous cells; however, its role in vascular smooth muscle cell (VSMC) proliferation is not known. The present study investigated the effect of gingerol on VSMC proliferation in cell culture and during neointima formation after balloon injury. METHOD AND RESULTS: Rat VSMCs or carotid arteries were harvested at 15 minutes, 30 minutes, 1, 6, 12, and 24 hours of fetal bovine serum (FBS; 10%) stimulation or balloon injury, respectively. Gingerol prevented FBS (10%)-induced proliferation of VSMCs in a dose-dependent manner (50 µmol/L-400 µmol/L). The FBS-induced proliferating cell nuclear antigen (PCNA) upregulation and p27(Kip1) downregulation were also attenuated in gingerol (200 µmol/L) pretreated cells. Fetal bovine serum-induced p38 mitogen-activated protein kinase (MAPK) activation, PCNA upregulation, and p27(Kip1) downregulation were abrogated in gingerol (200 µmol/L) and p38 MAPK inhibitor (SB203580, 10 µmol/L) pretreated cells. Balloon injury induced time-dependent p38 MAPK activation in the carotid artery. Pretreatment with gingerol (200 µmol/L) significantly attenuated injury-induced p38 MAPK activation, PCNA upregulation, and p27(Kip1) downregulation. After 14 days of balloon injury, intimal thickening, neointimal proliferation, and endothelial dysfunction were significantly prevented in gingerol pretreated arteries. In isolated organ bath studies, gingerol (30 nmol/L-300 µmol/L) inhibited phenylephrine-induced contractions and induced dose-dependent relaxation of rat thoracic aortic rings in a partially endothelium-dependent manner. CONCLUSION: Gingerol prevented FBS-induced VSMC proliferation and balloon injury-induced neointima formation by regulating p38 MAPK. Vasodilator effect of gingerol observed in the thoracic aorta was partially endothelium dependent. Gingerol is thus proposed as an attractive agent for modulating VSMC proliferation, vascular reactivity, and progression of vascular proliferative diseases.


Assuntos
Catecóis/uso terapêutico , Proliferação de Células/fisiologia , Álcoois Graxos/uso terapêutico , Músculo Liso Vascular/enzimologia , Neointima/tratamento farmacológico , Neointima/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Catecóis/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Álcoois Graxos/farmacologia , Hiperplasia/tratamento farmacológico , Hiperplasia/enzimologia , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
11.
J Mol Cell Cardiol ; 89(Pt B): 185-94, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26582463

RESUMO

UNLABELLED: Elevated levels of reactive oxygen species (ROS) in the vascular wall play a key role in the development of neointimal hyperplasia. Nox4-based NADPH oxidase is a major ROS generating enzyme in the vasculature, but its roles in neointimal hyperplasia remain unclear. OBJECTIVE: Our purpose was to investigate the role of smooth muscle cell (SMC) Nox4 in neointimal hyperplasia. APPROACH AND RESULTS: Mice overexpressing a human Nox4 mutant form, carrying a P437H dominant negative mutation (Nox4DN) and driven by SM22α promoter, to achieve specific expression in SMC, were generated in a FVB/N genetic background. After wire injury-induced endothelial denudation, Nox4DN had significantly decreased neointima formation compared with non-transgenic littermate controls (NTg). ROS production, serum-induced proliferation and migration, were significantly decreased in aortic SMCs isolated from Nox4DN compared with NTg. Both mRNA and protein levels of thrombospondin 1 (TSP1) were significantly downregulated in Nox4DN SMCs. Downregulation of TSP1 by siRNA decreased cell proliferation and migration in SMCs. Similar to Nox4DN, downregulation of Nox4 by siRNA significantly decreased TSP1 expression level, cell proliferation and migration in SMCs. CONCLUSIONS: Downregulation of smooth muscle Nox4 inhibits neointimal hyperplasia by suppressing TSP1, which in part can account for inhibition of SMC proliferation and migration.


Assuntos
Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , NADPH Oxidases/metabolismo , Neointima/enzimologia , Neointima/patologia , Animais , Aorta/metabolismo , Aorta/patologia , Artérias Carótidas/patologia , Movimento Celular , Proliferação de Células , Regulação para Baixo , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Técnicas de Silenciamento de Genes , Humanos , Peróxido de Hidrogênio/metabolismo , Hiperplasia , Masculino , Camundongos Transgênicos , NADPH Oxidase 4 , RNA Interferente Pequeno/metabolismo , Trombospondina 1/metabolismo
12.
Vascul Pharmacol ; 71: 108-15, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25866325

RESUMO

Creation of an autologous arteriovenous fistula (AVF) for vascular access in haemodialysis is the modality of choice. However neointimal hyperplasia and loss of the luminal compartment result in AVF patency rates of ~60% at 12months. The exact cause of neointimal hyperplasia in the AVF is poorly understood. Vascular trauma has long been associated with hyperplasia. With this in mind in our rabbit model of AVF we simulated cannulation autologous to that undertaken in vascular access procedures and observed significant neointimal hyperplasia as a direct consequence of cannulation. The neointimal hyperplasia was completely inhibited by topical transdermal delivery of the non-steroidal anti-inflammatory (NSAID) diclofenac. In addition to the well documented anti-inflammatory properties we have identified novel anti-proliferative mechanisms demonstrating diclofenac increases AMPK-dependent signalling and reduced expression of the cell cycle protein cyclin D1. In summary prophylactic transdermal delivery of diclofenac to the sight of AVF cannulation prevents adverse neointimal hyperplasic remodelling and potentially offers a novel treatment option that may help prolong AVF patency and flow rates.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Fístula Arteriovenosa/prevenção & controle , Cateterismo/efeitos adversos , Diclofenaco/administração & dosagem , Neointima/tratamento farmacológico , Grau de Desobstrução Vascular/efeitos dos fármacos , Administração Cutânea , Animais , Fístula Arteriovenosa/enzimologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Sistemas de Liberação de Medicamentos/métodos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Hiperplasia/enzimologia , Hiperplasia/prevenção & controle , Neointima/enzimologia , Coelhos , Grau de Desobstrução Vascular/fisiologia
13.
Arterioscler Thromb Vasc Biol ; 35(6): 1445-55, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25908764

RESUMO

OBJECTIVE: Reduced frequency of atherosclerotic plaques is observed in interleukin-1 receptor-associated kinase-1 (IRAK1)-deficient mice; however, the underlying mechanism is not clear. Therefore, this study investigate the role of IRAK1 in vascular smooth muscle cell proliferation and neointimal hyperplasia. APPROACH AND RESULTS: Stimulation of rat primary vascular smooth muscle cells with fetal bovine serum (10%) or platelet-derived growth factor-BB (20 ng/mL) for 15 minutes to 24 hours induced a time-dependent increase in IRAK1 and extracellular signal-regulated kinase (ERK) activation, proliferating cell nuclear antigen upregulation and p27Kip1 downregulation as assessed by Western blotting. Inhibitors of ERK pathway (U0126, 10 µmol/L), IRAK (IRAK1/4, 3 µmol/L), protein kinase C (PKC; Ro-31-8220, 1 µmol/L), siRNA of toll-like receptor-4 (200 nmol/L), and PKC-ε (200 nmol/L) significantly attenuated these changes. Platelet-derived growth factor induced endogenous IRAK-ERK-PKC-ε association in a toll-like receptor-4 and PKC-ε-dependent manner. A time-dependent increase in IRAK1 and ERK activation was observed after 15 minutes, 30 minutes, 1 hour, 6 hours, 12 hours, and 24 hours of carotid balloon injury in rats. Balloon injury induced endogenous IRAK-ERK-PKC-ε interaction. Perivascular application of IRAK1/4 inhibitor (100 µmol/L), U0126 (100 µmol/L), and IRAK1 siRNA (220 and 360 nmol/L) in pluronic gel abrogated balloon injury-induced ERK phosphorylation, activation, and p27Kip1 downregulation. Hematoxylin and eosin staining and immunohistochemistry of proliferating cell nuclear antigen and smooth muscle actin demonstrated that balloon injury-induced intimal thickening and neointimal vascular smooth muscle cell proliferation were significantly abrogated in the presence of IRAK1/4 inhibitor, IRAK1 siRNA, and U0126. CONCLUSIONS: IRAK1 mediates vascular smooth muscle cell proliferation and neointimal hyperplasia by regulating PKC-ε-IRAK1-ERK axis.


Assuntos
Artérias Carótidas/enzimologia , Lesões das Artérias Carótidas/enzimologia , Proliferação de Células , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Neointima/enzimologia , Animais , Artérias Carótidas/citologia , Lesões das Artérias Carótidas/patologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Miócitos de Músculo Liso/citologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Ratos , Ratos Sprague-Dawley , Soroalbumina Bovina/farmacologia
16.
Circulation ; 131(13): 1202-1213, 2015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25712206

RESUMO

BACKGROUND: Genome-wide association studies have established ADAMTS7 as a locus for coronary artery disease in humans. However, these studies fail to provide directionality for the association between ADAMTS7 and coronary artery disease. Previous reports have implicated ADAMTS7 in the regulation of vascular smooth muscle cell migration, but a role for and the direction of impact of this gene in atherogenesis have not been shown in relevant model systems. METHODS AND RESULTS: We bred an Adamts7 whole-body knockout mouse onto both the Ldlr and Apoe knockout hyperlipidemic mouse models. Adamts7(-/-)/Ldlr(-/-) and Adamts7(-/-)/Apoe(-/-) mice displayed significant reductions in lesion formation in aortas and aortic roots compared with controls. Adamts7 knockout mice also showed reduced neointimal formation after femoral wire injury. Adamts7 expression was induced in response to injury and hyperlipidemia but was absent at later time points, and primary Adamts7 knockout vascular smooth muscle cells showed reduced migration in the setting of tumor necrosis factor-α stimulation. ADAMTS7 localized to cells positive for smooth muscle cell markers in human coronary artery disease lesions, and subcellular localization studies in cultured vascular smooth muscle cells placed ADAMTS7 at the cytoplasm and cell membrane, where it colocalized with markers of podosomes. CONCLUSIONS: These data represent the first in vivo experimental validation of the association of Adamts7 with atherogenesis, likely through modulation of vascular cell migration and matrix in atherosclerotic lesions. These results demonstrate that Adamts7 is proatherogenic, lending directionality to the original genetic association and supporting the concept that pharmacological inhibition of ADAMTS7 should be atheroprotective in humans, making it an attractive target for novel therapeutic interventions.


Assuntos
Proteínas ADAM/análise , Proteínas ADAM/fisiologia , Aterosclerose/prevenção & controle , Doença das Coronárias/enzimologia , Neointima/enzimologia , Remodelação Vascular/fisiologia , Proteínas ADAM/deficiência , Proteínas ADAM/genética , Proteína ADAMTS7 , Sequência de Aminoácidos , Animais , Aorta/enzimologia , Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/etiologia , Aterosclerose/genética , Aterosclerose/patologia , Divisão Celular , Movimento Celular , Células Cultivadas , Doença das Coronárias/patologia , Dieta Ocidental/efeitos adversos , Células Endoteliais/metabolismo , Feminino , Artéria Femoral/lesões , Artéria Femoral/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Hiperlipidemias/complicações , Hiperlipidemias/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/ultraestrutura , Neointima/patologia , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/farmacologia , Receptores de LDL/deficiência , Receptores de LDL/genética , Fator de Necrose Tumoral alfa/farmacologia
17.
Circulation ; 131(13): 1191-201, 2015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25712208

RESUMO

BACKGROUND: ADAMTS-7, a member of the disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) family, was recently identified to be significantly associated genomewide with coronary artery disease. However, the mechanisms that link ADAMTS-7 and coronary artery disease risk remain elusive. We have previously demonstrated that ADAMTS-7 promotes vascular smooth muscle cell migration and postinjury neointima formation via degradation of a matrix protein cartilage oligomeric matrix protein. Because delayed endothelium repair renders neointima and atherosclerosis plaque formation after vessel injury, we examined whether ADAMTS-7 also inhibits re-endothelialization. METHODS AND RESULTS: Wire injury of the carotid artery and Evans blue staining were performed in Adamts7(-/-) and wild-type mice. Adamts-7 deficiency greatly promoted re-endothelialization at 3, 5, and 7 days after injury. Consequently, Adamts-7 deficiency substantially ameliorated neointima formation in mice at days 14 and 28 after injury in comparison with the wild type. In vitro studies further indicated that ADAMTS-7 inhibited both endothelial cell proliferation and migration. Surprisingly, cartilage oligomeric matrix protein deficiency did not affect endothelial cell proliferation/migration and re-endothelialization in mice. In a further examination of other potential vascular substrates of ADAMTS-7, a label-free liquid chromatography-tandem mass spectrometry secretome analysis revealed thrombospondin-1 as a potential ADAMTS-7 target. The subsequent studies showed that ADAMTS-7 was directly associated with thrombospondin-1 by its C terminus and degraded thrombospondin-1 in vivo and in vitro. The inhibitory effect of ADAMTS-7 on postinjury endothelium recovery was circumvented in Tsp1(-/-) mice. CONCLUSIONS: Our study revealed a novel mechanism by which ADAMTS-7 affects neointima formation. Thus, ADAMTS-7 is a promising treatment target for postinjury vascular intima hyperplasia.


Assuntos
Proteínas ADAM/fisiologia , Lesões das Artérias Carótidas/enzimologia , Artéria Femoral/lesões , Neointima/enzimologia , Trombospondina 1/fisiologia , Remodelação Vascular/fisiologia , Proteínas ADAM/deficiência , Proteínas ADAM/genética , Proteína ADAMTS7 , Sequência de Aminoácidos , Animais , Lesões das Artérias Carótidas/patologia , Artéria Carótida Primitiva/enzimologia , Divisão Celular , Células Cultivadas , Células Endoteliais/metabolismo , Artéria Femoral/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Miócitos de Músculo Liso/metabolismo , Neointima/patologia , Mapeamento de Interação de Proteínas , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Ratos , Trombospondina 1/deficiência , Trombospondina 1/genética
18.
Circ Res ; 116(7): 1120-32, 2015 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-25608528

RESUMO

RATIONALE: Neointimal hyperplasia characterized by abnormal accumulation of vascular smooth muscle cells (SMCs) is a hallmark of occlusive disorders such as atherosclerosis, postangioplasty restenosis, vein graft stenosis, and allograft vasculopathy. Cyclic nucleotides are vital in SMC proliferation and migration, which are regulated by cyclic nucleotide phosphodiesterases (PDEs). OBJECTIVE: Our goal is to understand the regulation and function of PDEs in SMC pathogenesis of vascular diseases. METHODS AND RESULTS: We performed screening for genes differentially expressed in normal contractile versus proliferating synthetic SMCs. We observed that PDE1C expression was low in contractile SMCs but drastically elevated in synthetic SMCs in vitro and in various mouse vascular injury models in vivo. In addition, PDE1C was highly induced in neointimal SMCs of human coronary arteries. More importantly, injury-induced neointimal formation was significantly attenuated by PDE1C deficiency or PDE1 inhibition in vivo. PDE1 inhibition suppressed vascular remodeling of human saphenous vein explants ex vivo. In cultured SMCs, PDE1C deficiency or PDE1 inhibition attenuated SMC proliferation and migration. Mechanistic studies revealed that PDE1C plays a critical role in regulating the stability of growth factor receptors, such as PDGF receptor ß (PDGFRß) known to be important in pathological vascular remodeling. PDE1C interacts with low-density lipoprotein receptor-related protein-1 and PDGFRß, thus regulating PDGFRß endocytosis and lysosome-dependent degradation in an low-density lipoprotein receptor-related protein-1-dependent manner. A transmembrane adenylyl cyclase cAMP-dependent protein kinase cascade modulated by PDE1C is critical in regulating PDGFRß degradation. CONCLUSIONS: These findings demonstrated that PDE1C is an important regulator of SMC proliferation, migration, and neointimal hyperplasia, in part through modulating endosome/lysosome-dependent PDGFRß protein degradation via low-density lipoprotein receptor-related protein-1.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 1/fisiologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/enzimologia , Neointima/enzimologia , Animais , Lesões das Artérias Carótidas/enzimologia , Lesões das Artérias Carótidas/patologia , Divisão Celular , Movimento Celular , Células Cultivadas , AMP Cíclico/fisiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1/antagonistas & inibidores , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1/deficiência , Endocitose/fisiologia , Indução Enzimática , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Lisossomos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Miócitos de Músculo Liso/citologia , Neointima/fisiopatologia , Mapeamento de Interação de Proteínas , Estabilidade Proteica , Proteólise , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/fisiologia
19.
Cardiovasc Res ; 105(1): 75-85, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25388664

RESUMO

AIMS: Phosphatidylinositol 3 kinases (PI3Ks) play a pivotal role in vascular physiology and pathophysiology. We aimed to investigate the role of p55γ, a regulatory subunit of PI3Ks, in vascular smooth muscle cell (VSMC) proliferation and neointimal formation. METHODS AND RESULTS: We identified p55γ as an important factor that suppresses VSMC proliferation and injury-evoked neointimal formation. Western blot and mRNA analyses showed that p55γ expression declined in balloon-injured rat carotid arteries and in response to PDGF-BB and serum treatment in cultured VSMCs. Overexpression of p55γ inhibited, whereas short hairpin RNA knockdown of p55γ promoted PDGF-BB- and serum-induced VSMC proliferation. Importantly, in vivo adenoviral gene transfer of p55γ into carotid arteries attenuated, while knockdown of p55γ enhanced balloon injury-induced neointimal formation. Furthermore, p55γ sequentially up-regulated p53 and p21, resulting in cell-cycle arrest in S phase; small-interfering RNA knockdown of either p53 or p21 blocked p55γ-induced VSMC growth arrest. Mechanistically, p55γ interacted with and stabilized p53 protein by blocking mouse double minute 2 homologue-mediated p53 ubiquitination and degradation, subsequently activating its target gene p21. Concurrently, p55γ up-regulated Bcl-xl expression, resulting in non-apoptotic growth arrest effect. CONCLUSION: These findings mark p55γ as a novel upstream regulator of the p53-p21 signalling pathway that negatively regulates VSMC proliferation, suggesting that malfunction of p55γ may trigger vascular proliferative disorders.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/patologia , Neointima/enzimologia , Neointima/prevenção & controle , Animais , Lesões das Artérias Carótidas/enzimologia , Lesões das Artérias Carótidas/patologia , Pontos de Checagem do Ciclo Celular , Proliferação de Células , Células Cultivadas , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Masculino , Neointima/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima , Proteína bcl-X/antagonistas & inibidores , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
20.
J Exp Med ; 211(9): 1779-92, 2014 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-25073791

RESUMO

Interventional strategies to treat atherosclerosis, such as transluminal angioplasty and stent implantation, often cause vascular injury. This leads to intimal hyperplasia (IH) formation that induces inflammatory and fibroproliferative processes and ultimately restenosis. We show that phosphoinositide 3-kinase γ (PI3Kγ) is a key player in IH formation and is a valid therapeutic target in its prevention/treatment. PI3Kγ-deficient mice and mice expressing catalytically inactive PI3Kγ (PI3Kγ KD) showed reduced arterial occlusion and accumulation of monocytes and T cells around sites of vascular lesion. The transfer of PI3Kγ KD CD4(+) T cells into Rag2-deficient mice greatly reduced vascular occlusion compared with WT cells, clearly demonstrating the involvement of PI3Kγ in CD4(+) T cells during IH formation. In addition we found that IH is associated with increased levels of Th1 and Th17 cytokines. A specific decrease in the Th1 response was observed in the absence of PI3Kγ activity, leading to decreased CXCL10 and RANTES production by smooth muscle cells. Finally, we show that treatment with the PI3Kγ inhibitor AS-605240 is sufficient to decrease IH in both mouse and rat models, reinforcing the therapeutic potential of PI3Kγ inhibition. Altogether, these findings demonstrate a new role for PI3Kγ activity in Th1-controlled IH development.


Assuntos
Neointima/enzimologia , Neointima/imunologia , Inibidores de Fosfoinositídeo-3 Quinase , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Animais , Lesões das Artérias Carótidas/tratamento farmacológico , Lesões das Artérias Carótidas/enzimologia , Lesões das Artérias Carótidas/imunologia , Classe Ib de Fosfatidilinositol 3-Quinase/deficiência , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Inibidores Enzimáticos/farmacologia , Artéria Femoral/enzimologia , Artéria Femoral/imunologia , Artéria Femoral/lesões , Marcação de Genes , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neointima/tratamento farmacológico , Quinoxalinas/farmacologia , Ratos , Ratos Sprague-Dawley , Tiazolidinedionas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA