Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cardiovasc Res ; 119(12): 2244-2255, 2023 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-37486354

RESUMO

AIMS: Phenotypic transition of vascular smooth muscle cells (VSMCs) from a contractile to a synthetic state is involved in the development of cardiovascular diseases, including atherosclerosis, hypertension, and post-angioplasty restenosis. Arginine methylation catalyzed by protein arginine methyltransferases (PRMTs) has been implicated in multiple cellular processes, however, its role in VSMC biology remains undetermined. The objective of this study was to determine the role of PRMTs in VSMC phenotypic switch and vascular remodelling after injury. METHODS AND RESULTS: Our results show that PRMT5 is the most abundantly expressed PRMT in human aortic SMCs, and its expression is up-regulated in platelet-derived growth factor (PDGF)-stimulated VSMCs, human atherosclerotic lesions, and rat carotid arteries after injury, as determined by western blot and immunohistochemical staining. PRMT5 overexpression inhibits the expression of SMC marker genes and promotes VSMC proliferation and migration, while silencing PRMT5 exerts the opposite effects. Mechanistically, we found that PRMT5 overexpression led to histone di-methylation of H3R8 and H4R3, which in turn attenuates acetylation of H3K9 and H4, thus limiting recruitment of the SRF/myocardin complexes to the CArG boxes of SMC marker genes. Furthermore, both SMC-specific deletion of PRMT5 in mice and local delivery of lentivirus expressing shPRMT5 to rat carotid arteries significantly attenuated neointimal formation after injury. Likewise, pharmacological inhibition of PRMT5 by EPZ015666 markedly inhibited carotid artery ligation-induced neointimal formation in mice. CONCLUSIONS: Our results identify PRMT5 as a novel regulator in VSMC phenotypic switch and suggest that inhibition of PRMT5 may represent an effective therapeutic strategy for proliferative vascular diseases.


Assuntos
Aterosclerose , Músculo Liso Vascular , Proteína-Arginina N-Metiltransferases , Animais , Humanos , Camundongos , Ratos , Arginina , Aterosclerose/patologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Epigênese Genética , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Neointima , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo
2.
Elife ; 122023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-37070640

RESUMO

Proinflammatory agonists provoke the expression of cell surface adhesion molecules on endothelium in order to facilitate leukocyte infiltration into tissues. Rigorous control over this process is important to prevent unwanted inflammation and organ damage. Protein L-isoaspartyl O-methyltransferase (PIMT) converts isoaspartyl residues to conventional methylated forms in cells undergoing stress-induced protein damage. The purpose of this study was to determine the role of PIMT in vascular homeostasis. PIMT is abundantly expressed in mouse lung endothelium and PIMT deficiency in mice exacerbated pulmonary inflammation and vascular leakage to LPS(lipopolysaccharide). Furthermore, we found that PIMT inhibited LPS-induced toll-like receptor signaling through its interaction with TNF receptor-associated factor 6 (TRAF6) and its ability to methylate asparagine residues in the coiled-coil domain. This interaction was found to inhibit TRAF6 oligomerization and autoubiquitination, which prevented NF-κB transactivation and subsequent expression of endothelial adhesion molecules. Separately, PIMT also suppressed ICAM-1 expression by inhibiting its N-glycosylation, causing effects on protein stability that ultimately translated into reduced EC(endothelial cell)-leukocyte interactions. Our study has identified PIMT as a novel and potent suppressor of endothelial activation. Taken together, these findings suggest that therapeutic targeting of PIMT may be effective in limiting organ injury in inflammatory vascular diseases.


Assuntos
Lipopolissacarídeos , Proteína D-Aspartato-L-Isoaspartato Metiltransferase , Fator 6 Associado a Receptor de TNF , Animais , Camundongos , Células Endoteliais/metabolismo , Endotélio/metabolismo , Lipopolissacarídeos/metabolismo , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo , Proteína D-Aspartato-L-Isoaspartato Metiltransferase/genética , Proteína D-Aspartato-L-Isoaspartato Metiltransferase/metabolismo
3.
Circ Res ; 132(3): 306-319, 2023 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-36562299

RESUMO

BACKGROUND: NDRG-1 (N-myc downstream-regulated gene 1) is a member of NDRG family that plays essential roles in cell differentiation, proliferation, and stress responses. Although the expression of NDRG1 is regulated by fluid shear stress, its roles in vascular biology remain poorly understood. The purpose of the study is to determine the functional significance of NDRG1 in vascular inflammation and remodeling. METHODS AND RESULTS: By using quantitative polymerase chain reaction, western blot, and immunohistochemistry, we demonstrate that the expression of NDRG1 is markedly increased in cytokine-stimulated endothelial cells and in human and mouse atherosclerotic lesions. To determine the role of NDRG1 in endothelial activation, we performed loss-of-function studies using NDRG1 short hairpin RNA. Our results demonstrate that NDRG1 knockdown by lentivirus bearing NDRG1 short hairpin RNA substantially attenuates both IL-1ß (interleukin-1ß) and TNF-α (tumor necrosis factor-α)-induced expression of cytokines/chemokines and adhesion molecules. Intriguingly, inhibition of NDRG1 also significantly attenuates the expression of procoagulant molecules, such as PAI-1 (plasminogen activator inhibitor type 1) and TF (tissue factor), and increases the expression of TM (thrombomodulin) and t-PA (tissue-type plasminogen activator), thus exerting potent antithrombotic effects in endothelial cells. Mechanistically, we showed that NDRG1 interacts with orphan Nur77 (nuclear receptor) and functionally inhibits the transcriptional activity of Nur77 and NF-κB (nuclear factor Kappa B) in endothelial cells. Moreover, in NDRG1 knockdown cells, both cytokine-induced mitogen-activated protein kinase activation, c-Jun phosphorylation, and AP-1 (activator protein 1) transcriptional activity are substantially inhibited. Neointima and atherosclerosis formation induced by carotid artery ligation and arterial thrombosis were markedly attenuated in endothelial cell-specific NDRG1 knockout mice compared with their wild-type littermates. CONCLUSIONS: Our results for the first time identify NDRG1 as a critical mediator implicated in regulating endothelial inflammation, thrombotic responses, and vascular remodeling, and suggest that inhibition of NDRG1 may represent a novel therapeutic strategy for inflammatory vascular diseases, such as atherothrombosis and restenosis.


Assuntos
Células Endoteliais , Trombose , Humanos , Animais , Camundongos , Células Endoteliais/metabolismo , Remodelação Vascular , NF-kappa B/metabolismo , Citocinas/metabolismo , Inflamação/genética , Inflamação/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Trombose/metabolismo , RNA Interferente Pequeno/metabolismo
4.
JCI Insight ; 7(9)2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35531958

RESUMO

Arginine methylation mediated by protein arginine methyltransferases (PRMTs) has been shown to be an important posttranslational mechanism involved in various biological processes. Herein, we sought to investigate whether PRMT5, a major type II enzyme, is involved in pathological angiogenesis and, if so, to elucidate the molecular mechanism involved. Our results show that PRMT5 expression is significantly upregulated in ischemic tissues and hypoxic endothelial cells (ECs). Endothelial-specific Prmt5-KO mice were generated to define the role of PRMT5 in hindlimb ischemia-induced angiogenesis. We found that these mice exhibited impaired recovery of blood perfusion and motor function of the lower limbs, an impairment that was accompanied by decreased vascular density and increased necrosis as compared with their WT littermates. Furthermore, both pharmacological and genetic inhibition of PRMT5 significantly attenuated EC proliferation, migration, tube formation, and aortic ring sprouting. Mechanistically, we showed that inhibition of PRMT5 markedly attenuated hypoxia-induced factor 1-α (HIF-1α) protein stability and vascular endothelial growth factor-induced (VEGF-induced) signaling pathways in ECs. Our results provide compelling evidence demonstrating a crucial role of PRMT5 in hypoxia-induced angiogenesis and suggest that inhibition of PRMT5 may provide novel therapeutic strategies for the treatment of abnormal angiogenesis-related diseases, such as cancer and diabetic retinopathy.


Assuntos
Células Endoteliais , Neovascularização Fisiológica , Proteína-Arginina N-Metiltransferases , Animais , Arginina/metabolismo , Células Endoteliais/metabolismo , Hipóxia/metabolismo , Isquemia/genética , Isquemia/metabolismo , Camundongos , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica/genética , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Am J Respir Cell Mol Biol ; 65(3): 288-299, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33971110

RESUMO

Inflammasomes are intracellular multiprotein complexes that help trigger and maintain the inflammatory response as part of the innate immune system. Recently, it has been increasingly recognized that aberrant inflammasome activation is critically involved in endothelial dysfunction in a variety of human diseases, such as atherosclerosis, acute lung injury (ALI), and type 2 diabetes. The molecular mechanisms underlying endothelial inflammasome activation, however, have not been completely elucidated. In the present study, we identified orphan nuclear receptor Nur77 as a novel regulator in controlling inflammasome activation in vascular endothelial cells (ECs). We demonstrated that LPS-induced inflammasome activation was significantly inhibited by ectopic overexpression of Nur77, predominantly through transcriptional suppression of caspase-1 expression in vascular ECs. Consistent with this observation, we found that LPS-induced inflammasome activation was significantly augmented in lung ECs isolated from Nur77-knockout mice. Mechanistically, we showed that Nur77-induced inhibition of caspase-1 expression was due to an inhibition of IRF1 (IFN regulatory factor 1) expression and its subsequent binding to the caspase-1 promoter. Importantly, in a mouse model of LPS-induced ALI, Nur77 knockout led to a marked activation of caspase-1 in the lung, increased alveolar and circulating IL-1ß levels, and exacerbated ALI, all of which were substantially inhibited by administration of caspase-1 inhibitor. Together, our results support the presence of an important role for Nur77 in controlling inflammasome activation in vascular ECs and suggest that Nur77 could be a novel therapeutic target for the treatment of human diseases associated with aberrant inflammasome activation, such as ALI and atherosclerosis.


Assuntos
Caspase 1/biossíntese , Regulação Enzimológica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Inflamassomos/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Caspase 1/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Inflamassomos/genética , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Camundongos , Camundongos Knockout , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética
6.
J Cell Physiol ; 234(1): 369-381, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-30076719

RESUMO

Abnormal airway smooth muscle cell (ASMC) proliferation and migration contribute significantly to increased ASM mass associated with asthma. MicroRNA (miR)-638 is a primate-specific miRNA that plays important roles in development, DNA damage repair, hematopoiesis, and tumorigenesis. Although it is highly expressed in ASMCs, its function in ASM remodeling remains unknown. In the current study, we found that in response to various mitogenic stimuli, including platelet-derived growth factor-two B chains (PDGF-BB), transforming growth factor ß1, and fetal bovine serum, the expression of miR-638, as determined by quantitative real-time polymerase chain reaction (qRT-PCR), was significantly downregulated in the proliferative human ASMCs. Both gain- and loss-of-function studies were performed to study the role of miR-638 in ASMC proliferation and migration. We found that adenovirus-mediated miR-638 overexpression markedly inhibits ASMC proliferation and migration, while ablation of miR-638 by anti-miR-638 markedly increases cell proliferation and migration, as determined by WST-8 proliferation and scratch wound assays. Dual-luciferase reporter assay, qRT-PCR, and immunoblot analysis were used to investigate the effects of miR-638 on the expression of the downstream target genes in ASMCs. Our results demonstrated that miR-638 overexpression significantly reduced the expression of downstream target cyclin D1 and NOR1, both of which have been shown to be essential for cell proliferation and migration. Together, our study provides the first in vitro evidence highlighting the antiproliferative and antimigratory roles of miR-638 in human ASMC remodeling and suggests that targeted overexpression of miR-638 in ASMCs may provide a novel therapeutic strategy for preventing ASM hyperplasia associated with asthma.


Assuntos
Remodelação das Vias Aéreas/genética , Asma/genética , Ciclina D1/genética , Proteínas de Membrana Transportadoras/genética , MicroRNAs/genética , Asma/patologia , Becaplermina/genética , Movimento Celular/genética , Proliferação de Células/genética , Regulação da Expressão Gênica/genética , Humanos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia
7.
Am J Physiol Heart Circ Physiol ; 309(5): H1017-28, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26254337

RESUMO

Blood chemerin concentration shows positive correlation not only with body mass index and serum triglyceride level but also with systolic blood pressure. While it seems likely that chemerin influences vascular smooth muscle cell (SMC) proliferation and migration, which are crucial to the development of hypertension, this remains to be clarified. In the present study, we investigated whether chemerin controls SMC proliferation and migration in vitro and also affects blood pressure in vivo. In vitro, chemerin significantly stimulated rat mesenteric arterial SMC proliferation and migration, as determined by a cell counting assay and Boyden chamber assay, respectively. The migratory effect of chemerin was confirmed in human aortic SMCs. Chemerin significantly increased ROS production in SMCs and phosphorylation of Akt (Ser(473)) and ERK, as measured by fluorescent staining and Western blot analysis, respectively. Various inhibitors (ROS inhibitor: N-acetyl-l-cysteine, phosphatidylinositol 3-kinase inhibitor: LY-294002, MAPKK inhibitor: PD-98059, NADPH oxidase inhibitor: gp91 ds-tat, and xanthine oxidase inhibitor: allopurinol) as well as chemokine-like receptor 1 small interfering RNA significantly inhibited chemerin-induced SMC proliferation and migration. Furthermore, chemerin-neutralizing antibody prevented carotid neointimal hyperplasia in the mouse ligation model. In vivo, chronic chemerin treatment (6 µg/kg, 6 wk) increased systolic blood pressure as well as phosphorylation of Akt and ERK in the mouse isolated aorta. In summary, we, for the first time, demonstrate that chemerin/chemokine-like receptor 1 stimulates SMC proliferation and migration via ROS-dependent phosphorylation of Akt/ERK, which may lead to vascular structural remodeling and an increase in systolic blood pressure.


Assuntos
Pressão Sanguínea , Movimento Celular , Proliferação de Células , Quimiocinas/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Animais , Artérias/efeitos dos fármacos , Artérias/metabolismo , Artérias/fisiopatologia , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
9.
Am J Physiol Heart Circ Physiol ; 308(10): H1298-305, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25770246

RESUMO

Pulmonary arterial (PA) hypertension (PAH) is a progressive and lethal disease that is caused by increased vascular contractile reactivity and structural remodeling. These changes contribute to increasing pulmonary peripheral vascular resistance, finally leading to right heart failure and death. Eukaryotic elongation factor 2 kinase (eEF2K) is a Ca(2+)/calmodulin-dependent protein kinase. We previously revealed that eEF2K protein increases in the mesenteric artery from spontaneously hypertensive rats and partly mediates the development of hypertension via a promotion of ROS-dependent vascular inflammatory responses and proliferation and migration of vascular smooth muscle cells. However, a role of eEF2K in the pathogenesis of PAH is unknown. In the present study, we tested the hypothesis that eEF2K may be involved in the pathogenesis of PAH. PAH was induced by a single intraperitoneal injection of monocrotaline (MCT; 60 mg/kg) to rats. A specific eEF2K inhibitor, A-484954 (2.5 mg·kg(-1)·day(-1)), was intraperitoneally injected for 14 days. Long-term A-484954 treatment inhibited MCT-induced increased PA pressure. It was revealed that A-484954 inhibited MCT-induced PA hypertrophy and fibrosis but not impairment of endothelium-dependent and -independent relaxation. Furthermore, A-484954 inhibited MCT-induced NADPH oxidase-1 expression and ROS generation as well as matrix metalloproteinase-2 activation. In conclusion, the present results suggest that eEF2K at least partly mediates MCT-induced PAH via stimulation of vascular structural remodeling perhaps through NADPH oxidase-1/ROS/matrix metalloproteinase-2 pathway.


Assuntos
Quinase do Fator 2 de Elongação/metabolismo , Hipertensão Pulmonar/metabolismo , Piridinas/farmacologia , Pirimidinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Remodelação Vascular , Animais , Quinase do Fator 2 de Elongação/antagonistas & inibidores , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/patologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Monocrotalina/toxicidade , NADH NADPH Oxirredutases/genética , NADH NADPH Oxirredutases/metabolismo , NADPH Oxidase 1 , Piridinas/uso terapêutico , Pirimidinas/uso terapêutico , Ratos , Ratos Wistar
10.
Biochem Biophys Res Commun ; 457(4): 602-7, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25600813

RESUMO

Omentin is a relatively novel adipocyte-derived cytokine mainly expressed in visceral adipose tissues. Blood omentin level decreases in the patients with obesity, hypertension, type 2 diabetes and atherosclerosis. We have previously demonstrated that omentin inhibits key pathological processes for hypertension development, including vascular inflammatory responses, contractile reactivity and structural remodeling. In addition, there are several reports demonstrating that omentin prevents cardiac hypertrophy and myocardial ischemic injury. Doxorubicin (DOX) is an effective anti-cancer drug with cardiotoxic side effect. Here we tested the hypothesis that omentin may prevent DOX-induced cardiac cytotoxicity. H9c2 rat cardiomyoblasts were treated with DOX in the absence or presence of omentin. Omentin (300 ng/ml, 3 h pretreatment) significantly inhibited DOX (1 µM, 18 h)-induced decreases in living cell number as determined by a colorimetric cell counting assay. Omentin (300 ng/ml, 3 h) significantly inhibited DOX (1 µM, 12 h)-induced cleaved caspase-3 expression as determined by Western blotting. Omentin (300 ng/ml, 3 h) significantly inhibited DOX (1 µM, 6 h)-induced mitochondrial reactive oxygen species (ROS) production as determined by a MitoSOX Red fluorescent staining. In addition, a mitochondrial respiratory chain complex I inhibitor, rotenone (0.5 µM, 3 h pretreatment), significantly inhibited DOX (1 µM, 6-18 h)-induced decreases of living cell number, cleaved caspase-3 expression and mitochondrial ROS production. In summary, we for the first time demonstrate that omentin prevents DOX-induced H9c2 cells apoptosis through the inhibition of mitochondrial ROS production. These results indicate omentin as an attractive pharmaco-therapeautic target against DOX-induced cardiac side effect.


Assuntos
Adipocinas/farmacologia , Antibióticos Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Cardiotônicos/farmacologia , Doxorrubicina/toxicidade , Mioblastos Cardíacos/efeitos dos fármacos , Animais , Linhagem Celular , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/metabolismo , Mioblastos Cardíacos/patologia , Ratos , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/farmacologia
11.
Biochem Biophys Res Commun ; 452(1): 142-6, 2014 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-25152392

RESUMO

Omentin is a novel adipocytokine mainly expressed in visceral rather than subcutaneous adipose tissue. Several epidemiological studies demonstrated the negative relationship between blood omentin level and occurrence of obesity, type 2 diabetes and hypertension. Increases of inflammatory responses, contractile reactivity and structural remodeling of vascular wall contribute to hypertension development. Our in vitro studies previously demonstrated that omentin inhibited those hypertension-related pathological processes. In addition, our in vivo study demonstrated that intravenously injected omentin acutely inhibited agonists-induced increases of blood pressure in rats. However, the chronic effects of omentin on hypertension development are not determined. In the present study, we tested the hypothesis that chronic omentin treatment may inhibit pulmonary arterial (PA) hypertension (PAH). PAH was induced by a single intraperitoneal injection of monocrotaline (MCT: 60 mg/kg) to rats. Omentin (18 µg/kg/day) was intraperitoneally treated for 14 days. Chronic omentin treatment inhibited MCT-induced increases in PA pressure. Omentin inhibited MCT-induced right ventricular hypertrophy as well as increase of lung to body weight ratio. Histologically, omentin inhibited MCT-induced PA hyperplasia. Further, omentin inhibited the impairment of both endothelium-dependent and -independent relaxations mediated by acetylcholine and sodium nitroprusside, respectively. In conclusion, we for the first time demonstrate that chronic omentin treatment inhibits MCT-induced PAH in rats via inhibiting vascular structural remodeling and abnormal contractile reactivity.


Assuntos
Citocinas/fisiologia , Hipertensão Pulmonar/prevenção & controle , Lectinas/fisiologia , Monocrotalina/antagonistas & inibidores , Animais , Hipertensão Pulmonar/induzido quimicamente , Masculino , Monocrotalina/efeitos adversos , Ratos , Ratos Wistar
12.
Clin Sci (Lond) ; 127(8): 539-48, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24814693

RESUMO

Death-associated protein kinase 3 (DAPK3) also known as zipper-interacting kinase is a serine/threonine kinase that mainly regulates cell death and smooth muscle contraction. We have previously found that protein expression of DAPK3 increases in the mesenteric artery from spontaneously hypertensive rats (SHRs) and that DAPK3 mediates the development of hypertension in SHRs partly through promoting reactive oxygen species-dependent vascular inflammation. However, it remains to be clarified how DAPK3 controls smooth muscle cell (SMC) proliferation and migration, which are also important processes for hypertension development. We, therefore, sought to investigate whether DAPK3 affects SMC proliferation and migration. siRNA against DAPK3 significantly inhibited platelet-derived growth factor (PDGF)-BB-induced SMC proliferation and migration as determined by bromodeoxyuridine (BrdU) incorporation and a cell counting assay as well as a Boyden chamber assay respectively. DAPK3 siRNA or a pharmacological inhibitor of DAPK3 inhibited PDGF-BB-induced lamellipodia formation as determined by rhodamine-phalloidin staining. DAPK3 siRNA or the DAPK inhibitor significantly reduced PDGF-BB-induced activation of p38 and heat-shock protein 27 (HSP27) as determined by Western blotting. In ex vivo studies, PDGF-BB-induced SMC out-growth was significantly inhibited by the DAPK inhibitor. In vivo, the DAPK inhibitor significantly prevented carotid neointimal hyperplasia in a mouse ligation model. The present results, for the first time, revealed that DAPK3 mediates PDGF-BB-induced SMC proliferation and migration through activation of p38/HSP27 signals, which may lead to vascular structural remodelling including neointimal hyperplasia. The present study suggests DAPK3 as a novel pharmaceutical target for the prevention of hypertensive cardiovascular diseases.


Assuntos
Movimento Celular/genética , Proliferação de Células , Proteínas Quinases Associadas com Morte Celular/fisiologia , Animais , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/patologia , Contagem de Células , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Proteínas Quinases Associadas com Morte Celular/antagonistas & inibidores , Proteínas Quinases Associadas com Morte Celular/genética , Proteínas Quinases Associadas com Morte Celular/metabolismo , Humanos , Hiperplasia/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/enzimologia , Neointima/patologia , Oxazolona/análogos & derivados , Oxazolona/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Ratos , Ratos Wistar , Transdução de Sinais
13.
Am J Physiol Heart Circ Physiol ; 306(12): H1714-9, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24727494

RESUMO

Omentin is a novel adipocytokine expressed in visceral adipose tissue. Secretion and blood concentration of omentin decrease in the obese subjects. We previously demonstrated that omentin is anti-inflammatory in vascular smooth muscle cells (SMCs). While vascular remodeling via migration of SMCs is also important for hypertension development, it remains to be clarified whether omentin affects this process. Here we examined whether omentin controls SMC migration. Omentin (300 ng/ml, 2 h) significantly inhibited platelet-derived growth factor (PDGF)-BB (10 ng/ml, 6 h)-induced migration of rat mesenteric arterial SMCs, as determined by Boyden chamber assay. Omentin (300 ng/ml, 2 h) significantly inhibited PDGF-BB (10 ng/ml, 30 min)-induced phosphorylation of p38 and heat shock protein (HSP) 27. Omentin (300 ng/ml, 2 h) significantly inhibited PDGF-BB (10 ng/ml, 30 min)-induced NADPH oxidase (NOX) activation as determined by lucigenin assay. Omentin (300 ng/ml, 24 h) significantly inhibited fetal bovine serum (5%, 4 days)-induced SMC outgrowth from rat isolated mesenteric artery. In vivo, omentin significantly inhibited carotid intimal hyperplasia in mouse ligation model. In summary, we for the first time demonstrate that omentin prevents PDGF-BB-induced SMC migration by preventing NOX/O2(-)/p38/HSP27 pathways, which might be at least partly responsible for the preventive effects on neointimal hyperplasia. Our data suggest that omentin may be protective against hypertension development by inhibiting vascular structural remodeling.


Assuntos
Adipocinas/farmacologia , Antioxidantes/fisiologia , Movimento Celular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis/farmacologia , Transdução de Sinais/fisiologia , Adipocinas/fisiologia , Animais , Becaplermina , Artérias Carótidas/citologia , Artérias Carótidas/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Citocinas/farmacologia , Proteínas Ligadas por GPI , Proteínas de Choque Térmico HSP27/fisiologia , Técnicas In Vitro , Lectinas/farmacologia , Masculino , Artérias Mesentéricas/citologia , Artérias Mesentéricas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Miócitos de Músculo Liso/citologia , NADPH Oxidases/fisiologia , Oxigênio/fisiologia , Proteínas Proto-Oncogênicas c-sis/fisiologia , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
14.
J Vet Med Sci ; 75(8): 1029-34, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23546685

RESUMO

Omentin is a recently identified adipocytokine, and we previously demonstrated that omentin played anti-inflammatory roles in vascular endothelial and smooth muscle cells. We also demonstrated that omentin induced vasodilation in rat isolated blood vessels. However, effects of omentin on blood pressure (BP) are not determined. Here, we examined whether intravenously injected omentin acutely alters BP of Wistar rats. Omentin (0.06-18 µg/kg) alone did not alter BP of Wistar rats. On the other hand, omentin (18 µg/kg) significantly inhibited noradrenaline (NA; 2 µg/kg)-induced increases in systolic BP and mean BP. Omentin (18 µg/kg) significantly inhibited angiotensin II (1 µg/kg)-induced increases in diastolic BP. Omentin (18 µg/kg) significantly inhibited dimorpholamine (3 mg/kg)-induced increases in diastolic BP. Omentin (18 µg/kg) failed to inhibit the NA (0.02-2 µg/kg)-induced increases of systolic BP in the nitric oxide (NO) synthase inhibitor, N(G)-nitro-l-arginine methyl ester (80 mg/kg, 1 day)-treated Wistar rats. In summary, we for the first time demonstrated that omentin inhibited agonists-induced increases in BP. The effect of omentin was suggested to be mediated likely via NO-dependent mechanism.


Assuntos
Angiotensinas/antagonistas & inibidores , Pressão Sanguínea/efeitos dos fármacos , Citocinas/farmacologia , Lectinas/farmacologia , Norepinefrina/antagonistas & inibidores , Análise de Variância , Animais , Citocinas/administração & dosagem , Proteínas Ligadas por GPI/administração & dosagem , Proteínas Ligadas por GPI/farmacologia , Injeções Intravenosas , Lectinas/administração & dosagem , Morfolinas/antagonistas & inibidores , Ácido Nítrico/metabolismo , Ratos , Ratos Wistar
15.
Eur J Pharmacol ; 686(1-3): 116-23, 2012 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-22554771

RESUMO

Omentin is a recently identified adipocytokine and its effect in vasculature is largely unknown. Here we examined the effects of omentin on smooth muscle cells (SMCs) inflammatory states. Western blotting was performed to analyze inflammatory signal transduction in cultured SMCs. Phosphorylation of nuclear factor-κB (NF-κB), p38 and JNK, and expression of vascular cell adhesion molecule (VCAM)-1 and cyclooxygenase-2 were not induced by omentin (50-300ng/ml, 20min or 24h). On the other hand, tumor necrosis factor-α (TNF-α; 10ng/ml, 20min)-induced phosphorylation of p38 and JNK was significantly inhibited by omentin pretreatment in a concentration-dependent manner (50-300ng/ml, 30min). TNF-α (24h)-induced expression of VCAM-1 was also significantly inhibited by omentin pretreatment in a concentration-dependent manner. Both inhibitor of p38 (SB203580) and JNK (SP600125) significantly inhibited TNF-α-induced VCAM-1 expression. Omentin (300ng/ml, 30min) inhibited TNF-α (1h)-induced nicotinamide adenine dinucleotide phosphate oxidase activity as determined by lucigenin assay. An antioxidant drug, N-acetyl-l-cysteine significantly inhibited TNF-α-induced phosphorylation of p38 and JNK. Furthermore, omentin (300ng/ml, 30min) significantly inhibited TNF-α (24h)-induced monocytic cells adhesion to SMCs. In rat isolated thoracic aorta, omentin (300ng/ml, 30min) inhibited TNF-α (24h)-induced VCAM-1 expression. The present results demonstrate for the first time that omentin plays an anti-inflammatory role by preventing the TNF-α-induced VCAM-1 expression in SMCs. It is suggested that omentin inhibits TNF-α-induced VCAM-1 expression via preventing the activation of p38 and JNK at least in part through inhibition of superoxide production.


Assuntos
Anti-Inflamatórios/farmacologia , Citocinas/farmacologia , Lectinas/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Superóxidos/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiologia , Adesão Celular/efeitos dos fármacos , Proteínas Ligadas por GPI/farmacologia , Humanos , Técnicas In Vitro , Proteínas Quinases JNK Ativadas por Mitógeno/imunologia , Masculino , Monócitos/efeitos dos fármacos , Monócitos/fisiologia , Músculo Liso Vascular/fisiologia , Ratos , Ratos Wistar , Proteínas Recombinantes/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Células U937 , Molécula 1 de Adesão de Célula Vascular/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
16.
Otolaryngol Head Neck Surg ; 138(6): 738-42, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18503846

RESUMO

OBJECTIVE: To evaluate the effect of our large-sized silicone sheet upon postoperative recovery of mastoid aeration in ears after surgery including mastoidectomy and soft-wall reconstruction (SWR). STUDY DESIGN AND SETTING: Retrospective chart review in a tertiary care center. SUBJECTS AND METHODS: Recovery of mastoid aeration was assessed by CT 4 to 12 months after surgery on 72 ears (69 patients), in which the silicone sheet covering from the eustachian tube (ET) to the mastoid was placed after SWR procedure with mastoidectomy were done for their chronic otitis media. Results were compared with those with a small silicone sheet. RESULTS: Recovery of mastoid aeration was significantly better in the large-silicone-sheet group than in the small-silicone-sheet group (chi(2) value = 11.7146, P = 0.0006). CONCLUSION: This preliminary study suggested that our large-sized silicone sheet may be effective for postoperative recovery of mastoid aeration even in ears operated with SWR procedure.


Assuntos
Colesteatoma da Orelha Média/cirurgia , Processo Mastoide/cirurgia , Prótese Ossicular , Silicones , Timpanoplastia/instrumentação , Adolescente , Adulto , Idoso , Criança , Colesteatoma da Orelha Média/patologia , Estudos de Coortes , Meato Acústico Externo/cirurgia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Desenho de Prótese , Estudos Retrospectivos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA