Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cardiovasc Res ; 117(13): 2652-2663, 2021 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-33751034

RESUMO

AIMS: Recent evidence suggests that 'vulnerable plaques', which have received intense attention as underlying mechanism of acute coronary syndromes over the decades, actually rarely rupture and cause clinical events. Superficial plaque erosion has emerged as a growing cause of residual thrombotic complications of atherosclerosis in an era of increased preventive measures including lipid lowering, antihypertensive therapy, and smoking cessation. The mechanisms of plaque erosion remain poorly understood, and we currently lack validated effective diagnostics or therapeutics for superficial erosion. Eroded plaques have a rich extracellular matrix, an intact fibrous cap, sparse lipid, and few mononuclear cells, but do harbour neutrophil extracellular traps (NETs). We recently reported that NETs amplify and propagate the endothelial damage at the site of arterial lesions that recapitulate superficial erosion in mice. We showed that genetic loss of protein arginine deiminase (PAD)-4 function inhibited NETosis and preserved endothelial integrity. The current study used systemic administration of targeted nanoparticles to deliver an agent that limits NETs formation to probe mechanisms of and demonstrate a novel therapeutic approach to plaque erosion that limits endothelial damage. METHODS AND RESULTS: We developed Collagen IV-targeted nanoparticles (Col IV NP) to deliver PAD4 inhibitors selectively to regions of endothelial cell sloughing and collagen IV-rich basement membrane exposure. We assessed the binding capability of the targeting ligand in vitro and evaluated Col IV NP targeting to areas of denuded endothelium in vivo in a mouse preparation that recapitulates features of superficial erosion. Delivery of the PAD4 inhibitor GSK484 reduced NET accumulation at sites of intimal injury and preserved endothelial continuity. CONCLUSIONS: NPs directed to Col IV show selective uptake and delivery of their payload to experimentally eroded regions, illustrating their translational potential. Our results further support the role of PAD4 and NETs in superficial erosion.


Assuntos
Aterosclerose/tratamento farmacológico , Colágeno Tipo IV/metabolismo , Portadores de Fármacos , Células Endoteliais/efeitos dos fármacos , Inibidores Enzimáticos/administração & dosagem , Armadilhas Extracelulares/metabolismo , Nanopartículas , Proteína-Arginina Desiminase do Tipo 4/antagonistas & inibidores , Animais , Aterosclerose/enzimologia , Aterosclerose/patologia , Membrana Basal/metabolismo , Técnicas de Cultura de Células em Três Dimensões , Células Cultivadas , Colágeno Tipo IV/química , Modelos Animais de Doenças , Composição de Medicamentos , Liberação Controlada de Fármacos , Células Endoteliais/enzimologia , Células Endoteliais/patologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , Camundongos Knockout para ApoE , Nanotecnologia , Placa Aterosclerótica , Ligação Proteica , Proteína-Arginina Desiminase do Tipo 4/metabolismo , Propriedades de Superfície , Distribuição Tecidual
2.
J Am Heart Assoc ; 9(15): e015878, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32750308

RESUMO

Background Activated vascular cells produce submicron prothrombotic and proinflammatory microparticle vesicles. Atherosclerotic plaques contain high levels of microparticles. Plasma microparticle levels increase during acute coronary syndromes and the thrombotic consequences of plaque rupture likely involve macrophage-derived microparticles (MΦMPs). The activation pathways that promote MΦMP production remain poorly defined. This study tested the hypothesis that signals implicated in atherogenesis also stimulate MΦMP production. Methods and Results We stimulated human primary MΦs with proinflammatory cytokines and atherogenic lipids, and measured MΦMP production by flow cytometry. Oxidized low-density lipoprotein (oxLDL; 25 µg/mL) induced MΦMP production in a concentration-dependent manner (293% increase; P<0.001), and these oxLDL MΦMP stimulatory effects were mediated by CD36. OxLDL stimulation increased MΦMP tissue factor content by 78% (P<0.05), and oxLDL-induced MΦMP production correlated with activation of caspase 3/7 signaling pathways. Salvionolic acid B, a CD36 inhibitor and a CD36 inhibitor antibody reduced oxLDL-induced MΦMP by 67% and 60%, respectively. Caspase 3/7 inhibition reduced MΦMP release by 52% (P<0.01) and caspase 3/7 activation increased MΦMP production by 208% (P<0.01). Mevastatin pretreatment (10 µM) decreased oxLDL-induced caspase 3/7 activation and attenuated oxLDL-stimulated MΦMP production and tissue factor content by 60% (P<0.01) and 43% (P<0.05), respectively. Conclusions OxLDL induces the production of prothrombotic microparticles in macrophages. This process depends on caspases 3 and 7 and CD36 and is inhibited by mevastatin pretreatment. These findings link atherogenic signaling pathways, inflammation, and plaque thrombogenicity and identify a novel potential mechanism for antithrombotic effects of statins independent of LDL lowering.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Lipoproteínas LDL/farmacologia , Macrófagos/metabolismo , Trombose/etiologia , Citometria de Fluxo , Humanos , Lipoproteínas LDL/metabolismo , Macrófagos/efeitos dos fármacos , Trombina/metabolismo , Tromboplastina/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 38(8): 1901-1912, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29976772

RESUMO

Objective- Coronary artery thrombosis can occur in the absence of plaque rupture because of superficial erosion. Erosion-prone atheromata associate with more neutrophil extracellular traps (NETs) than lesions with stable or rupture-prone characteristics. The effects of NETs on endothelial cell (EC) inflammatory and thrombogenic properties remain unknown. We hypothesized that NETs alter EC functions related to erosion-associated thrombosis. Approach and Results- Exposure of human ECs to NETs increased VCAM-1 (vascular cell adhesion molecule 1) and ICAM-1 (intercellular adhesion molecule 1) mRNA and protein expression in a time- and concentration-dependent manner. THP-1 monocytoid cells and primary human monocytes bound more avidly to NET-treated human umbilical vein ECs than to unstimulated cells under flow. Treatment of human ECs with NETs augmented the expression of TF (tissue factor) mRNA, increased EC TF activity, and hastened clotting of recalcified plasma. Anti-TF-neutralizing antibody blocked NET-induced acceleration of clotting by ECs. NETs alone did not exhibit TF activity or acceleration of clotting in cell-free assays. Pretreatment of NETs with anti-interleukin (IL)-1α-neutralizing antibody or IL-1Ra (IL-1 receptor antagonist)-but not with anti-IL-1ß-neutralizing antibody or control IgG-blocked NET-induced VCAM-1, ICAM-1, and TF expression. Inhibition of cathepsin G, a serine protease abundant in NETs, also limited the effect of NETs on EC activation. Cathepsin G potentiated the effect of IL-1α on ECs by cleaving the pro-IL-1α precursor and releasing the more potent mature IL-1α form. Conclusions- NETs promote EC activation and increased thrombogenicity through concerted action of IL-1α and cathepsin G. Thus, NETs may amplify and propagate EC dysfunction related to thrombosis because of superficial erosion.


Assuntos
Coagulação Sanguínea , Catepsina G/metabolismo , Armadilhas Extracelulares/enzimologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Interleucina-1alfa/metabolismo , Neutrófilos/enzimologia , Comunicação Parácrina , Tromboplastina/metabolismo , Adesão Celular , Técnicas de Cocultura , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Transdução de Sinais , Células THP-1 , Tromboplastina/genética , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
4.
Circ Res ; 123(1): 33-42, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29572206

RESUMO

RATIONALE: Neutrophils likely contribute to the thrombotic complications of human atheromata. In particular, neutrophil extracellular traps (NETs) could exacerbate local inflammation and amplify and propagate arterial intimal injury and thrombosis. PAD4 (peptidyl arginine deiminase 4) participates in NET formation, but an understanding of this enzyme's role in atherothrombosis remains scant. OBJECTIVE: This study tested the hypothesis that PAD4 and NETs influence experimental atherogenesis and in processes implicated in superficial erosion, a form of plaque complication we previously associated with NETs. METHODS AND RESULTS: Bone marrow chimeric Ldlr deficient mice reconstituted with either wild-type or PAD4-deficient cells underwent studies that assessed atheroma formation or procedures designed to probe mechanisms related to superficial erosion. PAD4 deficiency neither retarded fatty streak formation nor reduced plaque size or inflammation in bone marrow chimeric mice that consumed an atherogenic diet. In contrast, either a PAD4 deficiency in bone marrow-derived cells or administration of DNaseI to disrupt NETs decreased the extent of arterial intimal injury in mice with arterial lesions tailored to recapitulate characteristics of human atheroma complicated by erosion. CONCLUSIONS: These results indicate that PAD4 from bone marrow-derived cells and NETs do not influence chronic experimental atherogenesis, but participate causally in acute thrombotic complications of intimal lesions that recapitulate features of superficial erosion.


Assuntos
Armadilhas Extracelulares/fisiologia , Hidrolases/fisiologia , Placa Aterosclerótica/etiologia , Trombose/etiologia , Animais , Transplante de Medula Óssea , Doenças das Artérias Carótidas/etiologia , Doenças das Artérias Carótidas/patologia , Morte Celular , Desoxirribonuclease I/farmacologia , Armadilhas Extracelulares/efeitos dos fármacos , Humanos , Hidrolases/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/fisiologia , Osteomielite/etiologia , Placa Aterosclerótica/patologia , Proteína-Arginina Desiminase do Tipo 4 , Trombose/prevenção & controle , Túnica Íntima/lesões
5.
PLoS One ; 11(3): e0152474, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27032103

RESUMO

AIMS: Mice deficient in IL-1 receptor 1 (hence unresponsive to both IL-1 isoforms α and ß) have impaired expansive arterial remodeling due to diminished expression of matrix-degrading enzymes, especially MMP-3. Emergence of IL-1 as a target in cardiovascular disease prompted the investigation of the redundancy of IL-1α and IL-1ß in the induction of MMP-3 and other matrix-remodeling enzymes in human cells. METHODS AND RESULTS: Human primary vascular smooth muscle cells (VSMCs) and carotid endarterectomy specimens were stimulated with equimolar concentrations of IL-1α or IL-1ß and analyzed protease expression by immunoblot and ELISA. Either IL-1α or IL-1ß increased the expression of pro-MMP-3 in VSMCs, facilitated VSMC migration through Matrigel, and induced MMP-3 production in specimens from atheromatous plaques. VSMCs also secreted MMP-1 and Cathepsin S (CatS) upon stimulation with IL-1α or IL-1ß. IL-1 isoforms similarly increased MMP-1 and MMP-9 expression in carotid endarterectomy specimens. We examined the expression of MMP-3 and IL-1 isoforms by immunostaining of carotid atheromata, calculated the % positive areas, and tested associations by linear regression. MMP-3 colocalized with IL-1 isoforms in atheromata. MMP-3+ area in plaques positively associated with IL-1α+ (R2 = 0.61, P<0.001) and with IL-1ß + areas (R2 = 0.68, P<0.001). MMP-3+ area within atheroma also associated with CD68+ area, but not with α-smooth muscle actin area. CONCLUSIONS: Either IL-1α or IL-1ß can induce the expression of enzymes implicated in remodeling of the arterial extracellular matrix, and facilitate human VSMC migration in vitro. Human atheromata contain both IL-1 isoforms in association with immunoreactive MMP-3. This redundancy of IL-1 isoforms suggests that selective blocking of one IL-1 isoform should not impair expansive arterial remodeling, a finding with important clinical implications for therapeutic targeting of IL-1 in atherosclerosis.


Assuntos
Interleucina-1/análise , Transdução de Sinais , Animais , Artérias/metabolismo , Catepsinas/genética , Catepsinas/metabolismo , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Immunoblotting , Imuno-Histoquímica , Interleucina-1/farmacologia , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/análise , Metaloproteinase 3 da Matriz/genética , Metaloproteinase 3 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Isoformas de Proteínas/análise , Isoformas de Proteínas/farmacologia , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Circ Res ; 115(10): 875-83, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25185259

RESUMO

RATIONALE: Inflammation drives atherogenesis. Animal and human studies have implicated interleukin-1ß (IL-1ß) in this disease. Moderate hypoxia, a condition that prevails in the atherosclerotic plaque, may conspire with inflammation and contribute to the evolution and complications of atherosclerosis through mechanisms that remain incompletely understood. OBJECTIVE: This study investigated the links between hypoxia and inflammation by testing the hypothesis that moderate hypoxia modulates IL-1ß production in activated human macrophages. METHODS AND RESULTS: Our results demonstrated that hypoxia enhances pro-IL-1ß protein, but not mRNA, expression in lipopolysaccharide-stimulated human macrophages. We show that hypoxia limits the selective targeting of pro-IL-1ß to autophagic degradation, thus prolonging its half-life and promoting its intracellular accumulation. Furthermore, hypoxia increased the expression of NLRP3, a limiting factor in NLRP3 inflammasome function, and augmented caspase-1 activation in lipopolysaccharide-primed macrophages. Consequently, hypoxic human macrophages secreted higher amounts of mature IL-1ß than did normoxic macrophages after treatment with crystalline cholesterol, an endogenous danger signal that contributes to atherogenesis. In human atherosclerotic plaques, IL-1ß localizes predominantly to macrophage-rich regions that express activated caspase-1 and the hypoxia markers hypoxia-inducible factor 1α and hexokinase-2, as assessed by immunohistochemical staining of carotid endarterectomy specimens. CONCLUSIONS: These results indicate that hypoxia potentiates IL-1ß expression in cultured human macrophages and in the context of atheromata, therefore unveiling a novel proinflammatory mechanism that may participate in atherogenesis.


Assuntos
Interleucina-1beta/biossíntese , Macrófagos/metabolismo , Hipóxia Celular/fisiologia , Células Cultivadas , Humanos , Leucócitos Mononucleares/metabolismo
7.
Circ Cardiovasc Imaging ; 7(6): 920-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25170063

RESUMO

BACKGROUND: Studies have shown the feasibility of imaging plaques with 2-deoxy-2-[(18)F]fluoroglucose (FDG) positron emission tomography and dynamic contrast-enhanced magnetic resonance imaging with inconsistent results. We sought to investigate the relationship between markers of inflammatory activation, plaque microvascularization, and vessel wall permeability in subjects with carotid plaques using a multimodality approach combining FDG positron emission tomography, dynamic contrast-enhanced magnetic resonance imaging, and histopathology. METHODS AND RESULTS: Thirty-two subjects with carotid stenoses underwent noninvasive imaging with FDG positron emission tomography and dynamic contrast-enhanced magnetic resonance imaging, 46.9% (n=15) before carotid endarterectomy. We measured FDG uptake (target:background ratio [TBR]) by positron emission tomography and K(trans) (reflecting microvascular permeability and perfusion) by magnetic resonance imaging and correlated imaging with immunohistochemical markers of macrophage content (CD68), activated inflammatory cells (major histocompatibility complex class II), and microvessels (CD31) in plaque and control regions. TBR and K(trans) correlated significantly with tertiles of CD68(+) (P=0.009 and P=0.008, respectively), major histocompatibility complex class II(+) (P=0.003 and P<0.001, respectively), and CD31(+) (P=0.004 and P=0.008, respectively). Regions of plaques were associated with increased CD68(+) (P=0.002), major histocompatibility complex class II(+) (P=0.002), CD31(+) (P=0.02), TBR (P<0.0001), and K(trans) (P<0.0001), as compared with those without plaques. Microvascularization correlated with macrophage content (rs=0.52; P=0.007) and inflammatory activity (rs=0.68; P=0.0001) and TBR correlated with K(trans) (rs=0.53; P<0.0001). In multivariable mixed linear regression modeling, TBR remained independently associated with K(trans) (ß[SE], 2.68[0.47]; P<0.0001). CONCLUSIONS: Plaque regions with active inflammation, as determined by macrophage content and major histocompatibility complex class II expression, showed increased FDG uptake, which correlated with increased K(trans) and microvascularization. The correlation between K(trans) and TBR was moderate, direct, highly significant, and independent of clinical symptoms and plaque luminal severity.


Assuntos
Permeabilidade Capilar , Artérias Carótidas , Estenose das Carótidas/diagnóstico , Inflamação/diagnóstico , Imageamento por Ressonância Magnética , Microvasos/patologia , Neovascularização Patológica , Tomografia por Emissão de Pósitrons , Tomografia Computadorizada por Raios X , Idoso , Biomarcadores/metabolismo , Artérias Carótidas/diagnóstico por imagem , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Estenose das Carótidas/diagnóstico por imagem , Estenose das Carótidas/metabolismo , Estenose das Carótidas/patologia , Meios de Contraste , Feminino , Fluordesoxiglucose F18 , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Inflamação/diagnóstico por imagem , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Modelos Lineares , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Imagem Multimodal , Análise Multivariada , Placa Aterosclerótica , Valor Preditivo dos Testes , Prognóstico , Estudos Prospectivos , Compostos Radiofarmacêuticos , Índice de Gravidade de Doença
8.
Arterioscler Thromb Vasc Biol ; 34(7): 1374-81, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24812325

RESUMO

OBJECTIVE: Obesity associates with increased numbers of inflammatory cells in adipose tissue (AT), including T cells, but the mechanism of T-cell recruitment remains unknown. This study tested the hypothesis that the chemokine (C-X-C motif) receptor 3 (CXCR3) participates in T-cell accumulation in AT of obese mice and thus in the regulation of local inflammation and systemic metabolism. APPROACH AND RESULTS: Obese wild-type mice exhibited higher mRNA expression of CXCR3 in periepididymal AT-derived stromal vascular cells compared with lean mice. We evaluated the function of CXCR3 in AT inflammation in vivo using CXCR3-deficient and wild-type control mice that consumed a high-fat diet. Periepididymal AT from obese CXCR3-deficient mice contained fewer T cells than obese controls after 8 and 16 weeks on high-fat diet, as assessed by flow cytometry. Obese CXCR3-deficient mice had greater glucose tolerance than obese controls after 8 weeks, but not after 16 weeks. CXCR3-deficient mice fed high-fat diet had reduced mRNA expression of proinflammatory mediators, such as monocyte chemoattractant protein-1 and regulated on activation, normal T cell expressed and secreted, and anti-inflammatory genes, such as Foxp3, IL-10, and arginase-1 in periepididymal AT, compared with obese controls. CONCLUSIONS: These results demonstrate that CXCR3 contributes to T-cell accumulation in periepididymal AT of obese mice. Our results also suggest that CXCR3 regulates the accumulation of distinct subsets of T cells and that the ratio between these functional subsets across time likely modulates local inflammation and systemic metabolism.


Assuntos
Tecido Adiposo/imunologia , Quimiotaxia de Leucócito , Obesidade/imunologia , Paniculite/imunologia , Receptores CXCR3/metabolismo , Subpopulações de Linfócitos T/imunologia , Tecido Adiposo/metabolismo , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/genética , Obesidade/metabolismo , Paniculite/genética , Paniculite/metabolismo , Receptores CXCR3/deficiência , Receptores CXCR3/genética , Transdução de Sinais , Subpopulações de Linfócitos T/metabolismo , Fatores de Tempo
9.
J Biol Chem ; 287(44): 36896-904, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22948153

RESUMO

Abundant experimental and clinical data support a modulatory role for adiponectin in inflammation, dysmetabolism, and disease. Because the activation of cells involved in innate and adaptive immunity contributes to the pathogenesis of diseases such as atherosclerosis and obesity, this study investigated the role of adiponectin in human macrophage polarization and T cell differentiation. Examination of the adiponectin-induced transcriptome in primary human macrophages revealed that adiponectin promotes neither classical (M1) nor alternative (M2) macrophage activation but elicits a pro-inflammatory response that resembles M1 more closely than M2. Addition of adiponectin to polyclonally activated CD4(+) T lymphocytes did not affect cell proliferation but induced mRNA expression and protein secretion of interferon (IFN)-γ and interleukin (IL)-6. Adiponectin treatment of CD4(+) T cells increased phosphorylation of p38 mitogen-activated protein kinase (MAPK) and signal transducer and activation of transcription (STAT) 4 and augmented T-bet expression. Inhibition of p38 with SB203580 abrogated adiponectin-induced IFN-γ production, indicating that adiponectin enhances T(H)1 differentiation through the activation of the p38-STAT4-T-bet axis. Collectively, our results demonstrate that adiponectin can induce pro-inflammatory functions in isolated macrophages and T cells, concurring with previous observations that adiponectin induces a limited program of inflammatory activation that likely desensitizes these cells to further pro-inflammatory stimuli.


Assuntos
Adiponectina/fisiologia , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Células Th1/fisiologia , Imunidade Adaptativa , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/fisiologia , Diferenciação Celular , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Meios de Cultivo Condicionados , Expressão Gênica , Cadeias alfa de HLA-DR/genética , Cadeias alfa de HLA-DR/metabolismo , Humanos , Imunidade Inata , Macrófagos/metabolismo , Fosforilação , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/patologia , Processamento de Proteína Pós-Traducional , Fator de Transcrição STAT4/metabolismo , Transdução de Sinais , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Circulation ; 124(25): 2920-32, 2011 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-22144572

RESUMO

BACKGROUND: The calcium-binding proteins myeloid-related protein (MRP)-8 (S100A8) and MRP-14 (S100A9) form MRP-8/14 heterodimers (S100A8/A9, calprotectin) that regulate myeloid cell function and inflammatory responses and serve as early serum markers for monitoring acute allograft rejection. Despite functioning as a proinflammatory mediator, the pathophysiological role of MRP-8/14 complexes in cardiovascular disease is incompletely defined. This study investigated the role of MRP-8/14 in cardiac allograft rejection using MRP-14(-/-) mice that lack MRP-8/14 complexes. METHODS AND RESULTS: We examined parenchymal rejection after major histocompatibility complex class II allomismatched cardiac transplantation (bm12 donor heart and B6 recipients) in wild-type (WT) and MRP-14(-/-) recipients. Allograft survival averaged 5.9±2.9 weeks (n=10) in MRP-14(-/-) recipients compared with >12 weeks (n=15; P<0.0001) in WT recipients. Two weeks after transplantation, allografts in MRP-14(-/-) recipients had significantly higher parenchymal rejection scores (2.8±0.8; n=8) than did WT recipients (0.8±0.8; n=12; P<0.0001). Compared with WT recipients, allografts in MRP-14(-/-) recipients had significantly increased T-cell and macrophage infiltration and increased mRNA levels of interferon-γ and interferon-γ-associated chemokines (CXCL9, CXCL10, and CXCL11), interleukin-6, and interleukin-17 with significantly higher levels of Th17 cells. MRP-14(-/-) recipients also had significantly more lymphocytes in the adjacent para-aortic lymph nodes than did WT recipients (cells per lymph node: 23.7±0.7×10(5) for MRP-14(-/-) versus 6.0±0.2×10(5) for WT; P<0.0001). The dendritic cells (DCs) of the MRP-14(-/-) recipients of bm12 hearts expressed significantly higher levels of the costimulatory molecules CD80 and CD86 than did those of WT recipients 2 weeks after transplantation. Mixed leukocyte reactions with allo-endothelial cell-primed MRP-14(-/-) DCs resulted in significantly higher antigen-presenting function than reactions using WT DCs. Ovalbumin-primed MRP-14(-/-) DCs augmented proliferation of OT-II (ovalbumin-specific T cell receptor transgenic) CD4(+) T cells with increased interleukin-2 and interferon-γ production. Cardiac allografts of B6 major histocompatibility complex class II(-/-) hosts and of B6 WT hosts receiving MRP-14(-/-) DCs had significantly augmented inflammatory cell infiltration and accelerated allograft rejection compared with WT DCs from transferred recipient allografts. Bone marrow-derived MRP-14(-/-) DCs infected with MRP-8 and MRP-14 retroviral vectors showed significantly decreased CD80 and CD86 expression compared with controls, indicating that MRP-8/14 regulates B7-costimulatory molecule expression. CONCLUSIONS: Our results indicate that MRP-14 regulates B7 molecule expression and reduces antigen presentation by DCs and subsequent T-cell priming. The absence of MRP-14 markedly increased T-cell activation and exacerbated allograft rejection, indicating a previously unrecognized role for MRP-14 in immune cell biology.


Assuntos
Calgranulina A/imunologia , Calgranulina B/imunologia , Rejeição de Enxerto/metabolismo , Transplante de Coração/imunologia , Animais , Antígenos B7/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Calgranulina A/genética , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Citocinas/genética , Citocinas/metabolismo , Sobrevivência de Enxerto/imunologia , Histocompatibilidade/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Complexo Antígeno L1 Leucocitário/genética , Complexo Antígeno L1 Leucocitário/imunologia , Complexo Antígeno L1 Leucocitário/metabolismo , Teste de Cultura Mista de Linfócitos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores do Ácido Retinoico/imunologia , Receptores do Ácido Retinoico/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Transplante Homólogo , Receptor gama de Ácido Retinoico
11.
Int J Inflam ; 2011: 529061, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21837268

RESUMO

Obesity, long considered a condition characterized by the deposition of inert fat, is now recognized as a chronic and systemic inflammatory disease, where adipose tissue plays a crucial endocrine role through the production of numerous bioactive molecules, collectively known as adipokines. These molecules regulate carbohydrate and lipid metabolism, immune function and blood coagulability, and may serve as blood markers of cardiometabolic risk. Local inflammatory loops operate in adipose tissue as a consequence of nutrient overload, and crosstalk among its cellular constituents-adipocytes, endothelial and immune cells-results in the elaboration of inflammatory mediators. These mediators promote important systemic effects that can result in insulin resistance, dysmetabolism and cardiovascular disease. The understanding that inflammation plays a critical role in the pathogenesis of obesity-derived disorders has led to therapeutic approaches that target different points of the inflammatory network induced by obesity.

12.
J Am Coll Cardiol ; 58(6): 603-14, 2011 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-21798423

RESUMO

OBJECTIVES: This study investigated the regulation of glucose uptake in cells that participate in atherogenesis by stimuli relevant to this process, to gain mechanistic insight into the origin of the (18)fluorine-labeled 2-deoxy-D-glucose (FdG) uptake signals observed clinically. BACKGROUND: Patient studies suggest that positron emission tomography (PET) using FdG can detect "active" atherosclerotic plaques, yet the mechanism giving rise to FdG signals remains unknown. METHODS: We exposed cells to conditions thought to operate in atheroma and determined rates of glucose uptake. RESULTS: Hypoxia, but not pro-inflammatory cytokines, potently stimulated glucose uptake in human macrophages and foam cells. Statins attenuated this process in vitro, suggesting that these agents have a direct effect on human macrophages. Immunohistochemical study of human plaques revealed abundant expression of proteins regulating glucose utilization, predominantly in macrophage-rich regions of the plaques-regions previously proved hypoxic. Smooth-muscle cells and endothelial cells markedly increased rates of glucose uptake when exposed to pro-inflammatory cytokines. CONCLUSIONS: Glucose uptake and, probably, FdG uptake signals in atheroma may reflect hypoxia-stimulated macrophages rather than mere inflammatory burden. Cytokine-activated smooth-muscle cells also may contribute to the FdG signal.


Assuntos
Aterosclerose/diagnóstico por imagem , Aterosclerose/terapia , Glucose/farmacocinética , Hipóxia , Inflamação , Macrófagos/efeitos dos fármacos , Aorta/patologia , Aterosclerose/patologia , Citocinas/metabolismo , Células Endoteliais/citologia , Fluordesoxiglucose F18/farmacologia , Células Espumosas/metabolismo , Glucose/metabolismo , Humanos , Hidrólise , Macrófagos/metabolismo , Monócitos/citologia , Miócitos de Músculo Liso/metabolismo , Tomografia por Emissão de Pósitrons/métodos
13.
Sci Transl Med ; 3(84): 84ra45, 2011 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-21613624

RESUMO

New high-resolution molecular and structural imaging strategies are needed to visualize high-risk plaques that are likely to cause acute myocardial infarction, because current diagnostic methods do not reliably identify at-risk subjects. Although molecular imaging agents are available for low-resolution detection of atherosclerosis in large arteries, a lack of imaging agents coupled to high-resolution modalities has limited molecular imaging of atherosclerosis in the smaller coronary arteries. Here, we have demonstrated that indocyanine green (ICG), a Food and Drug Administration-approved near-infrared fluorescence (NIRF)-emitting compound, targets atheromas within 20 min of injection and provides sufficient signal enhancement for in vivo detection of lipid-rich, inflamed, coronary-sized plaques in atherosclerotic rabbits. In vivo NIRF sensing was achieved with an intravascular wire in the aorta, a vessel of comparable caliber to human coronary arteries. Ex vivo fluorescence reflectance imaging showed high plaque target-to-background ratios in atheroma-bearing rabbits injected with ICG compared to atheroma-bearing rabbits injected with saline. In vitro studies using human macrophages established that ICG preferentially targets lipid-loaded macrophages. In an early clinical study of human atheroma specimens from four patients, we found that ICG colocalized with plaque macrophages and lipids. The atheroma-targeting capability of ICG has the potential to accelerate the clinical development of NIRF molecular imaging of high-risk plaques in humans.


Assuntos
Artérias/patologia , Diagnóstico por Imagem/métodos , Verde de Indocianina , Inflamação/patologia , Lipídeos/química , Placa Aterosclerótica/diagnóstico , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Acetilação , Animais , Artérias/fisiopatologia , Permeabilidade Capilar/fisiologia , Bovinos , Endocitose , Fluorescência , Humanos , Inflamação/complicações , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Placa Aterosclerótica/complicações , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/fisiopatologia , Coelhos , Radiografia , Soroalbumina Bovina/metabolismo
14.
J Biol Chem ; 284(38): 25569-75, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19617629

RESUMO

Macrophages participate pivotally in the pathogenesis of many chronic inflammatory diseases including atherosclerosis. Adiponectin, a vasculoprotective molecule with insulin-sensitizing and anti-atherogenic properties, suppresses pro-inflammatory gene expression in macrophages by mechanisms that remain incompletely understood. This study investigated the effects of adiponectin on major pro-inflammatory signaling pathways in human macrophages. We demonstrate that pretreatment of these cells with adiponectin inhibits phosphorylation of nuclear factor kappaB inhibitor (IkappaB), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (MAPK), induced by either lipopolysaccharide (LPS) or tumor necrosis factor (TNF) alpha, as well as STAT3 phosphorylation induced by interleukin-6 (IL6). Antagonism of IL10 by either neutralizing antibodies or siRNA-mediated silencing did not abrogate the anti-inflammatory actions of adiponectin, indicating that the ability of adiponectin to render human macrophages tolerant to various pro-inflammatory stimuli does not require this cytokine. A systematic search for adiponectin-inducible genes with established anti-inflammatory properties revealed that adiponectin augmented the expression of A20, suppressor of cytokine signaling (SOCS) 3, B-cell CLL/lymphoma (BCL) 3, TNF receptor-associated factor (TRAF) 1, and TNFAIP3-interacting protein (TNIP) 3. These results suggest that adiponectin triggers a multifaceted response in human macrophages by inducing the expression of various anti-inflammatory proteins that act at different levels in concert to suppress macrophage activation.


Assuntos
Interleucina-10/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/metabolismo , Adiponectina/metabolismo , Adiponectina/farmacologia , Proteína 3 do Linfoma de Células B , Linhagem Celular , Proteínas de Ligação a DNA , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas I-kappa B/metabolismo , Inflamação/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Interleucina-6/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipopolissacarídeos/farmacologia , Proteínas Nucleares/biossíntese , Fosforilação/efeitos dos fármacos , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Fator de Transcrição STAT3/biossíntese , Fator 1 Associado a Receptor de TNF/biossíntese , Fatores de Transcrição/biossíntese , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Am J Pathol ; 174(2): 693-700, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19131590

RESUMO

Rupture of the collagenous, fibrous cap of an atherosclerotic plaque commonly causes thrombosis. Activated immune cells can secrete mediators that jeopardize the integrity of the fibrous cap. This study aimed to determine the relationship between T-cell-mediated inflammation and collagen turnover in a mouse model of experimental atherosclerosis. Both Apoe(-/-) x CD4dnTbetaRII mice with defective transforming growth factor-beta receptors in T cells (and hence released from tonic suppression of T-cell activation) and lesion size-matched Apoe(-/-) mice were used. Picrosirius red staining showed a lower content of thick mature collagen fibers in lesions of Apoe(-/-) x CD4dnTbetaRII mice, although both groups had similar levels of procollagen type I or III mRNA and total collagen content in lesions. Analysis of both gene expression and protein content showed a significant decrease of lysyl oxidase, the extracellular enzyme needed for collagen cross-linking, in aortas of Apoe(-/-)--CD4dnTbetaRII mice. T-cell-driven inflammation provoked a selective and limited increase in the expression of proteinases that catabolize the extracellular matrix. Atheromata of Apoe(-/-)--CD4dnTbetaRII mice had increased levels of matrix metalloproteinase-13 and cathepsin S mRNAs and of the active form of cathepsin S protein but no increase was detected in collagen fragmentation. Our results suggest that exaggerated T-cell-driven inflammation limits collagen maturation in the atherosclerotic plaque while having little effect on collagen degradation.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/imunologia , Colágeno/metabolismo , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Animais , Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/patologia , Western Blotting , Colágeno/genética , Colágeno/imunologia , Imunofluorescência , Expressão Gênica , Imuno-Histoquímica , Inflamação/imunologia , Inflamação/patologia , Camundongos , Camundongos Knockout , Proteína-Lisina 6-Oxidase/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Circ Res ; 103(5): 467-76, 2008 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-18658050

RESUMO

Adipose tissue (AT) can accumulate macrophages and secrete several inflammatory mediators. Despite its pivotal role in the progression of chronic inflammatory processes such as atherosclerosis, the adaptive role of immunity in obesity remains poorly explored. Visceral AT of diet-induced obese C57BL/6 mice had higher numbers of both CD4(+) and CD8(+) T cells than lean controls, monitored by flow cytometry. When stimulated in vitro, T cells from obese AT produced more interferon (IFN)gamma than those from controls. AT from obese animals also had more cells expressing I-A(b), a mouse class II histocompatibility marker implicated in antigen presentation, as determined by immunostaining. Differentiated 3T3-L1 cells stimulated with recombinant IFNgamma or T-helper 1-derived supernatant produced several chemokines and their mRNAs. Obese IFNgamma-deficient animals had significantly reduced AT expression of mRNA-encoding inflammatory genes such as tumor necrosis factor-alpha and monocyte chemoattractant protein-1, decreased AT inflammatory cell accumulation, and better glucose tolerance than control animals consuming the same diet. Obese mice doubly deficient for IFNgamma receptor and apolipoprotein (Apo)E on a mixed 129SvEv/C57BL/6 (129/B6) genetic background, despite exhibiting similar AT mRNA levels of tumor necrosis factor-alpha and monocyte chemoattractant protein-1 as 129/B6-ApoE(-/-) controls, had decreased expression of important T cell-related genes, such as IFNgamma-inducible protein-10 and I-A(b), and lower plasma triglycerides and glucose. These results indicate a role for T cells and IFNgamma, a prototypical T-helper 1 cytokine, in regulation of the inflammatory response that accompanies obesity.


Assuntos
Tecido Adiposo Branco/imunologia , Inflamação/imunologia , Interferon gama/metabolismo , Obesidade/imunologia , Células Th1/imunologia , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/imunologia , Adipócitos/metabolismo , Tecido Adiposo Branco/citologia , Tecido Adiposo Branco/metabolismo , Ração Animal , Animais , Apolipoproteínas E/genética , Glicemia/metabolismo , Colesterol/sangue , Expressão Gênica/imunologia , Inflamação/metabolismo , Resistência à Insulina , Interferon gama/genética , Interferon gama/imunologia , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/metabolismo , Leptina/sangue , Contagem de Leucócitos , Macrófagos/citologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Obesidade/metabolismo , Técnicas de Cultura de Órgãos , Receptores de Interferon/genética , Células Th1/citologia , Células Th1/metabolismo , Receptor de Interferon gama
17.
J Clin Invest ; 118(6): 2246-59, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18464931

RESUMO

Long QT syndrome (LQTS) is a heritable disease associated with ECG QT interval prolongation, ventricular tachycardia, and sudden cardiac death in young patients. Among genotyped individuals, mutations in genes encoding repolarizing K+ channels (LQT1:KCNQ1; LQT2:KCNH2) are present in approximately 90% of affected individuals. Expression of pore mutants of the human genes KCNQ1 (KvLQT1-Y315S) and KCNH2 (HERG-G628S) in the rabbit heart produced transgenic rabbits with a long QT phenotype. Prolongations of QT intervals and action potential durations were due to the elimination of IKs and IKr currents in cardiomyocytes. LQT2 rabbits showed a high incidence of spontaneous sudden cardiac death (>50% at 1 year) due to polymorphic ventricular tachycardia. Optical mapping revealed increased spatial dispersion of repolarization underlying the arrhythmias. Both transgenes caused downregulation of the remaining complementary IKr and IKs without affecting the steady state levels of the native polypeptides. Thus, the elimination of 1 repolarizing current was associated with downregulation of the reciprocal repolarizing current rather than with the compensatory upregulation observed previously in LQTS mouse models. This suggests that mutant KvLQT1 and HERG interacted with the reciprocal wild-type alpha subunits of rabbit ERG and KvLQT1, respectively. These results have implications for understanding the nature and heterogeneity of cardiac arrhythmias and sudden cardiac death.


Assuntos
Canal de Potássio KCNQ1/genética , Síndrome do QT Longo/genética , Síndrome do QT Longo/patologia , Potenciais de Ação , Animais , Animais Geneticamente Modificados , Morte Súbita , Modelos Animais de Doenças , Canal de Potássio ERG1 , Ecocardiografia , Eletrofisiologia/métodos , Canais de Potássio Éter-A-Go-Go , Genótipo , Ventrículos do Coração/patologia , Células Musculares/patologia , Fenótipo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Coelhos
18.
Am J Physiol Heart Circ Physiol ; 294(5): H2012-21, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18344374

RESUMO

The physiological function of ion channels is affected by protein-protein and protein-membrane interactions that modulate their activity and/or localization. Palmitoylation modulates protein function by facilitating targeted membrane association, interaction with other proteins, and determining subcellular localization. In this study, we demonstrate that the voltage-dependent potassium (Kv) channel Kv1.5 is palmitoylated and that the mutation of COOH-terminal cysteines is sufficient to abolish the palmitoylation of the Kv1.5 polypeptide in Chinese hamster ovary (CHO) cells. The labeling represented the thioester linkage of the labeled palmitic acid to cysteine rather than amide and oxygen ester linkages as judged by the release of the palmitic acid upon the treatment of the gel with hydroxylamine at a neutral pH. Site-directed mutagenesis and radiolabeling studies revealed that C593 was the sole site of palmitoylation. The elucidation of the biological function of palmitoylation revealed that the expression of the FLAG-Kv1.5 palmitoylation-deficient mutant (FL-Kv1.5(Palm-)) in stable CHO cells increased membrane expression as determined by the biotinylation of surface proteins and quantitative immunofluorescence analyses of these cells, in turn enhancing the outward potassium current. This enhanced surface expression and the currents were consequential to the slower rate of internalization, causing an increased localization of FL-Kv1.5(Palm-) in the plasma membrane compared with the wild-type FL-Kv1.5 channels. We conclude that the Kv1.5 channel is palmitoylated and that its palmitoylation modulates its biological functions and, therefore, might provide a physiological link between the metabolic state and the expression of Kv1.5 on the plasma membrane.


Assuntos
Canal de Potássio Kv1.5/metabolismo , Potássio/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Biotinilação , Células CHO , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Cisteína/metabolismo , Canal de Potássio Kv1.5/química , Canal de Potássio Kv1.5/genética , Lipoilação , Potenciais da Membrana , Microscopia de Fluorescência , Mutagênese Sítio-Dirigida , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Projetos de Pesquisa , Fatores de Tempo , Transfecção
19.
Circ Res ; 102(2): 218-25, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17991878

RESUMO

Obese individuals often have low plasma adiponectin and concomitant chronic inflammation with a predisposition to metabolic and cardiovascular diseases. The present study reports a novel antiinflammatory action of adiponectin in human monocyte-derived macrophages (MPhi) suppressing T-lymphocyte accumulation in atherogenesis. RNA profiling of lipopolysaccharide-stimulated human MPhi identified CXC chemokine ligands (CXCLs), such as IP-10 (interferon [IFN]-inducible protein 10) (CXCL10), I-TAC (IFN-inducible T-cell alpha chemoattractant) (CXCL11), and Mig (monokine induced by IFN-gamma) (CXCL9), T-lymphocyte chemoattractants associated with atherogenesis, among the top 14 transcripts suppressed by adiponectin. Real-time quantitative RT-PCR and ELISA verified that adiponectin inhibited expression of these chemokines at both the mRNA and protein levels in a concentration-dependent manner. Adiponectin reduced the release by lipopolysaccharide-stimulated MPhi of chemoattractant activity for CXC chemokine receptor 3-transfected (receptor for IP-10, Mig, and I-TAC) lymphocytes. Adiponectin decreased lipopolysaccharide-inducible IP-10 promoter activity in promoter-transfected THP-1 MPhi but did not change IP-10 mRNA stability. In lipopolysaccharide-stimulated MPhi, reduction of IFN-beta by adiponectin preceded inhibition of IP-10 mRNA expression. Immunoblot and chromatin immunoprecipitation analyses demonstrated that adiponectin attenuated activation of the transcription factor IFN regulatory factor 3, involved in the MyD88-independent pathway of Toll-like receptor 4 signaling, and subsequent IFN regulatory factor 3 binding to IFN-beta promoter. In vivo studies further demonstrated that apolipoprotein E/adiponectin double-deficient (apoE-/-APN-/-) mice had increased plasma IP-10 levels, accelerated T-lymphocyte accumulation in atheromata, and augmented atherogenesis compared with apoE single-deficient (apoE-/-APN+/+) mice. This study establishes that low levels of adiponectin associated with obesity, the metabolic syndrome, and diabetes favor T-lymphocyte recruitment and contribute to adaptive immune response during atherogenesis.


Assuntos
Adiponectina/farmacologia , Arteriosclerose/imunologia , Quimiocinas CXC/antagonistas & inibidores , Quimiotaxia de Leucócito/efeitos dos fármacos , Receptores CXCR3/metabolismo , Linfócitos T/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus , Relação Dose-Resposta a Droga , Humanos , Imunidade/efeitos dos fármacos , Macrófagos , Síndrome Metabólica , Obesidade , Receptores CXCR3/genética , Transfecção
20.
Atherosclerosis ; 192(2): 298-304, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17005185

RESUMO

BACKGROUND: Previous reports have suggested that advanced glycation end products (AGE) participate in the pathogenesis of diabetic macroangiopathy. However, current understanding of the mechanisms by which AGE may accelerate atherogenesis remains incomplete. METHODS AND RESULTS: Microarray and reverse transcription real-time PCR analyses revealed that exposure to AGE-BSA (BSA, bovine serum albumin) reduced mRNA levels (60%) in the ATP-binding cassette transporter G1 (ABCG1) but not ABCA1 in human macrophages. AGE-BSA also reduced ABCG1 protein levels. These effects occurred mainly through the receptor for AGE (RAGE), as an anti-RAGE antibody significantly limited ABCG1 mRNA reduction. Functional studies demonstrated that exposure to AGE-BSA decreased cholesterol efflux to high-density lipoprotein (HDL) (P<0.05) but not to apolipoprotein AI, compared to BSA treatment. Although liver X receptors (LXR) augment ABCG1 expression, macrophages treated with AGE-BSA showed no reduction in LXR mRNA levels or in the binding of nuclear proteins to the LXR response element, compared with BSA. CONCLUSIONS: Our data show that AGE-BSA can decrease cholesterol efflux from macrophages to HDL via an LXR-independent pathway. This novel mechanism may contribute to accelerated foam cell production and atherogenesis in diabetic patients.


Assuntos
Transportadores de Cassetes de Ligação de ATP/biossíntese , Colesterol/metabolismo , Produtos Finais de Glicação Avançada/farmacologia , Lipoproteínas HDL/metabolismo , Macrófagos/fisiologia , Soroalbumina Bovina/farmacologia , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Proteínas de Ligação a DNA/metabolismo , Humanos , Receptores X do Fígado , Macrófagos/efeitos dos fármacos , Receptores Nucleares Órfãos , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...