Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nitric Oxide ; 146: 37-47, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38579899

RESUMO

AIM: The mechanism of NO bioavailability in endothelial dysfunction, the trigger for atherogenesis is still unclear as exogenous nitrate therapy fails to alleviate endothelial dysfunction. Recently, sialin, a nitrate transporter, has been linked to affect tissue nitrate/nitrite levels. Hence, we investigated the role of sialin in NO bioavailability in endothelial dysfunction. METHODS: Serum-starved HUVECs were stimulated with either TNFα or AT-2 for 24 h either alone or in the presence of autophagy inducer or autophagy inhibitor alone. Nitric oxide, nitrite, and nitrate levels were measured in cell supernatant and cell lysate. Quantitative real-time PCR, Annexin V-PI, and monocyte adhesion assays were performed. Immunofluorescence staining for sialin, vWF, and LC3 was performed. STRING database was used to create protein interacting partners for sialin. RESULTS: Sialin is strongly expressed in activated EC in vitro and atherosclerotic plaque as well as tumor neo-vessel ECs. Sialin mediates nitrate ion efflux and is negatively regulated by autophagy via mTOR pathway. Blocking sialin enhances NO bioavailability, autophagy, cell survival, and eNOS expression while decreasing monocyte adhesion. PPI shows LGALS8 to directly interact with sialin and regulate autophagy, cell-cell adhesion, and apoptosis. CONCLUSION: Sialin is a potential novel therapeutic target for treating endothelial dysfunction in atherosclerosis and cancer.


Assuntos
Autofagia , Células Endoteliais da Veia Umbilical Humana , Nitratos , Óxido Nítrico , Humanos , Óxido Nítrico/metabolismo , Nitratos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Inflamação/metabolismo , Adesão Celular , Sialomucinas/metabolismo
2.
Hypertension ; 66(6): 1220-6, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26483345

RESUMO

Chemokines mediate monocyte adhesion to dysfunctional endothelial cells (ECs) and promote arterial inflammation during atherosclerosis. Hypoxia-inducible factor (HIF)-1α is expressed in various cell types of atherosclerotic lesions and is associated with lesional inflammation. However, the impact of endothelial HIF-1α in atherosclerosis is unclear. HIF-1α was detectable in the nucleus of ECs covering murine and human atherosclerotic lesions. To study the role of endothelial HIF-1α in atherosclerosis, deletion of the Hif1a gene was induced in ECs from apolipoprotein E knockout mice (EC-Hif1a(-/-)) by Tamoxifen injection. The formation of atherosclerotic lesions, the lesional macrophage accumulation, and the expression of CXCL1 in ECs were reduced after partial carotid ligation in EC-Hif1a(-/-) compared with control mice. Moreover, the lesion area and the lesional macrophage accumulation were decreased in the aortas of EC-Hif1a(-/-) mice compared with control mice during diet-induced atherosclerosis. In vitro, mildly oxidized low-density lipoprotein or lysophosphatidic acid 20:4 increased endothelial CXCL1 expression and monocyte adhesion by inducing HIF-1α expression. Moreover, endothelial Hif1a deficiency resulted in downregulation of miR-19a in atherosclerotic arteries determined by microRNA profiling. In vitro, HIF-1α-induced miR-19a expression mediated the upregulation of CXCL1 in mildly oxidized low-density lipoprotein-stimulated ECs. These results indicate that hyperlipidemia upregulates HIF-1α expression in ECs by mildly oxidized low-density lipoprotein-derived unsaturated lysophosphatidic acid. Endothelial HIF-1α promoted atherosclerosis by triggering miR-19a-mediated CXCL1 expression and monocyte adhesion, indicating that inhibition of the endothelial HIF-1α/miR-19a pathway may be a therapeutic option against atherosclerosis.


Assuntos
Aterosclerose/genética , Células Endoteliais/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , MicroRNAs/genética , Monócitos/metabolismo , Animais , Aterosclerose/metabolismo , Western Blotting , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Células Endoteliais/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lipoproteínas LDL/farmacologia , Camundongos Knockout , Oligonucleotídeos Antissenso/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
Arterioscler Thromb Vasc Biol ; 33(4): 679-86, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23393393

RESUMO

OBJECTIVE: Unstable atherosclerotic lesions are prone to rupture, which leads to atherothrombosis. Chemokine (C-X-C motif) ligand 12 (CXCL12) promotes the mobilization and neointimal recruitment of smooth muscle progenitor cells (SPCs), and thereby mediates vascular repair. Moreover, treatment with SPCs stabilizes atherosclerotic lesions in mice. We investigated the role of CXCL12 in the treatment of unstable atherosclerotic lesions. APPROACH AND RESULTS: Intravenous injection of CXCL12 selectively increased the level of Sca1(+)Lin platelet derived growth factor receptor-ß(+) SPCs in the circulation as determined by flow cytometry. Macrophage-rich lesions were induced by partial ligation of the carotid artery in Apoe(-/-) mice. Repeated injection of CXCL12 reduced the macrophage content, increased the number of smooth muscle cells, increased the fibrous cap thickness, and increased the collagen content in these lesions. However, CXCL12 did not alter the lesion size or the luminal diameter of the carotid artery as determined by planimetry and micro-computed tomography, respectively. Recruitment of bone marrow-derived SPCs to the lesions was increased after treatment with CXCL12 in chimeric mice that expressed SM22-LacZ in bone marrow cells as determined by quantification of the number of lesional ß-galactosidase-expressing cells. CXCL12 expression was upregulated in atherosclerotic arteries after CXCL12 treatment. Silencing of arterial CXCL12 expression during atherosclerosis promoted lesion formation and reduced the lesional smooth muscle cell content in CXCL12-treated mice. CONCLUSIONS: Systemic treatment with CXCL12 promotes a more stable atherosclerotic lesion phenotype and enhances the accumulation of SPCs in these lesions without promoting atherosclerosis. Thus, CXCL12-induced SPC mobilization appears a promising approach to treat unstable atherosclerosis.


Assuntos
Apolipoproteínas E/deficiência , Artérias Carótidas/efeitos dos fármacos , Doenças das Artérias Carótidas/tratamento farmacológico , Quimiocina CXCL2/administração & dosagem , Miócitos de Músculo Liso/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Antígenos Ly/metabolismo , Apolipoproteínas E/genética , Biomarcadores/metabolismo , Transplante de Medula Óssea , Artérias Carótidas/imunologia , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/imunologia , Doenças das Artérias Carótidas/metabolismo , Doenças das Artérias Carótidas/patologia , Células Cultivadas , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Quimiotaxia/efeitos dos fármacos , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Células Endoteliais/patologia , Fibrose , Citometria de Fluxo , Genótipo , Injeções Intravenosas , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fenótipo , Placa Aterosclerótica , Proteínas Proto-Oncogênicas c-sis/metabolismo , Interferência de RNA , Células-Tronco/imunologia , Células-Tronco/metabolismo , Células-Tronco/patologia , Microtomografia por Raio-X
4.
J Clin Invest ; 122(11): 4190-202, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23041630

RESUMO

Macrophages in atherosclerotic plaques drive inflammatory responses, degrade lipoproteins, and phagocytose dead cells. MicroRNAs (miRs) control the differentiation and activity of macrophages by regulating the signaling of key transcription factors. However, the functional role of macrophage-related miRs in the immune response during atherogenesis is unknown. Here, we report that miR-155 is specifically expressed in atherosclerotic plaques and proinflammatory macrophages, where it was induced by treatment with mildly oxidized LDL (moxLDL) and IFN-γ. Leukocyte-specific Mir155 deficiency reduced plaque size and number of lesional macrophages after partial carotid ligation in atherosclerotic (Apoe-/-) mice. In macrophages stimulated with moxLDL/IFN-γ in vitro, and in lesional macrophages, loss of Mir155 reduced the expression of the chemokine CCL2, which promotes the recruitment of monocytes to atherosclerotic plaques. Additionally, we found that miR-155 directly repressed expression of BCL6, a transcription factor that attenuates proinflammatory NF-κB signaling. Silencing of Bcl6 in mice harboring Mir155-/- macrophages enhanced plaque formation and CCL2 expression. Taken together, these data demonstrated that miR-155 plays a key role in atherogenic programming of macrophages to sustain and enhance vascular inflammation.


Assuntos
Aterosclerose/metabolismo , Proteínas de Ligação a DNA/metabolismo , Macrófagos/metabolismo , MicroRNAs/metabolismo , Placa Aterosclerótica/metabolismo , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerose/genética , Aterosclerose/patologia , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/genética , Interferon gama/genética , Interferon gama/metabolismo , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , MicroRNAs/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Placa Aterosclerótica/genética , Placa Aterosclerótica/patologia , Proteínas Proto-Oncogênicas c-bcl-6 , Transdução de Sinais/genética , Vasculite/genética , Vasculite/metabolismo , Vasculite/patologia
5.
Eur Heart J ; 32(4): 476-84, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21036775

RESUMO

AIMS: We sought to determine whether circulating vascular progenitor cells, such as endothelial progenitor cells (EPCs) or smooth muscle progenitor cells (SPCs), were associated with the severity of cardiac allograft vasculopathy (CAV). METHODS AND RESULTS: CD34(+)CD140b(+) SPCs and CD34(+)KDR(+) EPCs were measured in the peripheral circulation of 187 adult heart transplant recipients by flow cytometry. Cardiac allograft vasculopathy was quantified by angiography using a CAV-specific scoring system. Cardiac allograft vasculopathy was present in 84 patients (44.7%) and was classified as mild in 59 and severe in 25 cases. Circulating SPCs were more frequently detectable in CAV patients than in patients without CAV. The number of CD34(+)CD140b(+) cells showed a stepwise increase in patients with moderate and severe CAV. Smooth muscle progenitor cell counts were higher in patients with coronary stent implant compared with unstented patients with CAV. In contrast, peripheral CD34(+)KDR(+) EPC counts were not changed in CAV patients. Plasma CXCL12 levels correlated with the degree of CAV and SPC counts. None of the different immunosuppressive drug regimes was related to the SPC count or the CXCL12 levels. A multivariate regression analysis revealed that the SPC count was independently associated with the presence of CAV. CONCLUSION: Circulating SPCs, but not EPCs, and plasma CXCL12 concentrations are elevated in CAV patients, indicating that they play prominent roles in transplant arteriosclerosis.


Assuntos
Antígenos CD34/metabolismo , Quimiocina CXCL12/metabolismo , Rejeição de Enxerto/etiologia , Transplante de Coração/patologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Células-Tronco/patologia , Adulto , Idoso , Proliferação de Células , Células Endoteliais/patologia , Endotélio Vascular/patologia , Feminino , Citometria de Fluxo , Rejeição de Enxerto/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Transplante Homólogo
6.
Circ Res ; 107(1): 96-105, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20360252

RESUMO

RATIONALE: The chemokine CXCL12 (CXC motif ligand 12) and its receptor CXCR 4 (CXC motif receptor 4) direct the recruitment of smooth muscle progenitor cells (SPCs) in neointima formation after vascular injury. Lysophosphatidic acid (LPA) induces CXCL12 and neointimal accumulation of smooth muscle cells (SMCs) in uninjured arteries. Thus, we hypothesize that LPA may regulate CXCL12-mediated vascular remodelling. OBJECTIVES: We evaluated the role of LPA receptors in initiating CXCL12-dependent vascular repair by SPCs. METHODS AND RESULTS: Wire-induced carotid injury was performed in apolipoprotein E(-/-) mice on western-type diet. LPA receptor expression was studied by immunostaining and quantitative RT-PCR. LPA receptors LPA(1) and LPA(3) were detected in the media of uninjured arteries and in the injury-induced neointima. LPA(3) mRNA was upregulated and LPA(1) mRNA downregulated at one week after injury. The LPA(1/3) antagonist Ki16425 inhibited neointima formation by 71% and reduced both relative neointimal SMCs and the macrophage content. Additionally, neointimal hypoxia-inducible factor-1alpha and CXCL12 expression, the injury-induced peripheral stem cell antigen-1 (Sca-1)(+)/Lin(-) SPC mobilization, and the neointimal recruitment of Sca-1(+)SMCs were inhibited by Ki16425. In wild type mice, LPA20:4 increased CXCL12 and hypoxia-inducible factor-1alpha expression in carotid arteries as early as 1 day following short-term endoluminal incubation. LPA20:4-induced SPC mobilization and neointima formation were blocked by Ki16425, LPA(1)- and LPA(3)-specific small interfering (si)RNA, and the CXCR4 antagonist POL5551. Ki16425 reduced LPA20:4-mediated neointimal recruitment of SPC as demonstrated by 2-photon microscopy in bone marrow chimeric mice after repopulation with SM22-LacZ transgenic, hematopoietic cells. Moreover, POL5551 decreased the neointimal accumulation of CXCR4(+) SMCs. CONCLUSIONS: LPA(1) and LPA(3) promote neointima formation through activation of CXCL12-mediated mobilization and recruitment of SPCs.


Assuntos
Movimento Celular/fisiologia , Quimiocina CXCL12/fisiologia , Miócitos de Músculo Liso/fisiologia , Receptores de Ácidos Lisofosfatídicos/fisiologia , Células-Tronco/fisiologia , Túnica Íntima/fisiologia , Animais , Artéria Carótida Primitiva/patologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/citologia , Células-Tronco/citologia , Túnica Íntima/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA