Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 13: 910112, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35837407

RESUMEN

Signaling lymphocytic activation molecule family 8 (SLAMF8) is involved in the negative modulation of NADPH oxidase activation. However, the impact of SLAMF8 downregulation on macrophage functionality and the microbicide mechanism remains elusive. To study this in depth, we first analyzed NADPH oxidase activation pathways in wild-type and SLAMF8-deficient macrophages upon different stimulus. Herein, we describe increased phosphorylation of the Erk1/2 and p38 MAP kinases, as well as increased phosphorylation of NADPH oxidase subunits in SLAMF8-deficient macrophages. Furthermore, using specific inhibitors, we observed that specific PI3K inhibition decreased the differences observed between wild-type and SLAMF8-deficient macrophages, stimulated with either PMA, LPS, or Salmonella typhimurium infection. Consequently, SLAMF8-deficient macrophages also showed increased recruitment of small GTPases such as Rab5 and Rab7, and the p47phox subunit to cytoplasmic Salmonella, suggesting an impairment of Salmonella-containing vacuole (SCV) progression in SLAMF8-deficient macrophages. Enhanced iNOS activation, NO production, and IL-6 expression were also observed in the absence of SLAMF8 upon Salmonella infection, either in vivo or in vitro, while overexpression of SLAMF8 in RAW264.7 macrophages showed the opposite phenotype. In addition, SLAMF8-deficient macrophages showed increased activation of Src kinases and reduced SHP-1 phosphate levels upon IFNγ and Salmonella stimuli in comparison to wild-type macrophages. In agreement with in vitro results, Salmonella clearance was augmented in SLAMF8-deficient mice compared to that in wild-type mice. Therefore, in conclusion, SLAMF8 intervention upon bacterial infection downregulates mouse macrophage activation, and confirmed that SLAMF8 receptor could be a potential therapeutic target for the treatment of severe or unresolved inflammatory conditions.


Asunto(s)
Antiinfecciosos , Proteínas de la Membrana/metabolismo , Infecciones por Salmonella , Animales , Antiinfecciosos/metabolismo , Macrófagos/metabolismo , Ratones , NADPH Oxidasas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Infecciones por Salmonella/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética
2.
Sci Rep ; 7: 46444, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28406244

RESUMEN

Atherosclerosis is a chronic inflammatory cardiovascular disease that is responsible of high mortality worldwide. Evidence indicates that maladaptive autoimmune responses in the arterial wall play critical roles in the process of atherosclerosis. Cortistatin is a neuropeptide expressed in the vascular system and atherosclerotic plaques that regulates vascular calcification and neointimal formation, and inhibits inflammation in different experimental models of autoimmune diseases. Its role in inflammatory cardiovascular disorders is largely unexplored. The aim of this study is to investigate the potential therapeutic effects of cortistatin in two well-established preclinical models of atherosclerosis, and the molecular and cellular mechanisms involved. Systemic treatment with cortistatin reduced the number and size of atherosclerotic plaques in carotid artery, heart, aortic arch and aorta in acute and chronic atherosclerosis induced in apolipoprotein E-deficient mice fed a high-lipid diet. This effect was exerted at multiple levels. Cortistatin reduced Th1/Th17-driven inflammatory responses and increased regulatory T cells in atherosclerotic arteries and lymphoid organs. Moreover, cortistatin reduced the capacity of endothelial cells to bind and recruit immune cells to the plaque and impaired the formation of foam cells by enhancing cholesterol efflux from macrophages. Cortistatin emerges as a new candidate for the treatment of the clinical manifestations of atherosclerosis.


Asunto(s)
Antiinflamatorios/administración & dosificación , Células Espumosas/efectos de los fármacos , Hiperlipidemias/complicaciones , Neuropéptidos/administración & dosificación , Placa Aterosclerótica/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Células Espumosas/metabolismo , Hiperlipidemias/genética , Ratones , Ratones Noqueados para ApoE , Neuropéptidos/farmacología , Placa Aterosclerótica/genética , Placa Aterosclerótica/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología
3.
Br J Pharmacol ; 174(3): 267-280, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27922195

RESUMEN

BACKGROUND AND PURPOSE: Myocarditis is an inflammatory and autoimmune cardiovascular disease that causes dilated myocardiopathy and is responsible for high morbidity and mortality worldwide. Cortistatin is a neuropeptide produced by neurons and cells of the immune and vascular systems. Besides its action in locomotor activity and sleep, cortistatin inhibits inflammation in different experimental models of autoimmune diseases. However, its role in inflammatory cardiovascular disorders is unexplored. Here, we investigated the therapeutic effects of cortistatin in a well-established preclinical model of experimental autoimmune myocarditis (EAM). EXPERIMENTAL APPROACH: We induced EAM by immunization with a fragment of cardiac myosin in susceptible Balb/c mice. Cortistatin was administered i.p. starting 7, 11 or 15 days after EAM induction. At day 21, we evaluated heart hypertrophy, myocardial injury, cardiac inflammatory infiltration and levels of serum and cardiac inflammatory cytokines, cortistatin and autoantibodies. We determined proliferation and cytokine production by heart draining lymph node cells in response to cardiac myosin restimulation. KEY RESULTS: Systemic injection of cortistatin during the effector phase of the disease significantly reduced its prevalence and signs of heart hypertrophy and injury (decreased the levels of brain natriuretic peptide) and impaired myocardial inflammatory cell infiltration. This effect was accompanied by a reduction in self-antigen-specific T-cell responses in lymph nodes and in the levels of cardiomyogenic antibodies and inflammatory cytokines in serum and myocardium. Finally, we found a positive correlation between cardiac and systemic cortistatin levels and EAM severity. CONCLUSIONS AND IMPLICATIONS: Cortistatin emerges as a new candidate to treat inflammatory dilated cardiomyopathy.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Cardiomiopatía Dilatada/tratamiento farmacológico , Miocarditis/tratamiento farmacológico , Neuropéptidos/farmacología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/fisiopatología , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Dilatada/fisiopatología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/fisiopatología , Ganglios Linfáticos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Miocarditis/inmunología , Miocarditis/fisiopatología , Neuropéptidos/administración & dosificación , Índice de Severidad de la Enfermedad , Linfocitos T/inmunología , Factores de Tiempo
4.
J Immunol ; 194(4): 1467-79, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25595782

RESUMEN

Premature atherosclerosis is a severe complication of lupus and other systemic autoimmune disorders. Gain-of-function polymorphisms in IFN regulatory factor 5 (IRF5) are associated with an increased risk of developing lupus, and IRF5 deficiency in lupus mouse models ameliorates disease. However, whether IRF5 deficiency also protects against atherosclerosis development in lupus is not known. In this study, we addressed this question using the gld.apoE(-/-) mouse model. IRF5 deficiency markedly reduced lupus disease severity. Unexpectedly, despite the reduction in systemic immune activation, IRF5-deficient mice developed increased atherosclerosis and also exhibited metabolic dysregulation characterized by hyperlipidemia, increased adiposity, and insulin resistance. Levels of the atheroprotective cytokine IL-10 were reduced in aortae of IRF5-deficient mice, and in vitro studies demonstrated that IRF5 is required for IL-10 production downstream of TLR7 and TLR9 signaling in multiple immune cell types. Chimera studies showed that IRF5 deficiency in bone marrow-derived cells prevents lupus development and contributes in part to the increased atherosclerosis. Notably, IRF5 deficiency in non-bone marrow-derived cells also contributes to the increased atherosclerosis through the generation of hyperlipidemia and increased adiposity. Together, our results reveal a protective role for IRF5 in lupus-associated atherosclerosis that is mediated through the effects of IRF5 in both immune and nonimmune cells. These findings have implications for the proposed targeting of IRF5 in the treatment of autoimmune disease as global IRF5 inhibition may exacerbate cardiovascular disease in these patients.


Asunto(s)
Aterosclerosis/etiología , Factores Reguladores del Interferón/inmunología , Lupus Eritematoso Sistémico/inmunología , Síndrome Metabólico/etiología , Animales , Aterosclerosis/inmunología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Factores Reguladores del Interferón/deficiencia , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/patología , Masculino , Síndrome Metabólico/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
PLoS Pathog ; 8(7): e1002799, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22807679

RESUMEN

Trypanosoma cruzi, the protozoan parasite responsible for Chagas' disease, causes severe myocarditis often resulting in death. Here, we report that Slamf1-/- mice, which lack the hematopoietic cell surface receptor Slamf1, are completely protected from an acute lethal parasite challenge. Cardiac damage was reduced in Slamf1-/- mice compared to wild type mice, infected with the same doses of parasites, as a result of a decrease of the number of parasites in the heart even the parasitemia was only marginally less. Both in vivo and in vitro experiments reveal that Slamf1-defIcient myeloid cells are impaired in their ability to replicate the parasite and show altered production of cytokines. Importantly, IFN-γ production in the heart of Slamf1 deficient mice was much lower than in the heart of wt mice even though the number of infiltrating dendritic cells, macrophages, CD4 and CD8 T lymphocytes were comparable. Administration of an anti-Slamf1 monoclonal antibody also reduced the number of parasites and IFN-γ in the heart. These observations not only explain the reduced susceptibility to in vivo infection by the parasite, but they also suggest human Slamf1 as a potential target for therapeutic target against T. cruzi infection.


Asunto(s)
Antígenos CD/fisiología , Cardiomiopatía Chagásica/parasitología , Enfermedad de Chagas/parasitología , Células Mieloides/metabolismo , Receptores de Superficie Celular/fisiología , Trypanosoma cruzi/fisiología , Animales , Anticuerpos Monoclonales , Anticuerpos Antiprotozoarios/biosíntesis , Antígenos CD/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Cardiomiopatía Chagásica/inmunología , Enfermedad de Chagas/inmunología , Citocinas/biosíntesis , Células Dendríticas/inmunología , Susceptibilidad a Enfermedades , Corazón/parasitología , Interferón gamma/biosíntesis , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células Mieloides/parasitología , Miocardio/metabolismo , Parasitemia , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Trypanosoma cruzi/inmunología
6.
J Immunol ; 188(12): 5829-32, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22593622

RESUMEN

Slamf8 (CD353) is a cell surface receptor that is expressed upon activation of macrophages (MΦs) by IFN-γ or bacteria. In this article, we report that a very high NADPH oxidase (Nox2) enzyme activity was found in Slamf8(-/-) MΦs in response to Escherichia coli or Staphylococcus aureus, as well as to PMA. The elevated Nox2 activity in Slamf8(-/-) MΦs was also demonstrated in E. coli or S. aureus phagosomes by using a pH indicator system and was further confirmed by a reduction in the enzyme activity after transfection of the receptor into Slamf8-deficient primary MΦs or RAW 264.7 cells. Upon exposure to bacteria or PMA, protein kinase C activity in Slamf8(-/-) MΦs is increased. This results in an enhanced phosphorylation of p40phox, one key component of the Nox2 enzyme complex, which, in turn, leads to greater Nox2 activity. Taken together, the data show that, in response to inflammation-associated stimuli, the inducible receptor Slamf8 negatively regulates inflammatory responses.


Asunto(s)
Antígenos CD/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Antígenos CD/inmunología , Western Blotting , Línea Celular , Regulación de la Expresión Génica/inmunología , Inflamación/inmunología , Inflamación/metabolismo , Macrófagos/inmunología , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , NADPH Oxidasa 2 , NADPH Oxidasas/inmunología , Fagosomas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Superficie Celular/inmunología , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Regulación hacia Arriba
7.
J Immunol ; 188(12): 6287-99, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22566565

RESUMEN

IL-17A (IL-17) is the signature cytokine produced by Th17 cells and has been implicated in host defense against infection and the pathophysiology of autoimmunity and cardiovascular disease. Little is known, however, about the influence of IL-17 on endothelial activation and leukocyte influx to sites of inflammation. We hypothesized that IL-17 would induce a distinct pattern of endothelial activation and leukocyte recruitment when compared with the Th1 cytokine IFN-γ. We found that IL-17 alone had minimal activating effects on cultured endothelium, whereas the combination of TNF-α and IL-17 produced a synergistic increase in the expression of both P-selectin and E-selectin. Using intravital microscopy of the mouse cremaster muscle, we found that TNF-α and IL-17 also led to a synergistic increase in E-selectin-dependent leukocyte rolling on microvascular endothelium in vivo. In addition, TNF-α and IL-17 enhanced endothelial expression of the neutrophilic chemokines CXCL1, CXCL2, and CXCL5 and led to a functional increase in leukocyte transmigration in vivo and CXCR2-dependent neutrophil but not T cell transmigration in a parallel-plate flow chamber system. By contrast, endothelial activation with TNF-α and IFN-γ preferentially induced the expression of the integrin ligands ICAM-1 and VCAM-1, as well as the T cell chemokines CXCL9, CXCL10, and CCL5. These effects were further associated with a functional increase in T cell but not neutrophil transmigration under laminar shear flow. Overall, these data show that IL-17 and TNF-α act in a synergistic manner to induce a distinct pattern of endothelial activation that sustains and enhances neutrophil influx to sites of inflammation.


Asunto(s)
Células Endoteliales/metabolismo , Inflamación/metabolismo , Interleucina-17/metabolismo , Infiltración Neutrófila/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Quimiocinas/biosíntesis , Células Endoteliales/inmunología , Citometría de Flujo , Inflamación/inmunología , Interleucina-17/inmunología , Rodamiento de Leucocito/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/inmunología
8.
J Immunol ; 188(3): 1421-30, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22219321

RESUMEN

T cell subset-specific migration to inflammatory sites is tightly regulated and involves interaction of the T cells with the endothelium. Th17 cells often appear at different inflammatory sites than Th1 cells, or both subsets appear at the same sites but at different times. Differences in T cell subset adhesion to endothelium may contribute to subset-specific migratory behavior, but this possibility has not been well studied. We examined the adhesion of mouse Th17 cells to endothelial adhesion molecules and endothelium under flow in vitro and to microvessels in vivo and we characterized their migratory phenotype by flow cytometry and quantitative RT-PCR. More Th17 than Th1 cells interacted with E-selectin. Fewer Th17 than Th1 cells bound to TNF-α-activated E-selectin-deficient endothelial cells, and intravital microscopy studies demonstrated that Th17 cells engage in more rolling interactions with TNF-α-treated microvessels than Th1 cells in wild-type mice but not in E-selectin-deficient mice. Th17 adhesion to ICAM-1 was dependent on integrin activation by CCL20, the ligand for CCR6, which is highly expressed by Th17 cells. In an air pouch model of inflammation, CCL20 triggered recruitment of Th17 but not Th1 cells. These data provide evidence that E-selectin- and ICAM-1-dependent adhesion of Th17 and Th1 cells with endothelium are quantitatively different.


Asunto(s)
Adhesión Celular/inmunología , Endotelio Vascular/inmunología , Células TH1/fisiología , Animales , Quimiocina CCL20/metabolismo , Selectina E/inmunología , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiología , Molécula 1 de Adhesión Intercelular/inmunología , Ratones , Células Th17
9.
J Immunol ; 187(7): 3521-9, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21873519

RESUMEN

The ability of regulatory T cells (Treg) to traffic to sites of inflammation supports their role in controlling immune responses. This feature supports the idea that adoptive transfer of in vitro expanded human Treg could be used for treatment of immune/inflammatory diseases. However, the migratory behavior of Treg, as well as their direct influence at the site of inflammation, remains poorly understood. To explore the possibility that Treg may have direct anti-inflammatory influences on tissues, independent of their well-established suppressive effects on lymphocytes, we studied the adhesive interactions between mouse Treg and endothelial cells, as well as their influence on endothelial function during acute inflammation. We show that Foxp3(+) adaptive/inducible Treg (iTreg), but not naturally occurring Treg, efficiently interact with endothelial selectins and transmigrate through endothelial monolayers in vitro. In response to activation by endothelial Ag presentation or immobilized anti-CD3ε, Foxp3(+) iTreg suppressed TNF-α- and IL-1ß-mediated endothelial selectin expression and adhesiveness to effector T cells. This suppression was contact independent, rapid acting, and mediated by TGF-ß-induced activin receptor-like kinase 5 signaling in endothelial cells. In addition, Foxp3(+) iTreg adhered to inflamed endothelium in vivo, and their secretion products blocked acute inflammation in a model of peritonitis. These data support the concept that Foxp3(+) iTreg help to regulate inflammation independently of their influence on effector T cells by direct suppression of endothelial activation and leukocyte recruitment.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Endotelio Vascular/inmunología , Inflamación/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Animales , Adhesión Celular/inmunología , Separación Celular , Endotelio Vascular/metabolismo , Citometría de Flujo , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Técnicas de Sustitución del Gen , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/metabolismo
10.
Circulation ; 124(2): 185-95, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21690490

RESUMEN

BACKGROUND: Regulatory T cells (Treg) are present in atherosclerotic lesions and can modulate disease. In this study we characterized changes in Treg responses associated with prolonged hypercholesterolemia and lesion progression. METHODS AND RESULTS: Low-density lipoprotein receptor null mice in which Treg express green fluorescent protein were fed a control or cholesterol-rich diet, and green fluorescent protein-positive cells were enumerated in lymphoid tissues and in aorta. Splenic Treg numbers increased after 4, 8, and 20 weeks in cholesterol-diet-fed mice. However, the number of circulating and lesional Treg peaked at 4 weeks and decreased significantly at 8 and 20 weeks, concomitant with increased numbers of CD4(+) effector T cells and increased lesion size over this period. Treg expression of selectin ligands and their ability to bind to aortic endothelium decreased after prolonged hypercholesterolemia, and apoptosis of lesional Treg increased. After 4 weeks of cholesterol-rich diet, a switch to a control diet for 4 weeks reduced serum cholesterol and stopped lesion growth, and the high aortic Treg content was maintained, compared with mice fed a cholesterol diet for 8 weeks. After the diet reversal, the splenic Treg retained the phenotype of Treg after 4 weeks of cholesterol diet. CONCLUSIONS: Prolonged hypercholesterolemia impairs Treg but not effector T cell accumulation in lesions, but reversal of hypercholesterolemia can prevent loss of lesional Treg. Therefore, cholesterol-lowering therapies may induce dynamic and beneficial changes in Treg:effector T cell ratios in atherosclerotic lesions.


Asunto(s)
Colesterol en la Dieta/sangre , Colesterol/sangre , Dieta Aterogénica , Endotelio Vascular/metabolismo , Hipercolesterolemia/sangre , Linfocitos T Reguladores/metabolismo , Animales , Aterosclerosis/sangre , Aterosclerosis/inmunología , Aterosclerosis/terapia , Recuento de Linfocito CD4 , Colesterol/efectos adversos , Colesterol/farmacología , Colesterol en la Dieta/efectos adversos , Colesterol en la Dieta/farmacología , Hipercolesterolemia/genética , Hipercolesterolemia/inmunología , Hipercolesterolemia/terapia , Ratones , Ratones Noqueados , Receptores de LDL/genética , Receptores de LDL/metabolismo , Linfocitos T Reguladores/inmunología
11.
Arterioscler Thromb Vasc Biol ; 31(5): 1100-7, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21393583

RESUMEN

OBJECTIVE: Programmed cell death-1 (PD-1) is a member of the CD28 superfamily that delivers negative signals on interaction with its 2 ligands, PD-L1 and PD-L2. We studied the contribution of the PD-1 pathway to regulation of T cells that promote atherosclerotic lesion formation and inflammation. METHODS AND RESULTS: We show that compared with Ldlr-/- control mice, Pd1-/-Ldlr-/- mice developed larger lesions with more abundant CD4+ and CD8+ T cells and macrophages, accompanied by higher levels of serum tumor necrosis factor-α. Iliac lymph node T cells from Pd1-/-Ldlr-/- mice proliferated more to αCD3 or oxidized low-density lipoprotein stimulation compared with controls. CD8+ T cells from Pd1-/-Ldlr-/- mice displayed more cytotoxic activity compared with controls in vivo and in vitro. Administration of a blocking anti-PD-1 antibody increased lesional inflammation in hypercholesterolemic Ldlr-/- mice with more lesional T cells and more activated T cells in paraaortic lymph nodes. The changes in lesional T-cell content when PD-1 was absent or blocked were also observed in bone marrow chimeric Ldlr-/- mice lacking PD-L1 and PD-L2 on hematopoietic cells. CONCLUSIONS: PD-1 has an important role in downregulating proatherogenic T-cell responses, and blockade of this molecule for treatment of viral infections or cancer may increase risk of cardiovascular complications.


Asunto(s)
Antígenos de Superficie/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Aterosclerosis/metabolismo , Inflamación/metabolismo , Activación de Linfocitos , Subgrupos de Linfocitos T/metabolismo , Animales , Anticuerpos Bloqueadores/farmacología , Antígenos de Superficie/genética , Apoptosis , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Antígeno B7-H1 , Trasplante de Médula Ósea , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Genes Codificadores de los Receptores de Linfocitos T , Genotipo , Hipercolesterolemia/genética , Hipercolesterolemia/inmunología , Hipercolesterolemia/metabolismo , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Lipoproteínas LDL/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Activación de Linfocitos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/deficiencia , Péptidos/genética , Fenotipo , Proteína 2 Ligando de Muerte Celular Programada 1 , Receptor de Muerte Celular Programada 1 , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transducción de Señal
12.
J Clin Invest ; 120(6): 1961-70, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20440076

RESUMEN

The transcription factor Krüppel-like factor 2 (KLF2) is required for the quiescent and migratory properties of naive T cells. Statins, a class of HMG-CoA reductase inhibitors, display pleiotropic immunomodulatory effects that are independent of their lipid-lowering capacity and may be beneficial as therapeutic agents for T cell-mediated inflammatory diseases. Statins upregulate KLF2 expression in endothelial cells, and this activity is associated with an antiinflammatory phenotype. We therefore hypothesized that the immunomodulatory effects of statins are due, in part, to their direct effects on T cell KLF2 gene expression. Here we report that lipophilic statin treatment of mouse and human T cells increased expression of KLF2 through a HMG-CoA/prenylation-dependent pathway. Statins also diminished T cell proliferation and IFN-gamma expression. shRNA blockade of KLF2 expression in human T cells increased IFN-gamma expression and prevented statin-induced IFN-gamma reduction. In a mouse model of myocarditis induced by heart antigen-specific CD8+ T cells, both statin treatment of the T cells and retrovirally mediated overexpression of KLF2 in the T cells had similar ameliorating effects on disease induction. We conclude that statins reduce inflammatory functions and pathogenic activity of T cells through KLF2-dependent mechanisms, and this pathway may be a potential therapeutic target for cardiovascular diseases.


Asunto(s)
Expresión Génica/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Factores de Transcripción de Tipo Kruppel/metabolismo , Linfocitos T/metabolismo , Factores de Transcripción/metabolismo , Acilcoenzima A , Animales , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/inmunología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Prenilación , Linfocitos T/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología
13.
Circ Res ; 103(9): 965-73, 2008 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-18832748

RESUMEN

Recent reports suggest dyslipidemia impairs dendritic cell (DC) function and adaptive immunity. This study aimed to characterize the effect of hypercholesterolemia on antigen-presenting cell function of DCs and DC-dependent CD4(+) T-cell responses. DCs incubated in vitro with acetylated low-density lipoprotein cholesterol with or without an acyl-coenzyme A:cholesterol acyl-transferase inhibitor maintained their ability to prime CD4(+) T cells. Analysis of T-cell proliferation and interferon-gamma and tumor necrosis factor-alpha production after ex vivo coculture of naïve CD4(+) T cells with splenic, inguinal, or iliac DCs from low-density lipoprotein receptor-deficient (LDLR(-/-)) or apolipoprotein E-deficient (ApoE(-/-)) mice fed an atherogenic diet highlighted DC efficacy in effector T-cell generation under hypercholesterolemic conditions. Adoptive transfer of carboxyfluorescein diacetate, succinimidyl ester (CFSE)-labeled naïve CD4(+) T cells in LDLR(-/-) recipients and subsequent immunization demonstrated effective priming of naïve T cells in hypercholesterolemic mice. CFSE dilution analyses revealed that hypercholesterolemic DCs were equipotent in naïve CD4(+) T-cell priming efficacy with normocholesterolemic DCs. Quantitative real-time PCR and flow cytometric analyses demonstrated that DC expression of multiple molecules involved in antigen processing, presentation, and T-cell stimulation remained unaltered by dyslipidemia. Finally, endogenous antigen-primed CD4(+) T cells responded equivalently to a secondary ex vivo antigenic challenge, regardless of whether they were primed in vivo under hypercholesterolemic or control conditions, demonstrating that all essential steps in CD4(+) T-cell responses remain intact under atherogenic conditions. This study affirms that the adaptive immune response prevails under the hypercholesterolemic conditions present in atherosclerosis. In particular, DCs remain functional antigen-presenting cells and maintain their ability to prime CD4(+) T cells even when cholesterol-loaded.


Asunto(s)
Presentación de Antígeno , Aterosclerosis/inmunología , Antígeno CD11c/metabolismo , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Hipercolesterolemia/inmunología , Activación de Linfocitos , Traslado Adoptivo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Linfocitos T CD4-Positivos/trasplante , Proliferación Celular , Células Cultivadas , Colesterol en la Dieta/sangre , LDL-Colesterol/metabolismo , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Hipercolesterolemia/complicaciones , Hipercolesterolemia/etiología , Interferón gamma/metabolismo , Lipoproteínas LDL/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Factor de Necrosis Tumoral alfa/metabolismo
14.
Traffic ; 9(8): 1299-315, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18445119

RESUMEN

Trypanosoma cruzi can infect and replicate in macrophages. During invasion, T. cruzi interacts with different macrophage receptors to induce its own phagocytosis. However, the nature of those receptors and the molecular mechanisms involved are poorly understood. In this study, we demonstrate that T. cruzi metacyclic trypomastigotes but not epimastigotes were able to induce Rab5 activation and binding to the early endosomes in peritoneal macrophages. In this process, active Rab5 colocalized with parasites in the phagosome and with the Rab5A effector molecule early endosomal antigen 1. Phagosome formation and T. cruzi internalization were inhibited in Raw 264.7 macrophages expressing a dominant-negative form of Rab5 [(S34N)Rab5]. Using T. cruzi membrane extracts, we verified that the Rab5 activation depends on the interaction between parasite surface molecules and macrophages surface molecule. In addition, during infection of macrophages, phosphatidylinositol 3-kinase (PI3K) pathway was activated. Assays carried out using a selective PI3K inhibitor (LY294002) showed that the PI3K activation is essential for Rab5 activation by T. cruzi infection and for the entrance and intracellular replication of T. cruzi in macrophages. Moreover, using macrophages from knockout mice, we found that activation of Rab5, fusion of early endosomes and phagocytosis induced by T. cruzi infection involved Toll-like receptor (TLR)2 but were independent of TLR4 receptors.


Asunto(s)
Receptor Toll-Like 2/química , Trypanosoma cruzi/metabolismo , Tripanosomiasis/metabolismo , Tripanosomiasis/parasitología , Proteínas de Unión al GTP rab5/química , Animales , Células Cultivadas , Endosomas/metabolismo , Femenino , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Microscopía Fluorescente , Fagosomas/metabolismo , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...