Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
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.
Immunol Res ; 72(1): 72-81, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37620509

RESUMO

Human γδ T cells are highly enriched in epithelial cell-dominated compartments like skin. Nonetheless, their function in the pathogenesis of pemphigus vulgaris (PV), an autoimmune skin disorder, is lacking. Therefore, we investigated the functional expression of human γδT cell subsets along with their homing chemokine receptor-ligand and inflammatory cytokines in the immunopathogenesis of PV. Estimation of the frequency of γδT cell subsets by flow cytometry revealed four major subsets of γδ T cells (γδT1, γδT2, γδT17, γδTreg) in both control and PV circulation. The elevated frequency of γδT17 cells producing IL17 and expressing CCR6 receptor suggests their inflammatory and migratory potential in PV. In vitro culture of γδ T cells from patients showed increased mRNA expression of inflammatory cytokines IL17, RORγt, IL23, IL1, and co-stimulatory markers, CD27 and CD70. These findings correlated the role of IL1 and IL23 cytokines that alleviate the Th17 population in PV. Cytotoxic activities of γδ T cells were higher and inflammatory γδT17 cells were localized in the skin of PV whereas γδTreg cells associated TGFß and FOXP3 were lowered. Hyperinflammatory phenotype of the γδT17 cell subset and its migratory potential might be aiding in the pathogenesis of PV, whereas γδTreg cells fail to suppress these inflammatory responses. Hence, γδT17 cell-associated markers can be targeted for identifying novel therapeutics in PV.


Assuntos
Pênfigo , Dermatopatias , Humanos , Interleucina-17/genética , Pele/metabolismo , Citocinas/metabolismo
3.
Int Rev Immunol ; 41(2): 123-134, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33439070

RESUMO

Vascular endothelial dysfunction (ED) forms the cornerstone in the development of atherosclerotic lesions that clinically manifest as ischemia, myocardial infarction, stroke or peripheral arterial disease. ED can be triggered by various risk factors including hypercholesterolemia, hypertension, hyperhomocystenemia and chronic low-grade inflammation. These risk factors also activate immune response systemically. Current drugs used for managing atherosclerosis not only aid in subsiding the risk factor but also suppress the immune activation. Nonetheless, their effectiveness in treating ED is still questionable. Here, we discuss how pathologic molecules and processes pertaining to ED can activate innate and adaptive arms of the immune system leading to disease progression even in the absence of cardiovascular risk factors and the potential of the current drugs, used in the management of atherosclerotic patients, in reversing them. We mainly focus on activated endothelium, endothelial microparticles, mechanically stretched endothelial cells, endothelial mesenchymal transition and endothelial glycocalyx sheds.


Assuntos
Aterosclerose , Células Endoteliais , Aterosclerose/tratamento farmacológico , Células Endoteliais/patologia , Endotélio Vascular , Humanos , Imunidade
4.
Cytokine Growth Factor Rev ; 45: 35-44, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30773437

RESUMO

Autoimmune skin diseases are a group of disorders that arise due to a deregulated immune system resulting in skin tissue destruction. In the majority of these conditions, either autoreactive immune cells or the autoantibodies are generated against self-antigens of the skin. Although the etiology of these diseases remains elusive, biochemical, genetic, and environmental factors such as infectious agents, toxins damage the skin tissue leading to self-antigen generation, autoantibody attack and finally results in autoimmunity of skin. Immune dysregulation, which involves predominantly T helper 1/17 (Th1/Th17) polarization and the inability of regulatory T cells to regress immune response, is implicated in autoimmune skin diseases. The emerging roles of immune cells, cytokines, and chemokines in the pathogenesis of common autoimmune skin diseases like pemphigus, vitiligo, and psoriasis are discussed in this review. The main focus is on the interplay between immune cell network including the innate and adaptive immune system, regulatory cells, immune checkpoints and recently identified tissue-resident memory cells (TRMs) in disease pathogenesis and relapse. We also attempt to highlight on the immune mechanisms common to these diseases which can be targeted for designing novel therapeutics.


Assuntos
Doenças Autoimunes/imunologia , Pênfigo/imunologia , Psoríase/imunologia , Pele/imunologia , Pele/patologia , Vitiligo/imunologia , Imunidade Adaptativa , Animais , Autoanticorpos/imunologia , Doenças Autoimunes/patologia , Citocinas/imunologia , Humanos , Imunidade Inata , Camundongos , Pênfigo/patologia , Psoríase/patologia , Linfócitos T Reguladores/imunologia , Vitiligo/patologia
5.
Arterioscler Thromb Vasc Biol ; 37(11): 2087-2101, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28882872

RESUMO

OBJECTIVE: Atherosclerosis develops near branches and bends of arteries that are exposed to low shear stress (mechanical drag). These sites are characterized by excessive endothelial cell (EC) proliferation and inflammation that promote lesion initiation. The transcription factor HIF1α (hypoxia-inducible factor 1α) is canonically activated by hypoxia and has a role in plaque neovascularization. We studied the influence of shear stress on HIF1α activation and the contribution of this noncanonical pathway to lesion initiation. APPROACH AND RESULTS: Quantitative polymerase chain reaction and en face staining revealed that HIF1α was expressed preferentially at low shear stress regions of porcine and murine arteries. Low shear stress induced HIF1α in cultured EC in the presence of atmospheric oxygen. The mechanism involves the transcription factor nuclear factor-κB that induced HIF1α transcripts and induction of the deubiquitinating enzyme Cezanne that stabilized HIF1α protein. Gene silencing revealed that HIF1α enhanced proliferation and inflammatory activation in EC exposed to low shear stress via induction of glycolysis enzymes. We validated this observation by imposing low shear stress in murine carotid arteries (partial ligation) that upregulated the expression of HIF1α, glycolysis enzymes, and inflammatory genes and enhanced EC proliferation. EC-specific genetic deletion of HIF1α in hypercholesterolemic apolipoprotein E-defecient mice reduced inflammation and endothelial proliferation in partially ligated arteries, indicating that HIF1α drives inflammation and vascular dysfunction at low shear stress regions. CONCLUSIONS: Mechanical low shear stress activates HIF1α at atheroprone regions of arteries via nuclear factor-κB and Cezanne. HIF1α promotes atherosclerosis initiation at these sites by inducing excessive EC proliferation and inflammation via the induction of glycolysis enzymes.


Assuntos
Aterosclerose/metabolismo , Células Endoteliais/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/metabolismo , Mecanotransdução Celular , Placa Aterosclerótica , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Endopeptidases/metabolismo , Células Endoteliais/patologia , Indução Enzimática , Feminino , Predisposição Genética para Doença , Glicólise , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Inflamação/genética , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Camundongos Knockout , NF-kappa B/metabolismo , Oxigênio/metabolismo , Fenótipo , Estabilidade Proteica , Proteólise , Interferência de RNA , Fluxo Sanguíneo Regional , Estresse Mecânico , Sus scrofa , Fatores de Tempo , Transfecção , Ubiquitinação , Regulação para Cima
6.
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
7.
J Proteomics ; 127(Pt A): 50-60, 2015 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-25956427

RESUMO

Cardiovascular disease has remained as the largest cause of morbidity and mortality worldwide. From dissecting the disease aetiology to identifying prognostic markers for better management of the disease is still a challenge for researchers. In the post human genome sequencing era much of the thrust has been focussed towards application of advanced genomic tools along with evaluation of traditional risk factors. With the advancement of next generation proteomics and metabolomics approaches it has now become possible to understand the protein interaction network & metabolic rewiring which lead to the perturbations of the disease phenotype. Further, elucidating different post translational modifications using advanced mass spectrometry based methods have provided an impetus towards in depth understanding of the proteome. The past decade has observed a plethora of studies where proteomics has been applied successfully to identify potential prognostic and diagnostic markers as well as to understand the disease mechanisms for various types of cardiovascular diseases. In this review, we attempted to document relevant proteomics based studies that have been undertaken either to identify potential biomarkers or have elucidated newer mechanistic insights into understanding the patho-physiology of cardiovascular disease, primarily coronary artery disease, cardiomyopathy, and myocardial ischemia. We have also provided a perspective on the potential of proteomics in combating this deadly disease. BIOLOGICAL SIGNIFICANCE: This review has catalogued recent studies on proteomics and metabolomics involved in understanding several cardiovascular diseases (CVDs). A holistic systems biology based approach, of which proteomics and metabolomics are two very important components, would help in delineating various pathways associated with complex disorders like CVD. This would ultimately provide better mechanistic understanding of the disease biology leading to development of prognostic biomarkers. This article is part of a Special Issue entitled: Proteomics in India.


Assuntos
Doenças Cardiovasculares/metabolismo , Metabolômica/métodos , Metabolômica/tendências , Proteômica/métodos , Proteômica/tendências , Animais , Biomarcadores/metabolismo , Humanos
8.
Arterioscler Thromb Vasc Biol ; 34(6): 1209-20, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24723559

RESUMO

OBJECTIVE: The Cxcl12/Cxcr4 chemokine ligand/receptor axis mediates the mobilization of smooth muscle cell progenitors, driving injury-induced neointimal hyperplasia. This study aimed to investigate the role of endothelial Cxcr4 in neointima formation. APPROACH AND RESULTS: ß-Galactosidase staining using bone marrow x kinase (Bmx)-CreER(T2) reporter mice and double immunofluorescence revealed an efficient and endothelial-specific deletion of Cxcr4 in Bmx-CreER(T2+) compared with Bmx-CreER(T2-) Cxcr4-floxed apolipoprotein E-deficient (Apoe(-/-)) mice (referred to as Cxcr4(EC-KO)ApoE(-/-) and Cxcr4(EC-WT) ApoE(-/-), respectively). Endothelial Cxcr4 deficiency significantly increased wire injury-induced neointima formation in carotid arteries from Cxcr4(EC-KO)ApoE(-/-) mice. The lesions displayed a higher number of macrophages, whereas the smooth muscle cell and collagen content were reduced. This was associated with a significant reduction in reendothelialization and endothelial cell proliferation in injured Cxcr4(EC-KO)ApoE(-/-) carotids compared with Cxcr4(EC-WT)ApoE(-/-) controls. Furthermore, stimulation of human aortic endothelial cells with chemokine (C-X-C motif) ligand 12 (CXCL12) significantly enhanced their wound-healing capacity in an in vitro scratch assay, an effect that could be reversed with the CXCR4 antagonist AMD3100. Also, flow cytometric analysis showed a reduced mobilization of Sca1(+)Flk1(+)Cd31(+) and of Lin(-)Sca1(+) progenitors in Cxcr4(EC-KO) ApoE(-/-) mice after vascular injury, although Cxcr4 surface expression was unaltered. No differences could be detected in plasma concentrations of Cxcl12, vascular endothelial growth factor, sphingosine 1-phosphate, or Flt3 (fms-related tyrosine kinase 3) ligand, all cytokines with an established role in progenitor cell mobilization. Nonetheless, double immunofluorescence revealed a significant reduction in local endothelial Cxcl12 staining in injured carotids from Cxcr4(EC-KO)ApoE(-/-) mice. CONCLUSIONS: Endothelial Cxcr4 is crucial for efficient reendothelialization after vascular injury through endothelial wound healing and proliferation, and through the mobilization of Sca1(+)Flk1(+)Cd31(+) cells, often referred to as circulating endothelial progenitor cells.


Assuntos
Aterosclerose/patologia , Lesões das Artérias Carótidas/patologia , Células Endoteliais/fisiologia , Neointima/patologia , Receptores CXCR4/fisiologia , Animais , Antígenos Ly/fisiologia , Apolipoproteínas E/fisiologia , Aterosclerose/fisiopatologia , Movimento Celular , Quimiocina CXCL12/fisiologia , Feminino , Hiperplasia , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Tirosina Quinases/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia
9.
Circulation ; 129(11): 1244-53, 2014 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-24374972

RESUMO

BACKGROUND: The aim of this study was to determine the role of the chemokine receptor CXCR7 in atherosclerosis and vascular remodeling. CXCR7 is the alternative receptor of CXCL12, which regulates stem cell-mediated vascular repair and limits atherosclerosis via its receptor, CXCR4. METHODS AND RESULTS: Wire-induced injury of the carotid artery was performed in mice with a ubiquitous, conditional deletion of CXCR7 and in mice treated with the synthetic CXCR7 ligand CCX771. The effect of CCX771 treatment on atherosclerosis was studied in apolipoprotein E-deficient (Apoe(-/-)) mice fed a high-fat diet for 12 weeks. Lipoprotein fractions were quantified in the plasma of Apoe(-/-) mice by fast protein liquid chromatography. Uptake of DiI-labeled very low-density lipoprotein to adipose tissue was determined by 2-photon microscopy. We show that genetic deficiency of Cxcr7 increased neointima formation and lesional macrophage accumulation in hyperlipidemic mice after vascular injury. This was related to increased serum cholesterol levels and subsequent hyperlipidemia-induced monocytosis. Conversely, administration of the CXCR7 ligand CCX771 to Apoe(-/-) mice inhibited lesion formation and ameliorated hyperlipidemia after vascular injury and during atherosclerosis. Treatment with CCX771 reduced circulating very low-density lipoprotein levels but not low-density lipoprotein or high-density lipoprotein levels and increased uptake of very low-density lipoprotein into Cxcr7-expressing white adipose tissue. This effect of CCX771 was associated with an enhanced lipase activity and reduced expression of Angptl4 in adipose tissue. CONCLUSIONS: CXCR7 regulates blood cholesterol by promoting its uptake in adipose tissue. This unexpected cholesterol-lowering effect of CXCR7 is beneficial for atherosclerotic vascular diseases, presumably via amelioration of hyperlipidemia-induced monocytosis, and can be augmented with a synthetic CXCR7 ligand.


Assuntos
Tecido Adiposo/metabolismo , Aterosclerose/metabolismo , Colesterol/metabolismo , Hiperlipidemias/metabolismo , Receptores CXCR/biossíntese , Animais , Aterosclerose/prevenção & controle , Hiperlipidemias/prevenção & controle , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CXCR/agonistas
10.
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
11.
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
12.
Radiology ; 260(3): 709-17, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21788527

RESUMO

PURPOSE: To develop and evaluate a user-friendly tool to enable efficient, accurate, and reproducible quantification of blood vessel stenosis in computed tomographic (CT) and magnetic resonance (MR) angiographic data sets. MATERIALS AND METHODS: All clinical experiments were approved by the institutional review board, and informed patient consent was acquired. Animal experiments were approved by the governmental review committee on animal care. A virtual elastic sphere passes through a blood vessel specified by user-provided start and end points, and the adapting diameter over the course of the vessel is recorded. The program was tested in phantoms to determine the accuracy of diameter estimation, and it was applied in micro-CT data sets of mice with induced vessel stenosis. Dual-energy CT angiography and MR angiography were performed in 16 patients with carotid artery stenosis, and reproducibility and required reader time of this automated technique were compared with manual measurements. Additionally, the effect of dual-energy CT-based discrimination between iodine- and calcium-based enhancement was investigated. Differences between carotid artery diameters of mice and between automated and manual measurement durations were assessed with a paired t test. Reproducibility of stenosis scores was evaluated with the Fisher z test. RESULTS: Phantom diameters were determined with an average error of 0.094 mm. Diameters of normal and injured carotid arteries of mice were significantly different (P < .01). For patient data, automated interreader variability was significantly (P < .01) lower than manual intra- and interreader variability, while time efficiency was improved (P < .01). CONCLUSION: The virtual elastic sphere tool is applicable to CT, dual-energy CT, and MR angiography, and it improves reproducibility and efficiency over that achieved with manual stenosis measurements.


Assuntos
Angiografia Coronária/métodos , Imageamento Tridimensional/métodos , Angiografia por Ressonância Magnética/métodos , Interpretação de Imagem Radiográfica Assistida por Computador/métodos , Tomografia Computadorizada por Raios X/métodos , Idoso , Animais , Feminino , Humanos , Masculino , Camundongos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
13.
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
14.
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
...