Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 9(1): 14547, 2019 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-31601924

RESUMO

The anti-apoptotic protein myeloid cell leukemia 1 (Mcl-1) plays an important role in survival and differentiation of leukocytes, more specifically of neutrophils. Here, we investigated the impact of myeloid Mcl-1 deletion in atherosclerosis. Western type diet fed LDL receptor-deficient mice were transplanted with either wild-type (WT) or LysMCre Mcl-1fl/fl (Mcl-1-/-) bone marrow. Mcl-1 myeloid deletion resulted in enhanced apoptosis and lipid accumulation in atherosclerotic plaques. In vitro, Mcl-1 deficient macrophages also showed increased lipid accumulation, resulting in increased sensitivity to lipid-induced cell death. However, plaque size, necrotic core and macrophage content were similar in Mcl-1-/- compared to WT mice, most likely due to decreased circulating and plaque-residing neutrophils. Interestingly, Mcl-1-/- peritoneal foam cells formed up to 45% more multinucleated giant cells (MGCs) in vitro compared to WT, which concurred with an increased MGC presence in atherosclerotic lesions of Mcl-1-/- mice. Moreover, analysis of human unstable atherosclerotic lesions also revealed a significant inverse correlation between MGC lesion content and Mcl-1 gene expression, coinciding with the mouse data. Taken together, these findings suggest that myeloid Mcl-1 deletion leads to a more apoptotic, lipid and MGC-enriched phenotype. These potentially pro-atherogenic effects are however counteracted by neutropenia in circulation and plaque.


Assuntos
Apoptose , Células Gigantes/citologia , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Células 3T3 , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Diferenciação Celular , Deleção de Genes , Humanos , Imuno-Histoquímica , Lipídeos/química , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Neutrófilos/metabolismo , Fenótipo , Placa Aterosclerótica/metabolismo
2.
Sci Rep ; 7(1): 3086, 2017 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-28596542

RESUMO

Proapoptotic Bcl-2 family member Bim is particularly relevant for deletion of autoreactive and activated T and B cells, implicating Bim in autoimmunity. As atherosclerosis is a chronic inflammatory process with features of autoimmune disease, we investigated the impact of hematopoietic Bim deficiency on plaque formation and parameters of plaque stability. Bim -/- or wild type bone marrow transplanted ldlr -/- mice were fed a Western type diet (WTD) for 5 or 10 weeks, after which they were immunophenotyped and atherosclerotic lesions were analyzed. Bim -/- transplanted mice displayed splenomegaly and overt lymphocytosis. CD4+ and CD8+ T cells were more activated (increased CD69 and CD71 expression, increased interferon gamma production). B cells were elevated by 147%, with a shift towards the pro-atherogenic IgG-producing B2 cell phenotype, resulting in a doubling of anti-oxLDL IgG1 antibody titers in serum of bim -/- mice. Bim -/- mice displayed massive intraplaque accumulation of Ig complexes and of lesional T cells, although this did not translate in changes in plaque size or stability features (apoptotic cell and macrophage content). The surprising lack in plaque phenotype despite the profound pro-atherogenic immune effects may be attributable to the sharp reduction of serum cholesterol levels in WTD fed bim -/- mice.


Assuntos
Aterosclerose/genética , Doenças Autoimunes/etiologia , Proteína 11 Semelhante a Bcl-2/deficiência , Inflamação/etiologia , Leucócitos/imunologia , Leucócitos/metabolismo , Receptores de LDL/deficiência , Animais , Apoptose/genética , Doenças Autoimunes/patologia , Proteína 11 Semelhante a Bcl-2/genética , Transplante de Medula Óssea , Modelos Animais de Doenças , Hiperlipidemias , Imunidade Humoral , Imunoglobulinas/imunologia , Inflamação/patologia , Contagem de Linfócitos , Camundongos , Camundongos Knockout , Receptores de LDL/genética , Esplenomegalia , Células Th1/imunologia , Células Th1/metabolismo
3.
Sci Rep ; 7: 42688, 2017 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-28202909

RESUMO

Toll-like receptors (TLR) provide a critical link between innate and adaptive immunity, both important players in atherosclerosis. Since evidence for the role of TLR5 is lacking, we aimed to establish this in the immune axis of atherosclerosis. We assessed the effect of the TLR5-specific ligand Flagellin on macrophage maturation and T-cell polarisation. Next, we generated TLR5-/-LDLr-/- chimeras to study the effect of hematopoietic TLR5 deficiency on atherosclerosis formation. Flagellin stimulation did not influence wildtype or TLR5-/- macrophage maturation. Only in wildtype macrophages, Flagellin exposure increased MCP-1 and IL6 expression. Flagellin alone reduced T-helper 1 proliferation, which was completely overruled in the presence of T-cell receptor activation. In vivo, hematopoietic TLR5 deficiency attenuated atherosclerotic lesion formation by ≈25% (1030*103 ± 63*103 vs. 792*103 ± 61*103 µm2; p = 0.013) and decreased macrophage area (81.3 ± 12.0 vs. 44.2 ± 6.6 µm2; p = 0.011). In TLR5-/- chimeric mice, we observed lower IL6 plasma levels (36.4 ± 5.6 vs. 15.1 ± 2.2 pg/mL; p = 0.003), lower (activated) splenic CD4+ T-cell content (32.3 ± 2.1 vs. 21.0 ± 1.2%; p = 0.0018), accompanied by impaired T-cell proliferative responses. In conclusion, hematopoietic TLR5 deficiency inhibits atherosclerotic lesion formation by attenuated macrophage accumulation and defective T-cell responsiveness.


Assuntos
Aterosclerose/etiologia , Aterosclerose/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Receptor 5 Toll-Like/deficiência , Animais , Aterosclerose/sangue , Aterosclerose/patologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Contagem de Leucócitos , Camundongos , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo , Placa Aterosclerótica/genética , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Subpopulações de Linfócitos T/citologia
4.
J Mol Cell Cardiol ; 74: 44-52, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24816217

RESUMO

AIMS: The SDF-1α/CXCR4 dyad was previously shown by us and others to be instrumental in intimal hyperplasia as well as early stage atherosclerosis. We here sought to investigate its impact on clinically relevant stages of atherosclerosis in mouse and man. METHODS AND RESULTS: Immunohistochemical analysis of CXCR4 expression in human atherosclerotic lesions revealed a progressive accumulation of CXCR4(+) cells during plaque progression. To address causal involvement of CXCR4 in advanced stages of atherosclerosis we reconstituted LDLr(-/-) mice with autologous bone marrow infected with lentivirus encoding SDF-1α antagonist or CXCR4 degrakine, which effects proteasomal degradation of CXCR4. Functional CXCR4 blockade led to progressive plaque expansion with disease progression, while also promoting intraplaque haemorrhage. Moreover, CXCR4 knockdown was seen to augment endothelial adhesion of neutrophils. Concordant with this finding, inhibition of CXCR4 function increased adhesive capacity and reduced apoptosis of neutrophils and resulted in hyperactivation of circulating neutrophils. Compatible with a role of the neutrophil CXCR4 in end-stage atherosclerosis, CXCR4 expression by circulating neutrophils was lowered in patients with acute cardiovascular syndromes. CONCLUSION: In conclusion, CXCR4 contributes to later stages of plaque progression by perturbing neutrophil function.


Assuntos
Aterosclerose/genética , Hemorragia/genética , Neutrófilos/metabolismo , Placa Aterosclerótica/genética , Receptores CXCR4/genética , Animais , Aterosclerose/metabolismo , Aterosclerose/patologia , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Adesão Celular , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Progressão da Doença , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Regulação da Expressão Gênica , Vetores Genéticos , Hemorragia/metabolismo , Hemorragia/patologia , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Camundongos , Camundongos Knockout , Neutrófilos/patologia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais
5.
Atherosclerosis ; 235(1): 196-203, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24858338

RESUMO

AIM: Neuropeptide Y is an abundantly expressed neurotransmitter capable of modulating both immune and metabolic responses related to the development of atherosclerosis. NPY receptors are expressed by a number of vascular wall cell types, among which mast cells. However, the direct effects of NPY on atherosclerotic plaque development and progression remain to be investigated. In this study we thus aimed to determine whether NPY is expressed in atherosclerotic plaques and to establish its role in atherosclerotic plaque development. METHODS AND RESULTS: NPY expression was seen to be increased up to 2-fold in unstable human endarterectomy plaques, as compared to stable plaques, and to be significantly upregulated during lesion progression in apoE(-/-) mice. In apoE(-/-) mice focal overexpression of NPY in the carotid artery significantly increased atherosclerotic plaque size compared to controls, while plaque composition was unaffected. Interestingly, perivascular mast cell activation was significantly higher in the NPY-overexpressing mice, suggesting that NPY may impact plaque progression in part via mast cell activation. Furthermore, in vitro NPY-induced murine mast cell activation resulted in the release of pro-atherogenic mediators including IL-6 and tryptase. CONCLUSIONS: Our data show that NPY expression is increased during atherogenesis and in particular in unstable plaques. Furthermore, perivascular overexpression of NPY promoted plaque development and perivascular mast cell activation, suggestive of a role for NPY-induced mast cell activation in lesion progression.


Assuntos
Aterosclerose/sangue , Mastócitos/citologia , Neuropeptídeo Y/sangue , Placa Aterosclerótica/patologia , Animais , Apolipoproteínas E/sangue , Progressão da Doença , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células HEK293 , Humanos , Imuno-Histoquímica , Inflamação/sangue , Interleucina-6/sangue , Lentivirus/genética , Camundongos , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Placa Aterosclerótica/sangue , Triptases/sangue
6.
Arterioscler Thromb Vasc Biol ; 33(10): 2355-65, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23825366

RESUMO

OBJECTIVE: Myocardin, a potent transcriptional coactivator of serum response factor, is involved in vascular development and promotes a contractile smooth muscle phenotype. Myocardin levels are reduced during vascular injury, in association with phenotypic switching of smooth muscle cells (SMCs). However, the direct role of myocardin in vascular disease is unclear. APPROACH AND RESULTS: We show that re-expression of myocardin prevents the vascular injury response in murine carotid arteries, with reduced neointima formation due to decreased SMC migration and proliferation. Myocardin reduced SMC migration by downregulating platelet-derived growth factor receptor-ß (PDGFRB) expression. Pdgfrb was regulated by myocardin-induced miR-24 and miR-29a expression, and antagonizing these microRNAs restored SMC migration. Furthermore, using miR-24 and miR-29a mimics, we demonstrated that miR-29a directly regulates Pdgfrb expression at the 3' untranslated region while miR-24 has an indirect effect on Pdgfrb levels. Myocardin heterozygous-null mice showed an augmented neointima formation with increased SMC migration and proliferation, demonstrating that endogenous levels of myocardin are a critical regulator of vessel injury responses. CONCLUSIONS: Our results extend the function of myocardin from a developmental role to a pivotal regulator of SMC phenotype in response to injury, and this transcriptional coactivator may be an attractive target for novel therapeutic strategies in vascular disease.


Assuntos
Lesões das Artérias Carótidas/metabolismo , Estenose das Carótidas/metabolismo , MicroRNAs/metabolismo , Músculo Liso Vascular/metabolismo , Proteínas Nucleares/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transativadores/metabolismo , Regiões 3' não Traduzidas , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Lesões das Artérias Carótidas/prevenção & controle , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/patologia , Estenose das Carótidas/genética , Estenose das Carótidas/patologia , Estenose das Carótidas/prevenção & controle , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/lesões , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Neointima , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Ratos , Ratos Wistar , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais , Fatores de Tempo , Transativadores/deficiência , Transativadores/genética , Transfecção
7.
PLoS One ; 8(5): e63360, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23700419

RESUMO

AIMS: Altered sphingosine 1-phosphate (S1P) homeostasis and signaling is implicated in various inflammatory diseases including atherosclerosis. As S1P levels are tightly controlled by S1P lyase, we investigated the impact of hematopoietic S1P lyase (Sgpl1(-/-)) deficiency on leukocyte subsets relevant to atherosclerosis. METHODS AND RESULTS: LDL receptor deficient mice that were transplanted with Sgpl1(-/-) bone marrow showed disrupted S1P gradients translating into lymphopenia and abrogated lymphocyte mitogenic and cytokine response as compared to controls. Remarkably however, Sgpl1(-/-) chimeras displayed mild monocytosis, due to impeded stromal retention and myelopoiesis, and plasma cytokine and macrophage expression patterns, that were largely compatible with classical macrophage activation. Collectively these two phenotypic features of Sgpl1 deficiency culminated in diminished atherogenic response. CONCLUSIONS: Here we not only firmly establish the critical role of hematopoietic S1P lyase in controlling S1P levels and T cell trafficking in blood and lymphoid tissue, but also identify leukocyte Sgpl1 as critical factor in monocyte macrophage differentiation and function. Its, partly counterbalancing, pro- and anti-inflammatory activity spectrum imply that intervention in S1P lyase function in inflammatory disorders such as atherosclerosis should be considered with caution.


Assuntos
Aldeído Liases/deficiência , Aterosclerose/enzimologia , Placa Aterosclerótica/enzimologia , Receptores de LDL/deficiência , Aldeído Liases/genética , Animais , Aterosclerose/imunologia , Aterosclerose/patologia , Células da Medula Óssea/enzimologia , Diferenciação Celular , Feminino , Hematopoese , Contagem de Linfócitos , Linfopenia/enzimologia , Linfopenia/imunologia , Lisofosfolipídeos/metabolismo , Macrófagos/enzimologia , Macrófagos/imunologia , Macrófagos/fisiologia , Camundongos , Camundongos Knockout , Neutrófilos/enzimologia , Fenótipo , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/patologia , Receptores de LDL/genética , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Baço/metabolismo
8.
J Lipid Res ; 54(5): 1265-74, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23396975

RESUMO

Lysophosphatidic acid (LPA), a bioactive lysophospholipid, accumulates in the atherosclerotic plaque. It has the capacity to activate mast cells, which potentially exacerbates plaque progression. In this study, we thus aimed to investigate whether LPA contributes to plaque destabilization by modulating mast cell function. We here show by an imaging mass spectrometry approach that several LPA species are present in atherosclerotic plaques. Subsequently, we demonstrate that LPA is a potent mast cell activator which, unlike other triggers, favors release of tryptase. Local perivascular administration of LPA to an atherosclerotic carotid artery segment increases the activation status of perivascular mast cells and promotes intraplaque hemorrhage and macrophage recruitment without impacting plaque cell apoptosis. The mast cell stabilizer cromolyn could prevent intraplaque hemorrhage elicited by LPA-mediated mast cell activation. Finally, the involvement of mast cells in these events was further emphasized by the lack of effect of perivascular LPA administration in mast cell deficient animals. We demonstrate that increased accumulation of LPA in plaques induces perivascular mast cell activation and in this way contributes to plaque destabilization in vivo. This study points to local LPA availability as an important factor in atherosclerotic plaque stability.


Assuntos
Inflamação/metabolismo , Lisofosfolipídeos/metabolismo , Mastócitos/metabolismo , Placa Aterosclerótica/patologia , Animais , Apoptose/efeitos dos fármacos , Artérias Carótidas/diagnóstico por imagem , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Células Cultivadas , Cromolina Sódica/farmacologia , Inflamação/patologia , Macrófagos/metabolismo , Mastócitos/efeitos dos fármacos , Mastócitos/patologia , Camundongos , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/metabolismo , Radiografia , Triptases/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 33(3): e75-83, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23288165

RESUMO

OBJECTIVE: Despite common disbelief that neutrophils are involved in atherosclerosis, evidence is accumulating for a causal role of neutrophils in atherosclerosis. CC chemokine ligand (CCL)3 is an inflammatory chemokine and its expression is significantly increased during atherosclerotic lesion formation in mice. It has recently been shown that under conditions of inflammation neutrophils can migrate along a CCL3 gradient. In this study, we aimed to elucidate the role of leukocyte-derived CCL3 in atherogenesis. METHODS AND RESULTS: Irradiated low density lipoprotein receptor(-/-) mice, reconstituted with CCL3(-/-) or littermate bone marrow showed markedly reduced CCL3 response to lipopolysaccharide treatment, establishing the critical relevance of leukocytes as source of CCL3. Hematopoietic deficiency of CCL3 significantly reduced aortic sinus lesion formation by 31% after 12 weeks of western-type diet. Interestingly, whereas plaque macrophage, collagen, and vascular smooth muscle cell content were unchanged, neutrophil adhesion to and presence in plaques was significantly attenuated in CCL3(-/-) chimeras. These mice had reduced circulating neutrophil numbers, which could be ascribed to an increased neutrophil turnover and CCL3(-/-) neutrophils were shown to be less responsive toward the neutrophil chemoattractant CXC chemokine ligand 1. CONCLUSIONS: Our data indicate that under conditions of acute inflammation leukocyte-derived CCL3 can induce neutrophil chemotaxis toward the atherosclerotic plaque, thereby accelerating lesion formation.


Assuntos
Doenças das Artérias Carótidas/prevenção & controle , Artéria Carótida Primitiva/imunologia , Quimiocina CCL3/deficiência , Quimiotaxia de Leucócito , Leucócitos/imunologia , Infiltração de Neutrófilos , Neutrófilos/imunologia , Animais , Apoptose , Transplante de Medula Óssea , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/imunologia , Doenças das Artérias Carótidas/patologia , Artéria Carótida Primitiva/patologia , Adesão Celular , Células Cultivadas , Quimiocina CCL3/genética , Quimiocina CXCL1/metabolismo , Ciclofosfamida , Gorduras na Dieta , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutropenia/induzido quimicamente , Neutropenia/imunologia , Placa Aterosclerótica , RNA Mensageiro/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Fatores de Tempo , Irradiação Corporal Total
10.
FASEB J ; 27(1): 265-76, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23047899

RESUMO

Leukocyte chemotaxis is deemed instrumental in initiation and progression of atherosclerosis. It is mediated by G-protein-coupled receptors (e.g., CCR2 and CCR5), the activity of which is controlled by G-protein-coupled receptor kinases (GRKs). In this study, we analyzed the effect of hematopoietic deficiency of a potent regulator kinase of chemotaxis (GRK2) on atherogenesis. LDL receptor-deficient (LDLr(-/-)) mice with heterozygous hematopoietic GRK2 deficiency, generated by bone marrow transplantation (n=15), displayed a dramatic attenuation of plaque development, with 79% reduction in necrotic core and increased macrophage content. Circulating monocytes decreased and granulocytes increased in GRK2(+/-) chimeras, which could be attributed to diminished granulocyte colony-forming units in bone marrow. Collectively, these data pointed to myeloid cells as major mediators of the impaired atherogenic response in GRK2(+/-) chimeras. LDLr(-/-) mice with macrophage/granulocyte-specific GRK2 deficiency (LysM-Cre GRK2(flox/flox); n=8) failed to mimic the aforementioned phenotype, acquitting these cells as major responsible subsets for GRK2 deficiency-associated atheroprotection. To conclude, even partial hematopoietic GRK2 deficiency prevents atherosclerotic lesion progression beyond the fatty streak stage, identifying hematopoietic GRK2 as a potential target for intervention in atherosclerosis.


Assuntos
Aterosclerose/patologia , Quinase 2 de Receptor Acoplado a Proteína G/genética , Receptores de LDL/genética , Animais , Apoptose , Aterosclerose/genética , Feminino , Citometria de Fluxo , Camundongos , Camundongos Knockout , Fagocitose
11.
Atherosclerosis ; 223(2): 332-41, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22748276

RESUMO

OBJECTIVE: The ABCA2 transporter shares high structural homology to ABCA1, which is crucial for the removal of excess cholesterol from macrophages and, by extension, in atherosclerosis. It has been suggested that ABCA2 sequesters cholesterol inside the lysosomes, however, little is known of the macrophage-specific role of ABCA2 in regulating lipid homeostasis in vivo and in modulating susceptibility to atherosclerosis. METHODS: Chimeras with dysfunctional macrophage ABCA2 were generated by transplantation of bone marrow from ABCA2 knockout (KO) mice into irradiated LDL receptor (LDLr) KO mice. RESULTS: Interestingly, lack of ABCA2 in macrophages resulted in a diminished lesion size in the aortic root (-24.5%) and descending thoracic aorta (-36.6%) associated with a 3-fold increase in apoptotic cells, as measured by both caspase 3 and TUNEL. Upon oxidized LDL exposure, macrophages from wildtype (WT) transplanted animals developed filipin-positive droplets in lysosomal-like compartments, corresponding to free cholesterol (FC) accumulation. In contrast, ABCA2-deficient macrophages displayed an abnormal diffuse distribution of FC over peripheral regions. The accumulation of neutral sterols in lipid droplets was increased in ABCA2-deficient macrophages, but primarily in cytoplasmic clusters and not in lysosomes. Importantly, apoptosis of oxLDL loaded macrophages lacking ABCA2 was increased 2.7-fold, probably as a consequence of the broad cellular distribution of FC. CONCLUSIONS: Lack of functional ABCA2 generates abnormalities in intracellular lipid distribution/trafficking in macrophages consistent with its lysosomal sequestering role, leading to an increased susceptibility to apoptosis in response to oxidized lipids and reduced atherosclerotic lesion development.


Assuntos
Transportadores de Cassetes de Ligação de ATP/deficiência , Aorta/metabolismo , Doenças da Aorta/prevenção & controle , Apoptose , Aterosclerose/prevenção & controle , Colesterol/metabolismo , Macrófagos/metabolismo , Receptores de LDL/deficiência , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Aorta/patologia , Doenças da Aorta/etiologia , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Aterosclerose/etiologia , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Transplante de Medula Óssea , Caspase 3/metabolismo , Colesterol/sangue , Modelos Animais de Doenças , Filipina/metabolismo , Células Espumosas/metabolismo , Células Espumosas/patologia , Homeostase , Marcação In Situ das Extremidades Cortadas , Lipoproteínas LDL/metabolismo , Lisossomos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Receptores de LDL/genética , Fatores de Tempo , Quimeras de Transplante , Irradiação Corporal Total
12.
Arterioscler Thromb Vasc Biol ; 32(9): 2197-205, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22743060

RESUMO

OBJECTIVE: The A(2B) adenosine receptor (A(2B)R) is highly expressed in macrophages and vascular smooth muscle cells and has been established as an important regulator of inflammation and vascular adhesion. Recently, it has been demonstrated that A(2B)R deficiency enhances neointimal lesion formation after vascular injury. Therefore, we hypothesize that A(2B)R agonism protects against injury-induced intimal hyperplasia. METHODS AND RESULTS: Apolipoprotein E-deficient mice were fed a Western-type diet for 1 week, after which the left common carotid artery was denuded. Mice were treated with the A(2B) receptor agonist BAY60-6583 or vehicle control for 18 days. Interestingly, lumen stenosis as defined by the neointima/lumen ratio was inhibited by treatment with the A(2B) receptor agonist, caused by reduced smooth muscle cell proliferation. Collagen content was significantly increased in the BAY60-6583-treated mice, whereas macrophage content remained unchanged. In vitro, vascular smooth muscle cell proliferation decreased dose dependently whereas collagen content of cultured smooth muscle cells was increased by BAY60-6583. CONCLUSIONS: Our data show that activation of the adenosine A(2B) receptor protects against vascular injury, while it also enhances plaque stability as indicated by increased collagen content. These outcomes thus point to A(2B) receptor agonism as a new therapeutic approach in the prevention of restenosis.


Assuntos
Agonistas do Receptor A2 de Adenosina/farmacologia , Aminopiridinas/farmacologia , Apolipoproteínas E/deficiência , Fármacos Cardiovasculares/farmacologia , Lesões das Artérias Carótidas/tratamento farmacológico , Estenose das Carótidas/prevenção & controle , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Receptor A2B de Adenosina/efeitos dos fármacos , Animais , Apolipoproteínas E/genética , Células CHO , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Artéria Carótida Primitiva/efeitos dos fármacos , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/patologia , Estenose das Carótidas/genética , Estenose das Carótidas/metabolismo , Estenose das Carótidas/patologia , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno/metabolismo , Cricetinae , Cricetulus , Gorduras na Dieta , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Músculo Liso Vascular/lesões , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Neointima , Ativação de Neutrófilo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Receptor A2B de Adenosina/genética , Receptor A2B de Adenosina/metabolismo , Fatores de Tempo , Transfecção
13.
Thromb Haemost ; 107(3): 552-61, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22234485

RESUMO

Sphingosine 1-phosphate (S1P), a lysosphingolipid associated with high-density lipoprotein (HDL), contributes to the anti-atherogenic potential attributed to this lipoprotein. This study examined whether a reduction of S1P plasma levels affects atherosclerosis in a murine model of disease. LDL-R(-/-)mice on Western diet were given ABC294640, an inhibitor of sphingosine kinase (SphK) for 16 weeks. ABC294640 decreased plasma S1P by approximately 30%. However, ABC294640 failed to affect atherosclerotic lesion formation. Plasma triglycerides were reduced whereas total and HDL-cholesterol remained unchanged in course of ABC294640 treatment. ABC294640 increased plasma interleukin (IL)-12p70 and RANTES concentration as well as IL-12p70, RANTES and interferon (IFN)-γ production by peritoneal cells and this was paralleled by enhanced activity of peritoneal and spleen dendritic cells as evidenced by up-regulation of CD86 and MHC-II on CD11c(+) cells. As a consequence, increased T-cell activation was noted in ABC294640-treated mice as indicated by enhanced CD4(+) splenocyte proliferation, IFN-γ and IL-2 production, and CD69 expression. Concomitantly, however, ABC294640 treatment redistributed CD4(+) and CD8(+) cells from blood to lymphatic organs and reduced T-cell number within atherosclerotic lesions. In addition, plasma sVCAM-1, sICAM-1, and MCP-1 levels as well as in vivo leukocyte adhesion and CCL19-induced T-cell penetration into peritoneum were lower in ABC294640-treated animals. In vitro experiments demonstrated reduced VCAM-1 and ICAM-1 expression and lymphocyte adhesion to endothelial cells exposed to ABC294640. In conclusion, treatment with SphK inhibitor leads to both pro- and anti-atherogenic effects in LDL-R(-/-) mice. As a consequence, SphK inhibition fails to affect atherosclerosis despite significant S1P reduction in plasma.


Assuntos
Adamantano/análogos & derivados , Aterosclerose/tratamento farmacológico , Células Dendríticas/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Inibidores Enzimáticos/administração & dosagem , Piridinas/administração & dosagem , Linfócitos T/efeitos dos fármacos , Adamantano/administração & dosagem , Adamantano/efeitos adversos , Animais , Aterosclerose/sangue , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Modelos Animais de Doenças , Progressão da Doença , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Inibidores Enzimáticos/efeitos adversos , Humanos , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Knockout , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Pró-Proteína Convertases/sangue , Piridinas/efeitos adversos , Receptores de LDL/genética , Serina Endopeptidases/sangue , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia
14.
Circ Res ; 110(2): 200-10, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22116820

RESUMO

RATIONALE: Nuclear factor of activated T-cells (NFAT) is importantly implicated in pathological cardiac remodeling and vascular lesion formation. NFAT functionality is mainly regulated by calcineurin, a Ca(2+)-dependent multi-effector phosphatase. Calcineurin inhibitors such as cyclosporine A (CsA) were shown to be effective in the treatment of restenosis and vascular inflammation but with adverse side effects. OBJECTIVE: This prompted the design of more selective inhibitors such as VIVIT and inhibitors of NFAT-calcineurin association, which unfortunately have a poor potency precluding clinical use. METHODS AND RESULTS: Here, we describe the rational design of a potent bipartite inhibitor of NFAT-calcineurin interaction, MCV1, which targets two separate calcineurin docking motifs. Modeling, site-directed mutagenesis, and functional studies demonstrated that MCV1 acts by allosteric modulation of calcineurin. Comparable to CsA, MCV1 prevents NFAT activation at nanomolar potency without impairing calcineurin phosphatase activity, nuclear factor-κB nuclear import, and general cell signaling. In contrast, CsA but not MCV1-activated basal level extracellular signal-regulated kinases activity and prevented nuclear import of calcineurin, independent of NFAT activation. In vivo MCV1 abrogated NFAT-mediated T-cell activation in a model of PMA-elicited peritonitis, whereas topical application of MCV1 markedly reduced neointima formation in a mouse model of restenosis. CONCLUSIONS: We designed a bipartite NFAT inhibitor that is more potent than VIVIT and more selective than CsA. MCV1 constitutes not only a powerful tool to unravel NFAT function but also a potential drug candidate for the treatment of diseases implicating NFAT activation.


Assuntos
Lesões das Artérias Carótidas/tratamento farmacológico , Artéria Carótida Primitiva/efeitos dos fármacos , Estenose das Carótidas/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Imunossupressores/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Fatores de Transcrição NFATC/antagonistas & inibidores , Peptídeos/farmacologia , Peritonite/tratamento farmacológico , Linfócitos T/efeitos dos fármacos , Motivos de Aminoácidos , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Células COS , Calcineurina/metabolismo , Lesões das Artérias Carótidas/imunologia , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Artéria Carótida Primitiva/imunologia , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/patologia , Estenose das Carótidas/imunologia , Estenose das Carótidas/metabolismo , Estenose das Carótidas/patologia , Chlorocebus aethiops , Ciclosporina/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Desenho de Fármacos , Células HEK293 , Humanos , Hiperplasia , Imunossupressores/química , Células Jurkat , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Estrutura Molecular , Mutagênese Sítio-Dirigida , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Peptídeos/química , Peritonite/imunologia , Peritonite/metabolismo , Recidiva , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transfecção
15.
Arterioscler Thromb Vasc Biol ; 31(5): 1160-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21350196

RESUMO

OBJECTIVE: Apolipoprotein E (apoE) exerts potent antiinflammatory effects. Here, we investigated the effect of apoE on the functional phenotype of macrophages. METHODS AND RESULTS: Human apoE receptors very-low-density lipoprotein receptor (VLDL-R) and apoE receptor-2 (apoER2) were stably expressed in RAW264.7 mouse macrophages. In these cells, apoE downregulated markers of the proinflammatory M1 phenotype (inducible nitric oxide synthase, interleukin [IL]-12, macrophage inflammatory protein-1α) but upregulated markers of the antiinflammatory M2 phenotype (arginase I, SOCS3, IL-1 receptor antagonist [IL-1RA]). In addition, M1 macrophage responses (migration, generation of reactive oxygen species, antibody-dependent cell cytotoxicity, phagocytosis), as well as poly(I:C)- or interferon-γ-induced production of proinflammatory cytokines; cyclooxygenase-2 expression; and activation of nuclear factor-κB, IκB, and STAT1, were suppressed in VLDL-R- or apoER2-expressing cells. Conversely, the suppression of the M2 phenotype and the enhanced response to poly(I:C) were observed in apoE-producing bone marrow macrophages derived from VLDL-R-deficient mice but not wild-type or low-density lipoprotein receptor-deficient mice. The modulatory effects of apoE on macrophage polarization were inhibited in apoE receptor-expressing RAW264.7 cells exposed to SB220025, a p38 mitogen-activated protein kinase inhibitor, and PP1, a tyrosine kinase inhibitor. Accordingly, apoE induced tyrosine kinase-dependent activation of p38 mitogen-activated protein kinase in VLDL-R- or apoER2-expressing macrophages. Under in vivo conditions, apoE-/- mice transplanted with apoE-producing wild-type bone marrow showed increased plasma IL-1RA levels, and peritoneal macrophages of transplanted animals were shifted to the M2 phenotype (increased IL-1RA production and CD206 expression). CONCLUSIONS: ApoE signaling via VLDL-R or apoER2 promotes macrophage conversion from the proinflammatory M1 to the antiinflammatory M2 phenotype. This effect may represent a novel antiinflammatory activity of apoE.


Assuntos
Apolipoproteínas E/metabolismo , Inflamação/prevenção & controle , Macrófagos/metabolismo , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Transplante de Medula Óssea , Linhagem Celular , Feminino , Genótipo , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Interferon gama/metabolismo , Proteínas Relacionadas a Receptor de LDL/deficiência , Proteínas Relacionadas a Receptor de LDL/genética , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Poli I-C/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais , Fatores de Tempo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
J Exp Med ; 208(2): 217-25, 2011 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-21242297

RESUMO

Growth differentiation factor (GDF) 15 is a member of the transforming growth factor ß (TGF-ß) superfamily, which operates in acute phase responses through a currently unknown receptor. Elevated GDF-15 serum levels were recently identified as a risk factor for acute coronary syndromes. We show that GDF-15 expression is up-regulated as disease progresses in murine atherosclerosis and primarily colocalizes with plaque macrophages. Hematopoietic GDF-15 deficiency in low density lipoprotein receptor(-/-) mice led to impaired initial lesion formation and increased collagen in later lesions. Although lesion burden in GDF-15(-/-) chimeras was unaltered, plaques had reduced macrophage infiltrates and decreased necrotic core formation, all features of improved plaque stability. In vitro studies pointed to a TGFßRII-dependent regulatory role of GDF-15 in cell death regulation. Importantly, GDF-15(-/-) macrophages displayed reduced CCR2 expression, whereas GDF-15 promoted macrophage chemotaxis in a strictly CCR2- and TGFßRII-dependent manner, a phenomenon which was not observed in G protein-coupled receptor kinase 2(+/-) macrophages. In conclusion, GDF-15 deletion has a beneficial effect both in early and later atherosclerosis by inhibition of CCR2-mediated chemotaxis and by modulating cell death. Our study is the first to identify GDF-15 as an acute phase modifier of CCR2/TGFßRII-dependent inflammatory responses to vascular injury.


Assuntos
Apoptose/fisiologia , Aterosclerose/prevenção & controle , Quimiotaxia/fisiologia , Regulação da Expressão Gênica/fisiologia , Fator 15 de Diferenciação de Crescimento/deficiência , Macrófagos/fisiologia , Receptores CCR2/metabolismo , Animais , Transplante de Medula Óssea , Primers do DNA/genética , Citometria de Fluxo , Fator 15 de Diferenciação de Crescimento/sangue , Fator 15 de Diferenciação de Crescimento/genética , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Fagocitose/fisiologia , Receptores de LDL/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Cardiovasc Res ; 89(1): 244-52, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20693162

RESUMO

AIMS: mast cells have been shown to accumulate in the adventitia of human atherosclerotic plaques and were recently demonstrated by us to contribute to plaque progression and instability. In this study, we investigated whether selective inhibition of mast cell chymases would affect the lesion development and stability. METHODS AND RESULTS: the protease inhibitor RO5066852 appeared to be a potent inhibitor of chymase activity in vitro and ex vivo. With this inhibitor, we provide three lines of evidence that chymase inhibition can prevent many pro-atherogenic activities. First, oral administration of RO5066852 reduced spontaneous atherosclerosis in the thoracic aorta of apoE(-/-) mice. Second, chymase inhibition prevented the accelerated plaque progression observed in apoE(-/-) mice that were exposed to repetitive episodes of systemic mast cell activation. Furthermore, RO5066852 enhanced lesional collagen content and reduced necrotic core size. Third, RO5066852 treatment almost completely normalized the increased frequency and size of intraplaque haemorrhages observed in apoE(-/-) mice after acute perivascular mast cell activation in advanced atherosclerosis. CONCLUSION: our data indicate that chymase inhibition can inhibit pro-atherogenic and plaque destabilizing effects which are associated with perivascular mast cell activation. Our study thus identifies pharmacological chymase inhibition as a potential therapeutic modality for atherosclerotic plaque stabilization.


Assuntos
Apolipoproteínas E/deficiência , Quimases/antagonistas & inibidores , Ácidos Indolacéticos/farmacologia , Mastócitos/efeitos dos fármacos , Mastócitos/enzimologia , Naftalenos/farmacologia , Placa Aterosclerótica/prevenção & controle , Animais , Apolipoproteínas E/genética , Quimases/genética , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placa Aterosclerótica/tratamento farmacológico , Placa Aterosclerótica/enzimologia , Placa Aterosclerótica/patologia , Inibidores de Proteases/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética
18.
Am J Pathol ; 176(6): 3073-84, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20431029

RESUMO

Lysophosphatidic acid (LPA) accumulates in the central atheroma of human atherosclerotic plaques and is the primary platelet-activating lipid constituent of plaques. Here, we investigated the enzymatic regulation of LPA homeostasis in atherosclerotic lesions at various stages of disease progression. Atherosclerotic lesions were induced in carotid arteries of low-density lipoprotein receptor-deficient mice by semiconstrictive collar placement. At 2-week intervals after collar placement, lipids and RNA were extracted from the vessel segments carrying the plaque. Enzymatic-and liquid chromatography-mass spectrometry-based lipid profiling revealed progressive accumulation of LPA species in atherosclerotic tissue preceded by an increase in lysophosphatidylcholine, a precursor in LPA synthesis. Plaque expression of LPA-generating enzymes cytoplasmic phospholipase A(2)IVA (cPLA(2)IVA) and calcium-independent PLA(2)VIA (iPLA(2)VIA) was gradually increased, whereas that of the LPA-hydrolyzing enzyme LPA acyltransferase alpha was quenched. Increased expression of cPLA(2)IVA and iPLA(2)VIA in advanced lesions was confirmed by immunohistochemistry. Moreover, LPA receptors 1 and 2 were 50% decreased and sevenfold upregulated, respectively. Therefore, key proteins in LPA homeostasis are increasingly dysregulated in the plaque during atherogenesis, favoring intracellular LPA production. This might at least partly explain the observed progressive accumulation of this thrombogenic proinflammatory lipid in human and mouse plaques. Thus, intervention in the enzymatic LPA production may be an attractive measure to lower intraplaque LPA content, thereby reducing plaque progression and thrombogenicity.


Assuntos
Aterosclerose/patologia , Homeostase , Lisofosfolipídeos/metabolismo , Aciltransferases/metabolismo , Animais , Aterosclerose/metabolismo , Artérias Carótidas/patologia , Dieta , Progressão da Doença , Perfilação da Expressão Gênica , Fosfolipases A2 do Grupo VI/metabolismo , Humanos , Lisofosfolipídeos/química , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Receptores de LDL/genética , Receptores de LDL/metabolismo
19.
Circ Res ; 106(1): 89-92, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-19926877

RESUMO

RATIONALE: Although we and others have recently shown that mast cells play an important role in plaque progression and destabilization, the nature of the actual trigger for (peri)vascular mast cell activation during atherosclerosis is still unresolved. OBJECTIVE: In this study, we confirm that perivascular mast cell content correlates with the number of nerve fibers in the adventitia of human coronary atherosclerotic plaque specimen. Because peripheral C-type nerve fibers secrete, among others, substance P, a potent mast cell activator, we set out to study effects of adventitial administration of this neuropeptide on mast cell dependent destabilization of carotid artery plaques in apolipoprotein E-deficient (apoE(-/-)) mice. METHODS AND RESULTS: Substance P treatment significantly enhanced the number and activation status of adventitial mast cells compared to controls and promoted intraplaque hemorrhages. These phenomena could be prevented by coadministration of the neurokinin-1 receptor antagonist spantide I and did not occur in mast cell deficient apoE(-/-) mice, establishing the critical involvement of mast cells in substance P-elicited plaque destabilization. CONCLUSIONS: Our data suggest that neurotransmitters such as substance P are capable of promoting mast cell dependent plaque destabilization and provide a new, direct link between neural factors and vascular inflammation.


Assuntos
Tecido Conjuntivo/metabolismo , Doença da Artéria Coronariana/metabolismo , Hemorragia/metabolismo , Mastócitos/metabolismo , Neurotransmissores/metabolismo , Substância P/metabolismo , Idoso , Analgésicos/farmacologia , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Tecido Conjuntivo/patologia , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/patologia , Feminino , Hemorragia/genética , Hemorragia/patologia , Humanos , Masculino , Mastócitos/patologia , Camundongos , Camundongos Knockout , Antagonistas dos Receptores de Neurocinina-1 , Neurotransmissores/farmacologia , Receptores da Neurocinina-1/genética , Receptores da Neurocinina-1/metabolismo , Substância P/análogos & derivados , Substância P/farmacologia , Vasculite/genética , Vasculite/metabolismo , Vasculite/patologia
20.
Circulation ; 115(4): 501-8, 2007 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-17242282

RESUMO

BACKGROUND: Numerous in vitro studies suggest that sphingosine 1-phosphate (S1P), a bioactive lysosphingolipid associated with high-density lipoproteins, accounts at least partly for the potent antiinflammatory properties of high-density lipoprotein and, thereby, contributes to the antiatherogenic potential attributed to high-density lipoproteins. The present study was undertaken to investigate whether modulation of S1P signaling would affect atherosclerosis in a murine model of disease. METHODS AND RESULTS: Low-density lipoprotein receptor-deficient mice on a cholesterol-rich diet were given FTY720, a synthetic S1P analogue, at low (0.04 mg/kg per day) or high (0.4 mg/kg per day) doses for 16 weeks. FTY720 dose-dependently reduced atherosclerotic lesion formation, both in the aortic root and brachiocephalic artery, and almost completely blunted necrotic core formation. Plasma lipids remained unchanged during the course of FTY720 treatment. However, FTY720 lowered blood lymphocyte count (at a high dose) and significantly interfered with lymphocyte function, as evidenced by reduced splenocyte proliferation and interferon-gamma levels in plasma. Plasma concentrations of proinflammatory cytokines such as tumor necrosis factor-alpha, interleukin (IL)-6, IL-12, and regulated on activation normal T cell expressed and secreted were reduced by FTY720 administration. Moreover, lipopolysaccharide-elicited generation of nitrite/nitrate and IL-6--two markers of classical (M1) macrophage activation--was inhibited, whereas IL-4-induced production of IL-1-receptor antagonist, a marker of alternative (M2) macrophage activation, was augmented in peritoneal macrophages from FTY720-treated low-density lipoprotein receptor-deficient mice. CONCLUSIONS: The present results demonstrate that an S1P analogue inhibits atherosclerosis by modulating lymphocyte and macrophage function, and these results are consistent with the notion that S1P contributes to the antiatherogenic potential of high-density lipoprotein.


Assuntos
Aterosclerose/tratamento farmacológico , Imunossupressores/farmacologia , Propilenoglicóis/farmacologia , Receptores de LDL/genética , Esfingosina/análogos & derivados , Animais , Aterosclerose/imunologia , Aterosclerose/patologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/imunologia , HDL-Colesterol/sangue , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Cloridrato de Fingolimode , Contagem de Linfócitos , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/patologia , Lisofosfolipídeos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Esfingosina/metabolismo , Esfingosina/farmacologia , Triglicerídeos/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...