Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Intervalo de año de publicación
1.
Immunity ; 47(4): 621-634, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-29045897

RESUMEN

Atherosclerosis is an arterial disease process characterized by the focal subendothelial accumulation of apolipoprotein-B-containing lipoproteins, immune and vascular wall cells, and extracellular matrix. The lipoproteins acquire features of damage-associated molecular patterns and trigger first an innate immune response, dominated by monocyte-macrophages, and then an adaptive immune response. These inflammatory responses often become chronic and non-resolving and can lead to arterial damage and thrombosis-induced organ infarction. The innate immune response is regulated at various stages, from hematopoiesis to monocyte changes and macrophage activation. The adaptive immune response is regulated primarily by mechanisms that affect the balance between regulatory and effector T cells. Mechanisms related to cellular cholesterol, phenotypic plasticity, metabolism, and aging play key roles in affecting these responses. Herein, we review select topics that shed light on these processes and suggest new treatment strategies.


Asunto(s)
Aterosclerosis/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Linfocitos T/inmunología , Inmunidad Adaptativa/inmunología , Animales , Humanos , Inmunidad Innata/inmunología , Lipoproteínas/inmunología , Modelos Inmunológicos
2.
Trends Immunol ; 40(6): 511-523, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31053497

RESUMEN

Oncology has recently undergone a revolutionary change with widespread adoption of immunotherapy for many cancers. Immunotherapy using monoclonal antibodies against checkpoint molecules, including programmed death (PD)-1, PD ligand (PD-L)1, and cytotoxic T lymphocyte-associated antigen (CTLA)-4, is effective in a significant subset of patients. However, immune-related adverse events (irAEs) have emerged as frequent complications of checkpoint blockade, likely due to the physiological role of checkpoint pathways in regulating adaptive immunity and preventing autoimmunity. As immunotherapy becomes more common, a better understanding of the etiology of irAEs and ways to limit these events is needed. At the same time, studying these new therapy-related disorders provides an opportunity to better understand naturally occurring human autoimmune and inflammatory disorders, with the potential to improve therapies for cancer and autoimmune diseases.


Asunto(s)
Antineoplásicos Inmunológicos/efectos adversos , Inmunoterapia/efectos adversos , Neoplasias/terapia , Animales , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/uso terapéutico , Autoinmunidad , Biomarcadores de Tumor , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Humanos , Inmunomodulación/efectos de los fármacos , Inmunoterapia/métodos , Neoplasias/etiología , Neoplasias/metabolismo , Neoplasias/patología , Evaluación del Resultado de la Atención al Paciente , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
3.
Immunity ; 38(6): 1092-104, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23809160

RESUMEN

According to the traditional view, atherosclerosis results from a passive buildup of cholesterol in the artery wall. Yet, burgeoning evidence implicates inflammation and immune effector mechanisms in the pathogenesis of this disease. Both innate and adaptive immunity operate during atherogenesis and link many traditional risk factors to altered arterial functions. Inflammatory pathways have become targets in the quest for novel preventive and therapeutic strategies against cardiovascular disease, a growing contributor to morbidity and mortality worldwide. Here we review current experimental and clinical knowledge of the pathogenesis of atherosclerosis through an immunological lens and how host defense mechanisms essential for survival of the species actually contribute to this chronic disease but also present new opportunities for its mitigation.


Asunto(s)
Arterias/metabolismo , Aterosclerosis/inmunología , Colesterol/metabolismo , Inmunidad Adaptativa , Animales , Aterosclerosis/terapia , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Inflamación/terapia , Ratones , Terapia Molecular Dirigida
4.
Arterioscler Thromb Vasc Biol ; 39(8): 1588-1601, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31294624

RESUMEN

OBJECTIVE: MR (mineralocorticoid receptor) activation is associated with cardiovascular ischemia in humans. This study explores the role of the MR in atherosclerotic mice of both sexes and identifies a sex-specific role for endothelial cell (EC)-MR in vascular inflammation. Approach and Results: In the AAV-PCSK9 (adeno-associated virus-proprotein convertase subtilisin/kexin type 9) mouse atherosclerosis model, MR inhibition attenuated vascular inflammation in males but not females. Further studies comparing male and female littermates with intact MR or EC-MR deletion revealed that although EC-MR deletion did not affect plaque size in either sex, it reduced aortic arch inflammation specifically in male mice as measured by flow cytometry. Moreover, MR-intact females had larger plaques but were protected from vascular inflammation compared with males. Intravital microscopy of the mesenteric vasculature demonstrated that EC-MR deletion attenuated TNFα (tumor necrosis factor α)-induced leukocyte slow rolling and adhesion in males, while females exhibited fewer leukocyte-endothelial interactions with no additional effect of EC-MR deletion. These effects corresponded with decreased TNFα-induced expression of the endothelial adhesion molecules ICAM-1 (intercellular adhesion molecule-1) and E-selectin in males with EC-MR deletion compared with MR-intact males and females of both genotypes. These observations were also consistent with MR and estrogen regulation of ICAM-1 transcription and E-selectin expression in primary cultured mouse ECs and human umbilical vein ECs. CONCLUSIONS: In male mice, EC-MR deletion attenuates leukocyte-endothelial interactions, plaque inflammation, and expression of E-selectin and ICAM-1, providing a potential mechanism by which the MR promotes vascular inflammation. In females, plaque inflammation and leukocyte-endothelial interactions are decreased relative to males and EC-MR deletion is not protective.


Asunto(s)
Aterosclerosis/complicaciones , Células Endoteliales/fisiología , Receptores de Mineralocorticoides/fisiología , Vasculitis/etiología , Animales , Células Cultivadas , Selectina E/genética , Femenino , Molécula 1 de Adhesión Intercelular/genética , Leucocitos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Caracteres Sexuales
5.
J Immunol ; 200(11): 3697-3710, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29669783

RESUMEN

Vasoactive intestinal peptide (VIP) is a neuropeptide that exerts various vascular and cardioprotective functions and regulates immune function and inflammatory response at multiple levels. However, its role in inflammatory cardiovascular disorders is largely unknown. Myocarditis and atherosclerosis are two inflammatory and autoimmune cardiovascular diseases that cause important adverse circulatory events. In this study, we investigate the therapeutic effects of VIP in various well-established preclinical models of experimental autoimmune myocarditis and atherosclerosis. Intraperitoneal injection of VIP during the effector phase of experimental autoimmune myocarditis in susceptible BALB/c mice significantly reduced its prevalence, ameliorated signs of heart hypertrophy and injury, attenuated myocardial inflammatory infiltration, and avoided subsequent profibrotic cardiac remodeling. This effect was accompanied by a reduction of Th17-driven cardiomyogenic responses in peripheral lymphoid organs and in the levels of myocardial autoantibodies. In contrast, acute and chronic atherosclerosis was induced in apolipoprotein E-deficient mice fed a hyperlipidemic diet and subjected to partial carotid ligation. Systemic VIP treatment reduced the number and size of atherosclerotic plaques in carotid, aorta, and sinus in hypercholesterolemic mice. VIP reduced Th1-driven inflammatory responses and increased regulatory T cells in atherosclerotic arteries and their draining lymph nodes. VIP also regulated cholesterol efflux in macrophages and reduced the formation of foam cells and their presence in atherosclerotic plaques. Finally, VIP inhibited proliferation and migration of smooth muscle cells and neointima formation in a mouse model of complete carotid ligation. These findings encourage further studies aimed to assess whether VIP can be used as a pharmaceutical agent to treat heart inflammation and atherosclerosis.


Asunto(s)
Aterosclerosis/inmunología , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Inflamación/inmunología , Miocarditis/inmunología , Péptido Intestinal Vasoactivo/inmunología , Animales , Apolipoproteínas E/inmunología , Autoanticuerpos/inmunología , Modelos Animales de Enfermedad , Femenino , Ganglios Linfáticos/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Músculo Liso/inmunología , Miocardio/inmunología , Neuropéptidos/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología
6.
N Engl J Med ; 375(18): 1749-1755, 2016 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-27806233

RESUMEN

Immune checkpoint inhibitors have improved clinical outcomes associated with numerous cancers, but high-grade, immune-related adverse events can occur, particularly with combination immunotherapy. We report the cases of two patients with melanoma in whom fatal myocarditis developed after treatment with ipilimumab and nivolumab. In both patients, there was development of myositis with rhabdomyolysis, early progressive and refractory cardiac electrical instability, and myocarditis with a robust presence of T-cell and macrophage infiltrates. Selective clonal T-cell populations infiltrating the myocardium were identical to those present in tumors and skeletal muscle. Pharmacovigilance studies show that myocarditis occurred in 0.27% of patients treated with a combination of ipilimumab and nivolumab, which suggests that our patients were having a rare, potentially fatal, T-cell-driven drug reaction. (Funded by Vanderbilt-Ingram Cancer Center Ambassadors and others.).


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Inmunoterapia/efectos adversos , Miocarditis/etiología , Miocardio/patología , Anciano , Anticuerpos Monoclonales/uso terapéutico , Arritmias Cardíacas/inducido químicamente , Electrocardiografía/efectos de los fármacos , Resultado Fatal , Femenino , Glucocorticoides/uso terapéutico , Bloqueo Cardíaco/diagnóstico , Bloqueo Cardíaco/etiología , Humanos , Ipilimumab , Masculino , Melanoma/complicaciones , Melanoma/tratamiento farmacológico , Persona de Mediana Edad , Miocarditis/tratamiento farmacológico , Miocarditis/patología , Miositis/inducido químicamente , Nivolumab
7.
J Immunol ; 197(12): 4651-4662, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27837103

RESUMEN

Dendritic cells (DCs) have been implicated as important regulators of innate and adaptive inflammation in many diseases, including atherosclerosis. However, the molecular mechanisms by which DCs mitigate or promote inflammatory pathogenesis are only partially understood. Previous studies have shown an important anti-inflammatory role for the transcription factor Krüppel-like factor 2 (KLF2) in regulating activation of various cell types that participate in atherosclerotic lesion development, including endothelial cells, macrophages, and T cells. We used a pan-DC, CD11c-specific cre-lox gene knockout mouse model to assess the role of KLF2 in DC activation, function, and control of inflammation in the context of hypercholesterolemia and atherosclerosis. We found that KLF2 deficiency enhanced surface expression of costimulatory molecules CD40 and CD86 in DCs and promoted increased T cell proliferation and apoptosis. Transplant of bone marrow from mice with KLF2-deficient DCs into Ldlr-/- mice aggravated atherosclerosis compared with control mice, most likely due to heightened vascular inflammation evidenced by increased DC presence within lesions, enhanced T cell activation and cytokine production, and increased cell death in atherosclerotic lesions. Taken together, these data indicate that KLF2 governs the degree of DC activation and hence the intensity of proatherogenic T cell responses.


Asunto(s)
Aterosclerosis/inmunología , Células de la Médula Ósea/fisiología , Células Dendríticas/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Linfocitos T/inmunología , Animales , Trasplante de Médula Ósea , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inmunidad Celular , Factores de Transcripción de Tipo Kruppel/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL/genética
8.
Circ Res ; 117(6): 502-512, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26199323

RESUMEN

RATIONALE: The development of molecular imaging approaches that assess specific immunopathologic mechanisms can advance the study of myocarditis. OBJECTIVE: This study validates a novel molecular imaging tool that enables the in vivo visualization of granzyme B activity, a major effector of cytotoxic CD8+ T lymphocytes. METHODS AND RESULTS: We synthesized and optimized a fluorogenic substrate capable of reporting on granzyme B activity and examined its specificity ex vivo in mice hearts with experimental cytotoxic CD8+ T lymphocyte-mediated myocarditis using fluorescence reflectance imaging, validated by histological examination. In vivo experiments localized granzyme B activity in hearts with acute myocarditis monitored by fluorescent molecular tomography in conjunction with coregistered computed tomography imaging. A model anti-inflammatory intervention (dexamethasone administration) in vivo reduced granzyme B activity (vehicle versus dexamethasone: 504±263 versus 194±77 fluorescence intensities in hearts; P=0.002). CONCLUSIONS: Molecular imaging of granzyme B activity can visualize T cell-mediated myocardial injury and monitor the response to an anti-inflammatory intervention.


Asunto(s)
Granzimas/metabolismo , Miocarditis/enzimología , Miocarditis/inmunología , Animales , Linfocitos T CD8-positivos/enzimología , Linfocitos T CD8-positivos/inmunología , Activación Enzimática/fisiología , Colorantes Fluorescentes/análisis , Granzimas/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocarditis/patología
9.
Arterioscler Thromb Vasc Biol ; 36(3): 456-65, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26821944

RESUMEN

OBJECTIVE: T cell immunoglobulin and mucin domain (Tim) proteins are expressed by numerous immune cells, recognize phosphatidylserine on apoptotic cells, and function as costimulators or coinhibitors. Tim-1 is expressed by activated T cells but is also found on dendritic cells and B cells. Tim-4, present on macrophages and dendritic cells, plays a critical role in apoptotic cell clearance, regulates the number of phosphatidylserine-expressing activated T cells, and is genetically associated with low low-density lipoprotein and triglyceride levels. Because these functions of Tim-1 and Tim-4 could affect atherosclerosis, their modulation has potential therapeutic value in cardiovascular disease. APPROACH AND RESULTS: ldlr(-/-) mice were fed a high-fat diet for 4 weeks while being treated with control (rat immunoglobulin G1) or anti-Tim-1 (3D10) or -Tim-4 (21H12) monoclonal antibodies that block phosphatidylserine recognition and phagocytosis. Both anti-Tim-1 and anti-Tim-4 treatments enhanced atherosclerosis by 45% compared with controls by impairment of efferocytosis and increasing aortic CD4(+)T cells. Consistently, anti-Tim-4-treated mice showed increased percentages of activated T cells and late apoptotic cells in the circulation. Moreover, in vitro blockade of Tim-4 inhibited efferocytosis of oxidized low-density lipoprotein-induced apoptotic macrophages. Although anti-Tim-4 treatment increased T helper cell (Th)1 and Th2 responses, anti-Tim-1 induced Th2 responses but dramatically reduced the percentage of regulatory T cells. Finally, combined blockade of Tim-1 and Tim-4 increased atherosclerotic lesion size by 59%. CONCLUSIONS: Blockade of Tim-4 aggravates atherosclerosis likely by prevention of phagocytosis of phosphatidylserine-expressing apoptotic cells and activated T cells by Tim-4-expressing cells, whereas Tim-1-associated effects on atherosclerosis are related to changes in Th1/Th2 balance and reduced circulating regulatory T cells.


Asunto(s)
Anticuerpos Monoclonales/toxicidad , Enfermedades de la Aorta/inducido químicamente , Aterosclerosis/inducido químicamente , Linfocitos T CD4-Positivos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Proteínas de la Membrana/antagonistas & inhibidores , Receptores de LDL/deficiencia , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apoptosis/efectos de los fármacos , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Receptor Celular 1 del Virus de la Hepatitis A , Lipoproteínas LDL/metabolismo , Activación de Linfocitos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Masculino , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones Noqueados , Fagocitosis/efectos de los fármacos , Placa Aterosclerótica , Receptores de LDL/genética , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th2/efectos de los fármacos , Células Th2/inmunología
10.
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
11.
Arterioscler Thromb Vasc Biol ; 35(2): 280-7, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25414253

RESUMEN

Regulatory T cells (Tregs) play an important role in the regulation of T-cell-mediated immune responses through suppression of T-cell proliferation and secretion of inhibitory cytokines, such as interleukin-10 and transforming growth factor-ß. Impaired Treg numbers and function have been associated with numerous diseases, and an imbalance between proinflammatory/proatherogenic cells and Tregs promotes atherosclerotic disease. Restoration of this balance by inducing Tregs has great therapeutic potential to prevent cardiovascular disease. In addition to suppressing differentiation and function of effector T cells, Tregs have been shown to induce anti-inflammatory macrophages, inhibit foam cell formation and to influence cholesterol metabolism. Furthermore, Tregs suppress immune responses of endothelial cells and innate lymphoid cells. In this review, we focus on the recent knowledge on Treg subsets, their activity and function in atherosclerosis, and discuss promising strategies to use Tregs as a therapeutic tool to prevent cardiovascular disease.


Asunto(s)
Traslado Adoptivo , Aterosclerosis/terapia , Inmunidad Celular/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Inflamación/terapia , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/trasplante , Animales , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Activación de Linfocitos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Fenotipo , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 35(12): 2526-35, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26494229

RESUMEN

OBJECTIVE: Innate lymphoid cells (ILCs) are a newly discovered subset of immune cells that promote tissue homeostasis and protect against pathogens. ILCs produce cytokines also produced by T lymphocytes that have been shown to affect atherosclerosis, but the influence of ILCs on atherosclerosis has not been explored. APPROACH AND RESULTS: We demonstrate that CD25(+) ILCs that produce type 2 cytokines (ILC2s) are present in the aorta of atherosclerotic immunodeficient ldlr(-/-)rag1(-/-) mice. To investigate the role of ILCs in atherosclerosis, ldlr(-/-)rag1(-/-) mice were concurrently fed an atherogenic diet and treated with either ILC-depleting anti-CD90.2 antibodies or IL-2/anti-IL-2 complexes that expand CD25(+) ILCs. Lesion development was not affected by anti-CD90.2 treatment, but was reduced in IL-2/anti-IL-2-treated mice. These IL-2-treated mice had reduced very low-density lipoprotein cholesterol and increased triglycerides compared with controls and reduced apolipoprotein B100 gene expression in the liver. IL-2/anti-IL-2 treatment caused expansion of ILC2s in aorta and other tissues, elevated levels of IL-5, systemic eosinophila, and hepatic eosinophilic inflammation. Blockade of IL-5 reversed the IL-2 complex-induced eosinophilia but did not change lesion size. CONCLUSIONS: This study demonstrates that expansion of CD25-expressing ILCs by IL-2/anti-IL-2 complexes leads to a reduction in very low-density lipoprotein cholesterol and atherosclerosis. Global depletion of ILCs by anti-CD90.2 did not significantly affect lesion size indicating that different ILC subsets may have divergent effects on atherosclerosis.


Asunto(s)
Anticuerpos/farmacología , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Proliferación Celular/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Interleucina-2/farmacología , Linfocitos/efectos de los fármacos , Animales , Aorta/efectos de los fármacos , Aorta/inmunología , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteína B-100 , Apolipoproteínas B/sangre , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Biomarcadores/sangre , VLDL-Colesterol/sangre , Modelos Animales de Enfermedad , Eosinofilia/inmunología , Eosinofilia/metabolismo , Eosinofilia/prevención & control , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Subunidad alfa del Receptor de Interleucina-2/inmunología , Interleucina-5/antagonistas & inhibidores , Interleucina-5/sangre , Interleucina-5/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL/deficiencia , Receptores de LDL/genética , Triglicéridos/sangre
13.
J Am Soc Nephrol ; 26(12): 3102-13, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25855773

RESUMEN

Progress in long-term renal allograft survival continues to lag behind the progress in short-term transplant outcomes. Dendritic cells are the most efficient antigen-presenting cells, but surprisingly little attention has been paid to their presence in transplanted kidneys. We used dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin as a marker of dendritic cells in 105 allograft biopsy samples from 105 kidney transplant recipients. High dendritic cell density was associated with poor allograft survival independent of clinical variables. Moreover, high dendritic cell density correlated with greater T cell proliferation and poor outcomes in patients with high total inflammation scores, including inflammation in areas of tubular atrophy. We then explored the association between dendritic cells and histologic variables associated with poor prognosis. Multivariate analysis revealed an independent association between the densities of dendritic cells and T cells. In biopsy samples with high dendritic cell density, electron microscopy showed direct physical contact between infiltrating lymphocytes and cells that have the ultrastructural morphologic characteristics of dendritic cells. The origin of graft dendritic cells was sought in nine sex-mismatched recipients using XY fluorescence in situ hybridization. Whereas donor dendritic cells predominated initially, the majority of dendritic cells in late allograft biopsy samples were of recipient origin. Our data highlight the prognostic value of dendritic cell density in allograft biopsy samples, suggest a new role for these cells in shaping graft inflammation, and provide a rationale for targeting dendritic cell recruitment to promote long-term allograft survival.


Asunto(s)
Aloinjertos/patología , Moléculas de Adhesión Celular/análisis , Células Dendríticas/química , Supervivencia de Injerto , Trasplante de Riñón , Riñón/patología , Lectinas Tipo C/análisis , Receptores de Superficie Celular/análisis , Adulto , Aloinjertos/inmunología , Biopsia , Células Dendríticas/patología , Células Dendríticas/ultraestructura , Femenino , Supervivencia de Injerto/inmunología , Humanos , Riñón/inmunología , Masculino , Microscopía Electrónica , Persona de Mediana Edad , Nefritis/patología , Valor Predictivo de las Pruebas , Linfocitos T/patología , Linfocitos T/ultraestructura
14.
Am J Pathol ; 182(1): 5-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23159526

RESUMEN

Chronic inflammation driven by immune responses to lipid deposition in the arterial wall is now understood to be fundamental to the pathogenesis of atherosclerosis. The frequent presence of T lymphocytes in human atherosclerotic lesions was first described in the 1980s, but experiments to test whether adaptive immunity influences lesion development and phenotype required animal models. The American journal of pathology has published many research articles focused on the role of inflammation and adaptive immunity in diet-induced and genetically manipulated murine models of atherosclerosis. Seminal articles in the 1990s were the first to describe the presence of T cells in mouse atherosclerotic lesions; other articles demonstrated the effects of defective adaptive immunity on lesion development in mice.


Asunto(s)
Aterosclerosis/inmunología , Modelos Animales de Enfermedad , Inmunidad Adaptativa , Animales , Aterosclerosis/etiología , Antígenos B7/inmunología , Citocinas/inmunología , Humanos , Hipercolesterolemia/complicaciones , Tolerancia Inmunológica , Ratones , Ratones Noqueados , Linfocitos T Colaboradores-Inductores/inmunología
15.
J Immunol ; 188(10): 4876-84, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22491251

RESUMEN

PD-1, a member of the CD28 family of immune regulatory molecules, is expressed on activated T cells, interacts with its ligands, PD-L1/B7-H1 and PD-L2/B7-DC, on other cells, and delivers inhibitory signals to the T cell. We studied the role of this pathway in modulating autoreactive T cell responses in two models of myocarditis. In a CD8(+) T cell-mediated adoptive transfer model, we found that compared with Pd1(+/+) CD8(+) T cells, Pd1(-/-) CD8(+) T cells cause enhanced disease, with increased inflammatory infiltrate, particularly rich in neutrophils. Additionally, we show enhanced proliferation in vivo and enhanced cytotoxic activity of PD-1-deficient T lymphocytes against myocardial endothelial cells in vitro. In experimental autoimmune myocarditis, a disease model dependent on CD4(+) T cells, we show that mice lacking PD-1 develop enhanced disease compared with wild-type mice. PD-1-deficient mice displayed increased inflammation, enhanced serum markers of myocardial damage, and an increased infiltration of inflammatory cells, including CD8(+) T cells. Together, these studies show that PD-1 plays an important role in limiting T cell responses in the heart.


Asunto(s)
Células Musculares/inmunología , Células Musculares/patología , Miocarditis/inmunología , Miocarditis/patología , Receptor de Muerte Celular Programada 1/fisiología , Subgrupos de Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Movimiento Celular/genética , Movimiento Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Inflamación/genética , Inflamación/patología , Inflamación/prevención & control , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Miocarditis/genética , Receptor de Muerte Celular Programada 1/deficiencia , Receptor de Muerte Celular Programada 1/genética , Subgrupos de Linfocitos T/patología
16.
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
17.
J Immunol ; 189(7): 3714-23, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22933624

RESUMEN

Systemic lupus erythematosus (SLE) is a chronic, multiorgan inflammatory autoimmune disorder associated with high levels of circulating autoantibodies and immune complexes. We report that passive transfer of human SLE sera into mice expressing the uniquely human FcγRIIA and FcγRIIIB on neutrophils induces lupus nephritis and in some cases arthritis only when the mice additionally lack the CD18 integrin, Mac-1. The prevailing view is that Mac-1 on macrophages is responsible for immune complex clearance. However, disease permitted by the absence of Mac-1 is not related to enhanced renal immune complex deposition or in situ C1q/C3 complement activation and proceeds even in the absence of macrophages. Instead, disease is associated with increased FcγRIIA-induced neutrophil accumulation that is enabled by Mac-1 deficiency. Intravital microscopy in the cremasteric vasculature reveals that Mac-1 mitigates FcγRIIA-dependent neutrophil recruitment in response to deposited immune complexes. Our results provide direct evidence that human SLE immune complexes are pathogenic, demonstrate that neutrophils are primary mediators of end organ damage in a novel humanized lupus mouse model, and identify Mac-1 regulation of FcγRIIA-mediated neutrophil recruitment as a key step in development of target organ damage.


Asunto(s)
Antígenos CD18/genética , Riñón/patología , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Antígeno de Macrófago-1/genética , Neutrófilos/inmunología , Neutrófilos/patología , Suero/inmunología , Animales , Antígenos CD18/metabolismo , Humanos , Pruebas Intradérmicas , Células K562 , Riñón/inmunología , Lupus Eritematoso Sistémico/patología , Antígeno de Macrófago-1/fisiología , Ratones , Ratones Noqueados , Conejos , Receptores de IgG/genética , Receptores de IgG/fisiología
18.
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
19.
J Autoimmun ; 45: 90-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23810579

RESUMEN

Myocardial inflammation and damage can lead to lethal acute or chronic cardiac failure. A variety of regulatory mechanisms limit the magnitude and duration of T cell responses in the heart. Insights into these regulatory mechanisms have come from studies of specific deficiencies in central or peripheral T cell tolerance which cause or enhance the severity of myocarditis. Under non-inflammatory conditions, constitutive DC presentation of cardiac peptides to naïve T cells in cardiac draining lymph nodes tolerizes recirculating naïve T cells specific for these antigens. Cardiac antigen-specific naïve T cells, especially those specific of α-myosin heavy chain peptides, become activated and differentiate into expanded clones of effector T cells under various conditions, such as cardiac infection and/or genetic variations in peripheral tolerance. The pathology that these effector cells cause in the myocardium is limited by PD-L1 expressed on myocardial cells in response to inflammatory cytokines, and by CTLA-4 dependent mechanisms. The PD-1:PD-L1 pathway works together with other control mechanisms to keep the heart safe from T cells, and combined impairment of this pathway along with other regulatory mechanisms synergize to cause myocarditis. T cell derived IFNγ contributes to the inflammatory damage to the heart in autoimmune myocarditis, but it also engages regulatory mechanisms that limit disease, including upregulation of PD-L1, and differentiation of TNF and iNOS expressing DCs from monocytes. iNOS derived from these DCs and other IFNγ stimulated cells inhibits expansion of T cells that cause myocarditis. Regulatory T cells also appear to be critical for suppression of effector T cells specific for myocardial antigens.


Asunto(s)
Insuficiencia Cardíaca/inmunología , Interferón gamma/inmunología , Miocarditis/inmunología , Linfocitos T/inmunología , Animales , Autoantígenos/inmunología , Antígeno B7-H1/metabolismo , Miosinas Cardíacas/inmunología , Humanos , Tolerancia Inmunológica , Ratones , Óxido Nítrico Sintasa de Tipo II/metabolismo , Transducción de Señal
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA