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.
J Lipid Res ; 61(12): 1577-1588, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32907987

RESUMO

Lipoproteins play a key role in transport of cholesterol to and from tissues. Recent studies have also demonstrated that red blood cells (RBCs), which carry large quantities of free cholesterol in their membrane, play an important role in reverse cholesterol transport. However, the exact role of RBCs in systemic cholesterol metabolism is poorly understood. RBCs were incubated with autologous plasma or isolated lipoproteins resulting in a significant net amount of cholesterol moved from RBCs to HDL, while cholesterol from LDL moved in the opposite direction. Furthermore, the bi-directional cholesterol transport between RBCs and plasma lipoproteins was saturable and temperature-, energy-, and time-dependent, consistent with an active process. We did not find LDLR, ABCG1, or scavenger receptor class B type 1 in RBCs but found a substantial amount of ABCA1 mRNA and protein. However, specific cholesterol efflux from RBCs to isolated apoA-I was negligible, and ABCA1 silencing with siRNA or inhibition with vanadate and Probucol did not inhibit the efflux to apoA-I, HDL, or plasma. Cholesterol efflux from and cholesterol uptake by RBCs from Abca1+/+ and Abca1-/- mice were similar, arguing against the role of ABCA1 in cholesterol flux between RBCs and lipoproteins. Bioinformatics analysis identified ABCA7, ABCG5, lipoprotein lipase, and mitochondrial translocator protein as possible candidates that may mediate the cholesterol flux. Together, these results suggest that RBCs actively participate in cholesterol transport in the blood, but the role of cholesterol transporters in RBCs remains uncertain.


Assuntos
Colesterol/metabolismo , Eritrócitos/metabolismo , Lipoproteínas/metabolismo , Transporte Biológico , Biologia Computacional , Humanos
2.
Arterioscler Thromb Vasc Biol ; 40(10): 2346-2359, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32787522

RESUMO

OBJECTIVE: AIBP (apolipoprotein A-I binding protein) is an effective and selective regulator of lipid rafts modulating many metabolic pathways originating from the rafts, including inflammation. The mechanism of action was suggested to involve stimulation by AIBP of cholesterol efflux, depleting rafts of cholesterol, which is essential for lipid raft integrity. Here we describe a different mechanism contributing to the regulation of lipid rafts by AIBP. Approach and Results: We demonstrate that modulation of rafts by AIBP may not exclusively depend on the rate of cholesterol efflux or presence of the key regulator of the efflux, ABCA1 (ATP-binding cassette transporter A-I). AIBP interacted with phosphatidylinositol 3-phosphate, which was associated with increased abundance and activation of Cdc42 and rearrangement of the actin cytoskeleton. Cytoskeleton rearrangement was accompanied with reduction of the abundance of lipid rafts, without significant changes in the lipid composition of the rafts. The interaction of AIBP with phosphatidylinositol 3-phosphate was blocked by AIBP substrate, NADPH (nicotinamide adenine dinucleotide phosphate), and both NADPH and silencing of Cdc42 interfered with the ability of AIBP to regulate lipid rafts and cholesterol efflux. CONCLUSIONS: Our findings indicate that an underlying mechanism of regulation of lipid rafts by AIBP involves PIP-dependent rearrangement of the cytoskeleton.


Assuntos
Citoesqueleto de Actina/enzimologia , Colesterol/metabolismo , Microdomínios da Membrana/enzimologia , Racemases e Epimerases/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Citoesqueleto de Actina/genética , Animais , Células HeLa , Humanos , Microdomínios da Membrana/genética , Camundongos , Fosfatidilinositol 3-Quinase/metabolismo , Transdução de Sinais , Células THP-1 , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo
3.
J Biol Chem ; 295(38): 13377-13392, 2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32732283

RESUMO

HIV-associated neurocognitive disorders (HANDs) are a frequent outcome of HIV infection. Effective treatment of HIV infection has reduced the rate of progression and severity but not the overall prevalence of HANDs, suggesting ongoing pathological process even when viral replication is suppressed. In this study, we investigated how HIV-1 protein Nef secreted in extracellular vesicles (exNef) impairs neuronal functionality. ExNef were rapidly taken up by neural cells in vitro, reducing the abundance of ABC transporter A1 (ABCA1) and thus cholesterol efflux and increasing the abundance and modifying lipid rafts in neuronal plasma membranes. ExNef caused a redistribution of amyloid precursor protein (APP) and Tau to lipid rafts and increased the abundance of these proteins, as well as of Aß42 ExNef further potentiated phosphorylation of Tau and activation of inflammatory pathways. These changes were accompanied by neuronal functional impairment. Disruption of lipid rafts with cyclodextrin reversed the phenotype. Short-term treatment of C57BL/6 mice with either purified recombinant Nef or exNef similarly resulted in reduced abundance of ABCA1 and elevated abundance of APP in brain tissue. The abundance of ABCA1 in brain tissue of HIV-infected human subjects diagnosed with HAND was lower, and the abundance of lipid rafts was higher compared with HIV-negative individuals. Levels of APP and Tau in brain tissue correlated with the abundance of Nef. Thus, modification of neuronal cholesterol trafficking and of lipid rafts by Nef may contribute to early stages of neurodegeneration and pathogenesis in HAND.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Infecções por HIV/metabolismo , HIV-1/metabolismo , Microdomínios da Membrana/metabolismo , Transtornos Neurocognitivos/metabolismo , Neurônios/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Proteínas tau/metabolismo , Transportador 1 de Cassete de Ligação de ATP/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Linhagem Celular Tumoral , Colesterol/genética , Colesterol/metabolismo , Infecções por HIV/complicações , Infecções por HIV/genética , Infecções por HIV/patologia , HIV-1/genética , Humanos , Microdomínios da Membrana/genética , Camundongos , Transtornos Neurocognitivos/etiologia , Transtornos Neurocognitivos/genética , Transtornos Neurocognitivos/patologia , Neurônios/patologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética , Proteínas tau/genética
4.
EMBO Rep ; 21(3): e48692, 2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-32072744

RESUMO

Dysregulation of lipid homeostasis is intimately associated with defects in insulin secretion, a key feature of type 2 diabetes. Here, we explore the role of the putative lipid transporter ABCA12 in regulating insulin secretion from ß-cells. Mice with ß-cell-specific deletion of Abca12 display impaired glucose-stimulated insulin secretion and eventual islet inflammation and ß-cell death. ABCA12's action in the pancreas is independent of changes in the abundance of two other cholesterol transporters, ABCA1 and ABCG1, or of changes in cellular cholesterol or ceramide content. Instead, loss of ABCA12 results in defects in the genesis and fusion of insulin secretory granules and increases in the abundance of lipid rafts at the cell membrane. These changes are associated with dysregulation of the small GTPase CDC42 and with decreased actin polymerisation. Our findings establish a new, pleiotropic role for ABCA12 in regulating pancreatic lipid homeostasis and insulin secretion.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Transportador 1 de Cassete de Ligação de ATP/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Camundongos
5.
PLoS Pathog ; 15(7): e1007907, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31344124

RESUMO

HIV infection has a profound effect on "bystander" cells causing metabolic co-morbidities. This may be mediated by exosomes secreted by HIV-infected cells and containing viral factors. Here we show that exosomes containing HIV-1 protein Nef (exNef) are rapidly taken up by macrophages releasing Nef into the cell interior. This caused down-regulation of ABCA1, reduction of cholesterol efflux and sharp elevation of the abundance of lipid rafts through reduced activation of small GTPase Cdc42 and decreased actin polymerization. Changes in rafts led to re-localization of TLR4 and TREM-1 to rafts, phosphorylation of ERK1/2, activation of NLRP3 inflammasome, and increased secretion of pro-inflammatory cytokines. The effects of exNef on lipid rafts and on inflammation were reversed by overexpression of a constitutively active mutant of Cdc42. Similar effects were observed in macrophages treated with exosomes produced by HIV-infected cells or isolated from plasma of HIV-infected subjects, but not with exosomes from cells and subjects infected with ΔNef-HIV or uninfected subjects. Mice injected with exNef exhibited monocytosis, reduced ABCA1 in macrophages, increased raft abundance in monocytes and augmented inflammation. Thus, Nef-containing exosomes potentiated pro-inflammatory response by inducing changes in cholesterol metabolism and reorganizing lipid rafts. These mechanisms may contribute to HIV-associated metabolic co-morbidities.


Assuntos
Infecções por HIV/metabolismo , Infecções por HIV/virologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Animais , Efeito Espectador , Colesterol/metabolismo , Exossomos/metabolismo , Exossomos/virologia , Células HEK293 , HIV-1 , Humanos , Inflamação/metabolismo , Inflamação/virologia , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/virologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Células RAW 264.7 , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptor Gatilho 1 Expresso em Células Mieloides/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
6.
Arterioscler Thromb Vasc Biol ; 37(7): 1301-1306, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28522696

RESUMO

OBJECTIVE: Apolipoprotein A-I (apoA-I) mimetic peptides have antiatherogenic properties of high-density lipoprotein in vitro and have been shown to inhibit atherosclerosis in vivo. It is unclear, however, if each in vitro antiatherogenic property of these peptides translates to a corresponding activity in vivo, and if so, which of these contributes most to reduce atherosclerosis. APPROACH AND RESULTS: The effect of 7 apoA-I mimetic peptides, which were developed to selectively reproduce a specific component of the antiatherogenic properties of apoA-I, on the development of atherosclerosis was investigated in apolipoprotein E-deficient mice fed a high-fat diet for 4 or 12 weeks. The peptides include those that selectively upregulate cholesterol efflux, or are anti-inflammatory, or have antioxidation properties. All the peptides studied effectively inhibited the in vivo development of atherosclerosis in this model to the same extent. However, none of the peptides had the same selective effect in vivo as they had exhibited in vitro. None of the tested peptides affected plasma lipoprotein profile; capacity of plasma to support cholesterol efflux was increased modestly and similarly for all peptides. CONCLUSIONS: There is a discordance between the selective in vitro and in vivo functional properties of apoA-I mimetic peptides, and the in vivo antiatherosclerotic effect of apoA-I-mimetic peptides is independent of their in vitro functional profile. Comparing the properties of apoA-I mimetic peptides in plasma rather than in the lipid-free state is better for predicting their in vivo effects on atherosclerosis.


Assuntos
Doenças da Aorta/prevenção & controle , Apolipoproteína A-I/farmacologia , Aterosclerose/prevenção & controle , Hipolipemiantes/farmacologia , Macrófagos/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Animais , Doenças da Aorta/sangue , Doenças da Aorta/patologia , Apolipoproteína A-I/farmacocinética , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/sangue , Aterosclerose/patologia , Mimetismo Biológico , Biomarcadores/sangue , Colesterol/sangue , Dieta Hiperlipídica , Modelos Animais de Doenças , Predisposição Genética para Doença , Lipoproteínas/sangue , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fragmentos de Peptídeos/farmacocinética , Fenótipo , Placa Aterosclerótica , Células RAW 264.7 , Distribuição Tecidual
7.
Cell Rep ; 16(1): 186-200, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27320924

RESUMO

Cytomegalovirus (HCMV) contains cholesterol, but how HCMV interacts with host cholesterol metabolism is unknown. We found that, in human fibroblasts, HCMV infection increased the efflux of cellular cholesterol, despite reducing the abundance of ABCA1. Mechanistically, viral protein US28 was acting through CDC42, rearranging actin microfilaments, causing association of actin with lipid rafts, and leading to a dramatic change in the abundance and/or structure of lipid rafts. These changes displaced ABCA1 from the cell surface but created new binding sites for apolipoprotein A-I, resulting in enhanced cholesterol efflux. The changes also reduced the inflammatory response in macrophages. HCMV infection modified the host lipidome profile and expression of several genes and microRNAs involved in cholesterol metabolism. In mice, murine CMV infection elevated plasma triglycerides but did not affect the level and functionality of high-density lipoprotein. Thus, HCMV, through its protein US28, reorganizes lipid rafts and disturbs cell cholesterol metabolism.


Assuntos
Colesterol/metabolismo , Citomegalovirus/metabolismo , Interações Hospedeiro-Patógeno , Microdomínios da Membrana/metabolismo , Receptores de Quimiocinas/metabolismo , Transdução de Sinais , Proteínas Virais/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Transporte Biológico , Infecções por Citomegalovirus , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibroblastos/virologia , Humanos , Inflamação/patologia , Metabolismo dos Lipídeos , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Células RAW 264.7
8.
J Pharmacol Exp Ther ; 353(3): 490-5, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25788712

RESUMO

Cyclophilins exert both intracellular and extracellular activities related to immune responses and inflammation, which have been implicated in pathogenesis of atherosclerosis. Pan-inhibition of cyclophilins has both pro- and antiatherosclerotic properties, but specific contributions of extracellular and intracellular cyclophilins to these effects have not been characterized. Here, using selective inhibitor of extracellular cyclophilins, we investigated the role of these molecules in atherosclerosis. Apolipoprotein E-null mice fed a high-fat diet received intraperitoneal injections every second day of either vehicle or two analogs of cyclosporine A (CsA): [Melle](4)-CsA (NIM811), a nonimmunosupressive cell-permeable inhibitor of both intracellular and extracellular cyclophilins; and [(4R)-4-[(6-carboxy-1H-benzo[d]imidazol-2-yl)-methyl]-4-methyl-l-threonine](1)-CsA (MM284), cell-impermeable analog only inhibiting extracellular cyclophilins. Development of atherosclerosis and composition of plaques in aorta and innominate artery were studied. Both analogs increased abundance and cross-sectional size of the atherosclerotic plaques in aorta but did not affect development of atherosclerosis in innominate artery. Neither compound affected abundance of macrophages and amount of vascular cell adhesion molecule-1 or nitrotyrosine in the plaques of both arteries. Both compounds reduced the amount of collagen in innominate artery without affecting abundance of collagen in aortic sinus. MM284, but not NIM811, significantly reduced plasma concentration of tumor necrosis factor-α (TNFα); neither compound affected plasma concentrations of interleukin (IL)-6, IL-10 or monocyte chemoattractant protein-1. Ratio between different populations of immune cells in blood or isolated from lymph nodes and spleen as well as plasma lipoprotein profile were unaffected by both compounds. In conclusion, selective inhibition of extracellular cyclophilins reduced TNFα levels in plasma but increased atherosclerosis.


Assuntos
Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/induzido quimicamente , Aterosclerose/fisiopatologia , Ciclofilinas/antagonistas & inibidores , Ciclosporinas/farmacologia , Imunossupressores/farmacologia , Animais , Estudos Transversais , Citocinas/sangue , Lipoproteínas/sangue , Masculino , Camundongos , Camundongos Knockout , Placa Aterosclerótica/patologia
9.
FASEB J ; 28(7): 2828-39, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24642731

RESUMO

Patients with HIV are at an increased risk of cardiovascular disease. In this study we investigated the effect of Nef, a secreted HIV protein responsible for the impairment of cholesterol efflux, on the development of atherosclerosis in two animal models. ApoE(-/-) mice fed a high-fat diet and C57BL/6 mice fed a high-fat, high-cholesterol diet were injected with recombinant Nef (40 ng/injection) or vehicle, and the effects of Nef on development of atherosclerosis, inflammation, and dyslipidemia were assessed. In apoE(-/-) mice, Nef significantly increased the size of atherosclerotic lesions and caused vessel remodeling. Nef caused elevation of total cholesterol and triglyceride levels in the plasma while reducing high-density lipoprotein cholesterol levels. These changes were accompanied by a reduction of ABCA1 abundance in the liver, but not in the vessels. In C57BL/6 mice, Nef caused a significant number of lipid-laden macrophages presented in adventitia of the vessels; these cells were absent from the vessels of control mice. Nef caused sharp elevations of plasma triglyceride levels and body weight. Taken together, our findings suggest that Nef causes dyslipidemia and accumulation of cholesterol in macrophages within the vessel wall, supporting the role of Nef in pathogenesis of atherosclerosis in HIV-infected patients.-Cui, H. L., Ditiatkovski, M., Kesani, R., Bobryshev, Y. V., Liu, Y., Geyer, M., Mukhamedova, N., Bukrinsky, M., Sviridov, D. HIV protein Nef causes dyslipidemia and formation of foam cells in mouse models of atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Dislipidemias/metabolismo , Células Espumosas/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Animais , Apolipoproteínas E/metabolismo , Aterosclerose/sangue , Aterosclerose/etiologia , Colesterol/sangue , Glicosaminoglicanos/metabolismo , HIV/metabolismo , Infecções por HIV/sangue , Infecções por HIV/complicações , Infecções por HIV/metabolismo , Inflamação/metabolismo , Lipoproteínas HDL/sangue , Fígado/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Triglicerídeos/sangue
10.
Cell Metab ; 18(2): 225-38, 2013 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-23931754

RESUMO

ABCA12 is involved in the transport of ceramides in skin, but it may play a wider role in lipid metabolism. We show that, in Abca12-deficient macrophages, cholesterol efflux failed to respond to activation with LXR agonists. Abca12 deficiency caused a reduction in the abundance of Abca1, Abcg1, and Lxrß. Overexpression of Lxrß reversed the effects. Mechanistically, Abca12 deficiency did not affect expression of genes involved in cholesterol metabolism. Instead, a physical association between Abca1, Abca12, and Lxrß proteins was established. Abca12 deficiency enhanced interaction between Abca1 and Lxrß and the degradation of Abca1. Overexpression of ABCA12 in HeLa-ABCA1 cells increased the abundance and stability of ABCA1. Abca12 deficiency caused an accumulation of cholesterol in macrophages and the formation of foam cells, impaired reverse cholesterol transport in vivo, and increased the development of atherosclerosis in irradiated Apoe(-/-) mice reconstituted with Apoe(-/-)Abca12(-/-) bone marrow. Thus, ABCA12 regulates the cellular cholesterol metabolism via an LXRß-dependent posttranscriptional mechanism.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Colesterol/metabolismo , Macrófagos/metabolismo , Receptores Nucleares Órfãos/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/deficiência , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Transporte Biológico/genética , Linhagem Celular , Células Espumosas/metabolismo , Células HeLa , Humanos , Metabolismo dos Lipídeos/genética , Lipoproteínas/metabolismo , Receptores X do Fígado , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Nucleares Órfãos/biossíntese
11.
PLoS One ; 8(7): e68802, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874769

RESUMO

Apolipoprotein A-I (apoA-I) mimetic peptides are considered a promising novel therapeutic approach to prevent and/or treat atherosclerosis. An apoA-I mimetic peptide ELK-2A2K2E was designed with a reductionist approach and has shown exceptional activity in supporting cholesterol efflux but modest anti-inflammatory and anti-oxidant properties in vitro. In this study we compared these in vitro properties with the capacity of this peptide to modify rates of reverse cholesterol transport and development of atherosclerosis in mouse models. The peptide enhanced the rate of reverse cholesterol transport in C57BL/6 mice and reduced atherosclerosis in Apoe(-/-) mice receiving a high fat diet. The peptide modestly reduced the size of the plaques in aortic arch, but was highly active in reducing vascular inflammation and oxidation. Administration of the peptide to Apoe(-/-) mice on a high fat diet reduced the levels of total, high density lipoprotein and non-high density lipoprotein cholesterol and triglycerides. It increased the proportion of smaller HDL particles in plasma at the expense of larger HDL particles, and increased the capacity of the plasma to support cholesterol efflux. Thus, ELK-2A2K2E peptide reduced atherosclerosis in Apoe(-/-) mice, however, the functional activity profile after chronic in vivo administration was different from that found in acute in vitro studies.


Assuntos
Apolipoproteína A-I/química , Aterosclerose/metabolismo , Biomimética , Colesterol/metabolismo , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Apolipoproteína A-I/sangue , Apolipoproteínas E/genética , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Transporte Biológico/efeitos dos fármacos , Modelos Animais de Doenças , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/patologia , Masculino , Camundongos , Camundongos Knockout , Oxirredução/efeitos dos fármacos , Peptídeos/química , Peptídeos/farmacocinética
12.
Curr Pharm Des ; 19(33): 5904-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23438962

RESUMO

Atherosclerosis is a chronic disease with a significant inflammatory component. Recent studies indicate a role of extracellular cyclophilins as contributors to endothelial inflammation and pathogenesis of atherosclerosis. In this article, we review current literature on pro-inflammatory activities of extracellular cyclophilins and discuss possible approaches to selectively target this novel proinflammatory factor.


Assuntos
Aterosclerose/enzimologia , Fatores Quimiotáticos/imunologia , Ciclofilinas/imunologia , Endotélio Vascular/imunologia , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/imunologia , Basigina/imunologia , Ciclofilinas/antagonistas & inibidores , Ciclosporinas/farmacologia , Ciclosporinas/uso terapêutico , Endotélio Vascular/efeitos dos fármacos , Humanos , Metaloproteinases da Matriz/imunologia , Terapia de Alvo Molecular
13.
Cardiovasc Res ; 95(1): 77-85, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22492684

RESUMO

AIMS: The mechanisms underlying cardiac fibrosis in hypertension are yet to be defined, although inflammatory cells, fibroblasts, and cytokines have been implicated. Here, we investigated the role of interleukin-4 (IL-4) in cardiac fibrosis, which is elevated in the hypertensive heart. IL-4 has been shown to be pro-fibrotic in the liver and the lung, but its role in cardiac fibrosis has not been investigated. METHODS AND RESULTS: Cardiac fibrosis was induced in mice by constricting the aorta between the two carotid arteries. Fourteen days later marked left ventricular fibrosis developed together with expression of IL-4. Anti-IL-4 neutralizing antibodies attenuated this fibrosis without affecting blood pressure or expression of the transforming growth factor-beta system. The reduction in fibrosis was associated with reductions in interstitial fibroblasts and macrophages together with reductions in proliferating cells and expression of monocyte chemoattractant protein-1 (MCP-1). Since mast cells are a source of IL-4, we also assessed their role in fibrosis. Cromolyn, a mast cell inhibitor attenuated mast cell degranulation as well as IL-4 mRNA expression and cardiac fibrosis without affecting blood pressure. Treatment with Cromolyn also reduced interstitial fibroblasts and macrophages in regions of developing fibrosis as well MCP-1 expression. CONCLUSION: This study demonstrates for the first time that IL-4, most likely produced by mast cells in the heart during pressure overload, is a significant contributor to cardiac fibrosis. Targeting this cytokine may be a useful therapeutic strategy to limit cardiac fibrosis.


Assuntos
Hipertensão/patologia , Interleucina-4/fisiologia , Miocárdio/patologia , Animais , Coartação Aórtica/etiologia , Proliferação de Células , Fibrose , Macrófagos/fisiologia , Masculino , Mastócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Fator de Crescimento Transformador beta/fisiologia
14.
Arterioscler Thromb Vasc Biol ; 26(10): 2337-44, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16873730

RESUMO

OBJECTIVE: Granulocyte-macrophage colony-stimulating factor (GM-CSF) is expressed in atherosclerotic lesions but its significance for lesion development is unknown. Consequently, we investigated the significance of GM-CSF expression for development of atherosclerotic lesions in apolipoprotein E-deficient (apoE(-/-)) mice. METHODS AND RESULTS: We generated apoE(-/-) mice deficient in GM-CSF (apoE(-/-).GM-CSF(-/-) mice), fed them a high-fat diet, and compared lesion development with apoE(-/-) mice. We measured lesion size, macrophage, smooth muscle cell, and collagen accumulation at the aortic sinus, and expression of genes that regulate cholesterol transport and inflammation. No differences in serum cholesterol were found between the 2 groups. Lesion size in hyperlipidemic apoE(-/-).GM-CSF(-/-) increased by 30% (P<0.05), macrophage accumulation doubled, and collagen content reduced by 15% (P<0.05); smooth muscle cell accumulation and vascularity were unaffected. Analysis of PPAR-gamma, ABCA1, and CD36 in lesions showed reduced expression (50%, 65%, and 55%, respectively), whereas SR-A doubled. In peritoneal macrophages, PPAR-gamma and ABCA1 expression was also reduced by 50% and 70%, respectively, as was cholesterol efflux, by 50%. In lesions, pro-inflammatory MCP-1 and tumor necrosis factor (TNF)-alpha expression increased 2- and 3.5-fold, respectively, vascular cell adhesion molecule (VCAM)-1 expression enhanced and interleukin (IL)-1 receptor antagonist reduced by 50%. CONCLUSIONS: GM-CSF deficiency increases atherosclerosis under hypercholesterolemic conditions, indicating antiatherogenic role for GM-CSF. We suggest this protective role is mediated by PPAR-gamma and ABCA1, molecules that affect cholesterol homeostasis and inflammation.


Assuntos
Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Aterosclerose/patologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/deficiência , Macrófagos/metabolismo , PPAR gama/antagonistas & inibidores , Transportador 1 de Cassete de Ligação de ATP , Animais , Aterosclerose/etiologia , Quimiocina CCL2/metabolismo , Colesterol/metabolismo , Colágeno/antagonistas & inibidores , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Hiperlipidemias/complicações , Macrófagos/patologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/patologia , Seio Aórtico/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA