Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 40(3): 583-596, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31996026

RESUMO

OBJECTIVE: Inflammatory activation changes the mitochondrial function of macrophages from oxidative phosphorylation to reactive oxygen species production, which may promote necrotic core formation in atherosclerotic lesions. In hypoxic and cancer cells, HIF-1α (hypoxia-inducible factor) promotes oxygen-independent energy production by microRNAs. Therefore, we studied the role of HIF-1α in the regulation of macrophage energy metabolism in the context of atherosclerosis. Approach and Results: Myeloid cell-specific deletion of Hif1a reduced atherosclerosis and necrotic core formation by limiting macrophage necroptosis in apolipoprotein E-deficient mice. In inflammatory bone marrow-derived macrophages, deletion of Hif1a increased oxidative phosphorylation, ATP levels, and the expression of genes encoding mitochondrial proteins and reduced reactive oxygen species production and necroptosis. microRNA expression profiling showed that HIF-1α upregulates miR-210 and downregulates miR-383 levels in lesional macrophages and inflammatory bone marrow-derived macrophages. In contrast to miR-210, which inhibited oxidative phosphorylation and enhanced mitochondrial reactive oxygen species production, miR-383 increased ATP levels and inhibited necroptosis. The effect of miR-210 was due to targeting 2,4-dienoyl-CoA reductase, which is essential in the ß oxidation of unsaturated fatty acids. miR-383 affected the DNA damage repair pathway in bone marrow-derived macrophages by targeting poly(ADP-ribose)-glycohydrolase (Parg), which reduced energy consumption and increased cell survival. Blocking the targeting of Parg by miR-383 prevented the protective effect of Hif1a deletion in macrophages on atherosclerosis and necrotic core formation in mice. CONCLUSIONS: Our findings unveil a new mechanism by which activation of HIF-1α in inflammatory macrophages increases necroptosis through microRNA-mediated ATP depletion, thus increasing atherosclerosis by necrotic core formation.


Assuntos
Aorta/metabolismo , Aterosclerose/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/metabolismo , Macrófagos/metabolismo , MicroRNAs/metabolismo , Necroptose , Trifosfato de Adenosina/metabolismo , Animais , Aorta/patologia , Aterosclerose/genética , Aterosclerose/patologia , Células Cultivadas , Modelos Animais de Doenças , Metabolismo Energético , Regulação da Expressão Gênica , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Inflamação/genética , Inflamação/patologia , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , MicroRNAs/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
2.
Diabetes ; 66(12): 3072-3084, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28970282

RESUMO

A high-fat diet increases bacterial lipopolysaccharide (LPS) in the circulation and thereby stimulates glucagon-like peptide 1 (GLP-1)-mediated insulin secretion by upregulating interleukin-6 (IL-6). Although microRNA-155-5p (miR-155-5p), which increases IL-6 expression, is upregulated by LPS and hyperlipidemia and patients with familial hypercholesterolemia less frequently develop diabetes, the role of miR-155-5p in the islet stress response to hyperlipidemia is unclear. In this study, we demonstrate that hyperlipidemia-associated endotoxemia upregulates miR-155-5p in murine pancreatic ß-cells, which improved glucose metabolism and the adaptation of ß-cells to obesity-induced insulin resistance. This effect of miR-155-5p is because of suppression of v-maf musculoaponeurotic fibrosarcoma oncogene family, protein B, which promotes ß-cell function through IL-6-induced GLP-1 production in α-cells. Moreover, reduced GLP-1 levels are associated with increased obesity progression, dyslipidemia, and atherosclerosis in hyperlipidemic Mir155 knockout mice. Hence, induction of miR-155-5p expression in ß-cells by hyperlipidemia-associated endotoxemia improves the adaptation of ß-cells to insulin resistance and represents a protective mechanism in the islet stress response.


Assuntos
Hiperlipidemias/genética , Células Secretoras de Insulina/fisiologia , Fator de Transcrição MafB/genética , MicroRNAs/fisiologia , Animais , Células Cultivadas , Endotoxemia/genética , Peptídeo 1 Semelhante ao Glucagon/sangue , Glucose/metabolismo , Insulina/biossíntese , Interleucina-6/genética , Camundongos , Camundongos Endogâmicos C57BL , Receptores de LDL/fisiologia
3.
J Cell Mol Med ; 21(8): 1523-1531, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28211187

RESUMO

Platelets play an important role in the pathogenesis of vascular remodelling after injury. Junctional adhesion molecule A (JAM-A) was recently described to regulate platelet activation. Specific deletion of JAM-A from platelets resulted in increased reactivity and in accelerated progression of atherosclerosis. The aim of this study was to investigate the specific contribution of platelet-derived JAM-A to neointima formation after vascular injury. Mice with or without platelet-specific (tr)JAM-A-deficiency in an apolipoprotein e (apoe-/- ) background underwent wire-induced injury of the common carotid artery. Ex vivo imaging by two-photon microscopy revealed increased platelet coverage at the site of injury in trJAM-A-deficient mice. Cell recruitment assays showed increased adhesion of monocytic cells to activated JAM-A-deficient platelets than to control platelets. Inhibition of αM ß2 or GPIbα, but not of CD62P, suppressed those differences. Up to 4 weeks after wire injury, intimal neoplasia and neointimal cellular content were analysed. Neointimal lesion area was increased in trJAM-A-/- apoe-/- mice and the lesions showed an increased macrophage accumulation and proliferating smooth muscle cells compared with trJAM-A+/+ apoe-/- littermates 2 weeks, but not 4 weeks after injury. Re-endothelialization was decreased in trJAM-A-/- apoe-/- mice compared with controls 2 weeks after injury, yet it was complete in both groups after 4 weeks. A platelet gain of function by deletion of JAM-A accelerates neointima formation only during earlier phases after vascular injury, through an increased recruitment of mononuclear cells. Thus, the contribution of platelets might become less important when neointima formation progresses to later stages.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/genética , Lesões das Artérias Carótidas/genética , Moléculas de Adesão Celular/genética , Hiperlipidemias/genética , Neointima/genética , Receptores de Superfície Celular/genética , Animais , Apolipoproteínas E/deficiência , Aterosclerose/complicações , Aterosclerose/metabolismo , Aterosclerose/patologia , Plaquetas/metabolismo , Plaquetas/patologia , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/complicações , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Adesão Celular , Moléculas de Adesão Celular/deficiência , Feminino , Regulação da Expressão Gênica , Hiperlipidemias/complicações , Hiperlipidemias/metabolismo , Hiperlipidemias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Monócitos/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Neointima/complicações , Neointima/metabolismo , Neointima/patologia , Receptores de Superfície Celular/deficiência , Transdução de Sinais , Remodelação Vascular/genética
4.
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
5.
Circ Res ; 116(4): 587-99, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25472975

RESUMO

RATIONALE: Besides their essential role in hemostasis, platelets also have functions in inflammation. In platelets, junctional adhesion molecule (JAM)-A was previously identified as an inhibitor of integrin αIIbß3-mediated outside-in signaling and its genetic knockdown resulted in hyperreactivity. OBJECTIVE: This gain-of-function was specifically exploited to investigate the role of platelet hyperreactivity in plaque development. METHODS AND RESULTS: JAM-A-deficient platelets showed increased aggregation and cellular and sarcoma tyrosine-protein kinase activation. On αIIbß3 ligation, JAM-A was shown to be dephosphorylated, which could be prevented by protein tyrosine phosphatase nonreceptor type 1 inhibition. Mice with or without platelet-specific (tr)JAM-A-deficiency in an apolipoprotein e (apoe(-/-)) background were fed a high-fat diet. After ≤12 weeks of diet, trJAM-A(-/-)apoe-/- mice showed increased aortic plaque formation when compared with trJAM-A(+/+) apoe(-/-) controls, and these differences were most evident at early time points. At 2 weeks, the plaques of the trJAM-A(-/-) apoe(-/-) animals revealed increased macrophage, T cell, and smooth muscle cell content. Interestingly, plasma levels of chemokines CC chemokine ligand 5 and CXC-chemokine ligand 4 were increased in the trJAM-A(-/-) apoe(-/-)mice, and JAM-A-deficient platelets showed increased binding to monocytes and neutrophils. Whole-blood perfusion experiments and intravital microscopy revealed increased recruitment of platelets and monocytes to the inflamed endothelium in blood of trJAM-A(-/-) apoe(-/-)mice. Notably, these proinflammatory effects of JAM-A-deficient platelets could be abolished by the inhibition of αIIbß3 signaling in vitro. CONCLUSIONS: Deletion of JAM-A causes a gain-of-function in platelets, with lower activation thresholds and increased inflammatory activities. This leads to an increase of plaque formation, particularly in early stages of the disease.


Assuntos
Aorta/metabolismo , Doenças da Aorta/etiologia , Aterosclerose/etiologia , Plaquetas/metabolismo , Doenças das Artérias Carótidas/etiologia , Moléculas de Adesão Celular/deficiência , Hiperlipidemias/complicações , Agregação Plaquetária , Receptores de Superfície Celular/deficiência , Animais , Aorta/patologia , Doenças da Aorta/sangue , Doenças da Aorta/genética , Doenças da Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/patologia , Doenças das Artérias Carótidas/sangue , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/patologia , Adesão Celular , Moléculas de Adesão Celular/sangue , Moléculas de Adesão Celular/genética , Células Cultivadas , Quimiotaxia de Leucócito , Dieta Hiperlipídica , Modelos Animais de Doenças , Progressão da Doença , Feminino , Genótipo , Humanos , Hiperlipidemias/sangue , Hiperlipidemias/genética , Mediadores da Inflamação/metabolismo , Leucócitos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Placa Aterosclerótica , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Receptores de Superfície Celular/sangue , Receptores de Superfície Celular/genética , Trombose/sangue , Trombose/etiologia , Fatores de Tempo , Quinases da Família src/metabolismo
6.
Thromb Haemost ; 110(6): 1267-77, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24114205

RESUMO

The chemokine receptor CCR6 is expressed by various cell subsets implicated in atherogenesis, such as monocytes, Th17 and regulatory T cells. In order to further define the role of CCR6 in atherosclerosis, CCR6-deficient (Ccr6-/-) mice were crossed with low-density lipoprotein receptor-deficient (Ldlr-/-) mice to generate atherosclerosis-prone mice deficient in CCR6. Compared to Ldlr-/- controls, atherosclerotic burden in the aortic sinus and aorta were reduced in Ccr6-/-Ldlr-/- mice fed a high fat diet, associated with a profound depression in lesional macrophage accumulation. Local and systemic distributions of T cells, including frequencies of Th1, Th17 and regulatory T cells were unaltered. In contrast, circulating counts of both Gr-1(high) and Gr1(low) monocytes were reduced in Ccr6-/-Ldlr-/- mice. Moreover, CCR6 was revealed to promote monocyte adhesion to inflamed endothelium in vitro and leukocyte adhesion to carotid arteries in vivo. Finally, CCR6 selectively recruited monocytes but not T cells in an acute inflammatory air pouch model. We here show that CCR6 functions on multiple levels and regulates the mobilisation, adhesion and recruitment of monocytes/macrophages to the inflamed vessel, thereby promoting atherosclerosis, but is dispensable for hypercholesterolaemia-associated adaptive immune priming. Targeting CCR6 or its ligand CCL20 may therefore be a promising therapeutic strategy to alleviate atherosclerosis.


Assuntos
Aterosclerose/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Receptores CCR6/metabolismo , Linfócitos T/imunologia , Animais , Adesão Celular/genética , Movimento Celular/genética , Células Cultivadas , Dieta Hiperlipídica , Modelos Animais de Doenças , Suscetibilidade a Doenças , Inflamação/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CCR6/genética , Receptores CCR6/imunologia , Receptores de Superfície Celular/metabolismo , Receptores de LDL/genética
7.
Biochem Pharmacol ; 86(2): 279-85, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23665353

RESUMO

Two synthetic LXR agonists were recently reported to inhibit collagen-induced platelet aggregation and thrombus formation in mice. We therefore studied whether also natural LXR agonists inhibit human platelet activation and whether they can be fluorescence-labelled preserving their bioactivity for LXR-related functional imaging. The natural LXR agonist 22(R)-OH-cholesterol - but not its stereoisomer 22(S)-OH-cholesterol - inhibited collagen induced platelet shape change and aggregation similar to synthetic LXR agonists in a concentration- and time-dependent manner. First exposure to 22(S)-OH-cholesterol prevented the subsequent inhibition of platelets by 22(R)-OH-cholesterol but not vice versa. 22(R)- and 22(S)-OH-cholesterol could be fluorescence-labelled as 22(R)- and 22(S)-OH-cholesteryl-3-dodecanoic-3-BODIPY esters with high yield and purity using the Steglich acylation. Labelled 22(R)- and 22(S)-OH-cholesterol esters retained the stereo specific bioactivity of their parent compounds, were metabolically stable and not cytotoxic at LXR agonistic concentrations. Live staining with labelled 22(R)- or 22(S)-OH-cholesterol esters demonstrated stereo specific inhibition of platelet spreading and chiral handling by macrophages that reflect LXR activation. The rapid inhibition of platelet reactivity to collagen by natural and pharmacologic LXR agonists offers a mechanism that could attenuate platelet activation by denuded plaques that expose collagen and LXR agonistic oxysterols. Stable fluorescence labelled 22(R)- and 22(S)-OH-cholesterol analogues with preserved stereo specific bioactivity and staining characteristics provide valuable tools for LXR-related functional imaging in pathophysiologic studies, for binding assays and for LXR-targeted drug development.


Assuntos
Hidroxicolesteróis/farmacologia , Macrófagos/efeitos dos fármacos , Receptores Nucleares Órfãos/agonistas , Ativação Plaquetária/efeitos dos fármacos , Acilação , Corantes Fluorescentes , Humanos , Cinética , Receptores X do Fígado , Macrófagos/metabolismo , Microscopia de Fluorescência , Estereoisomerismo
8.
Cell Metab ; 13(5): 592-600, 2011 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-21531341

RESUMO

Oxidatively modified low-density lipoprotein (oxLDL) plays a key role in the initiation of atherosclerosis by increasing monocyte adhesion. The mechanism that is responsible for the oxLDL-induced atherogenic monocyte recruitment in vivo, however, still remains unknown. Oxidation of LDL generates lysophosphatidylcholine, which is the main substrate for the lysophosphatidic acid (LPA) generating enzyme autotaxin. We show that oxLDL requires endothelial LPA receptors and autotaxin to elicit CXCL1-dependent arterial monocyte adhesion. Unsaturated LPA releases endothelial CXCL1, which is subsequently immobilized on the cell surface and mediates LPA-induced monocyte adhesion. Local and systemic application of LPA accelerates the progression of atherosclerosis in mice. Blocking the LPA receptors LPA(1) and LPA(3) reduced hyperlipidemia-induced arterial leukocyte arrest and atherosclerosis in the presence of functional CXCL1. Thus, atherogenic monocyte recruitment mediated by hyperlipidemia and modified LDL crucially depends on LPA, which triggers endothelial deposition of CXCL1, revealing LPA signaling as a target for cardiovascular disease treatments.


Assuntos
Aterosclerose/induzido quimicamente , Aterosclerose/metabolismo , Quimiocina CXCL1/metabolismo , Endotélio Vascular/metabolismo , Hiperlipidemias , Lipoproteínas LDL/metabolismo , Lisofosfolipídeos/farmacologia , Animais , Apolipoproteínas E/fisiologia , Artérias Carótidas/citologia , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/metabolismo , Células Cultivadas , Dieta Aterogênica , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , RNA Interferente Pequeno/genética , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/metabolismo
9.
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
10.
Trends Cardiovasc Med ; 16(4): 103-8, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16713531

RESUMO

Circulating smooth muscle progenitor cells have been identified as a source for neointimal smooth muscle cells after various types of injuries to the vessel wall contributing to neointimal hyperplasia, implying a fundamental role of these progenitor cells in the vascular response to injury. Recent studies have provided insight into the molecular mechanisms of mobilization and local recruitment of smooth muscle progenitor cells. The CXC chemokine SDF-1alpha and its receptor CXCR4 have been identified as the central signaling axis regulating the homing of smooth muscle progenitor cells into the injured vessel wall. This review discusses the unique biologic functions of this chemokine and its contribution to stem-cell-based vascular repair and disease.


Assuntos
Quimiocinas CXC/farmacologia , Transplante de Células-Tronco/métodos , Células-Tronco/efeitos dos fármacos , Doenças Vasculares/cirurgia , Animais , Quimiocina CXCL12 , Humanos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Resultado do Tratamento , Doenças Vasculares/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA