Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
1.
Immunity ; 47(4): 621-634, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-29045897

RESUMO

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.


Assuntos
Aterosclerose/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Linfócitos T/imunologia , Imunidade Adaptativa/imunologia , Animais , Humanos , Imunidade Inata/imunologia , Lipoproteínas/imunologia , Modelos Imunológicos
2.
Trends Immunol ; 40(6): 511-523, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31053497

RESUMO

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.


Assuntos
Antineoplásicos Imunológicos/efeitos adversos , Imunoterapia/efeitos adversos , Neoplasias/terapia , Animais , Antineoplásicos Imunológicos/administração & dosagem , Antineoplásicos Imunológicos/uso terapêutico , Autoimunidade , Biomarcadores Tumorais , Gerenciamento Clínico , Suscetibilidade a Doenças , Humanos , Imunomodulação/efeitos dos fármacos , Imunoterapia/métodos , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Avaliação de Resultados da Assistência ao Paciente , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
3.
Immunity ; 38(6): 1092-104, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23809160

RESUMO

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.


Assuntos
Artérias/metabolismo , Aterosclerose/imunologia , Colesterol/metabolismo , Imunidade Adaptativa , Animais , Aterosclerose/terapia , Modelos Animais de Doenças , Humanos , Imunidade Inata , Inflamação/terapia , Camundongos , Terapia de Alvo Molecular
4.
Mol Cell ; 56(2): 219-231, 2014 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-25263595

RESUMO

Proinflammatory stimuli elicit rapid transcriptional responses via transduced signals to master regulatory transcription factors. To explore the role of chromatin-dependent signal transduction in the atherogenic inflammatory response, we characterized the dynamics, structure, and function of regulatory elements in the activated endothelial cell epigenome. Stimulation with tumor necrosis factor alpha prompted a dramatic and rapid global redistribution of chromatin activators to massive de novo clustered enhancer domains. Inflammatory super enhancers formed by nuclear factor-kappa B accumulate at the expense of immediately decommissioned, basal endothelial super enhancers, despite persistent histone hyperacetylation. Mass action of enhancer factor redistribution causes momentous swings in transcriptional initiation and elongation. A chemical genetic approach reveals a requirement for BET bromodomains in communicating enhancer remodeling to RNA Polymerase II and orchestrating the transition to the inflammatory cell state, demonstrated in activated endothelium and macrophages. BET bromodomain inhibition abrogates super enhancer-mediated inflammatory transcription, atherogenic endothelial responses, and atherosclerosis in vivo.


Assuntos
Aterosclerose/genética , Inflamação/genética , Subunidade p50 de NF-kappa B/imunologia , Proteínas Nucleares/antagonistas & inibidores , Fator de Transcrição RelA/imunologia , Fatores de Transcrição/antagonistas & inibidores , Acetilação , Animais , Aterosclerose/imunologia , Azepinas/farmacologia , Adesão Celular/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Cromatina/genética , Selectina E/biossíntese , Células Endoteliais , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Elementos Facilitadores Genéticos , Histonas/metabolismo , Humanos , Inflamação/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subunidade p50 de NF-kappa B/genética , Proteínas Nucleares/imunologia , Ligação Proteica , RNA Polimerase II/genética , Sequências Reguladoras de Ácido Nucleico , Fatores de Transcrição SOXF/genética , Transdução de Sinais , Fator de Transcrição RelA/genética , Fatores de Transcrição/imunologia , Iniciação da Transcrição Genética , Transcrição Gênica/efeitos dos fármacos , Triazóis/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/biossíntese
5.
Circulation ; 140(2): 80-91, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31390169

RESUMO

Recent developments in cancer therapeutics have improved outcomes but have also been associated with cardiovascular complications. Therapies harnessing the immune system have been associated with an immune-mediated myocardial injury described as myocarditis. Immune checkpoint inhibitors are one such therapy with an increasing number of case and cohort reports describing a clinical syndrome of immune checkpoint inhibitor­associated myocarditis. Although the full spectrum of immune checkpoint inhibitor­associated cardiovascular disease still needs to be fully defined, described cases of myocarditis range from syndromes with mild signs and symptoms to fatal events. These observations in the clinical setting stand in contrast to outcomes from randomized clinical trials in which myocarditis is a rare event that is investigator reported and lacking in a specific case definition. The complexities associated with diagnosis, as well as the heterogeneous clinical presentation of immune checkpoint inhibitor­associated myocarditis, have made ascertainment and identification of myocarditis with high specificity challenging in clinical trials and other data sets, limiting the ability to better understand the incidence, outcomes, and predictors of these rare events. Therefore, establishing a uniform definition of myocarditis for application in clinical trials of cancer immunotherapies will enable greater understanding of these events. We propose an operational definition of cancer therapy-associated myocarditis that may facilitate case ascertainment and report and therefore may enhance the understanding of the incidence, outcomes, and risk factors of this novel clinical syndrome.


Assuntos
Cardiologia/tendências , Oncologia/tendências , Miocardite/terapia , Neoplasias/terapia , Antineoplásicos Imunológicos/uso terapêutico , Cardiologia/métodos , Ensaios Clínicos como Assunto/métodos , Humanos , Imunoterapia/métodos , Imunoterapia/tendências , Oncologia/métodos , Miocardite/epidemiologia , Miocardite/imunologia , Neoplasias/epidemiologia , Neoplasias/imunologia
6.
Circ Res ; 123(1): 33-42, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29572206

RESUMO

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


Assuntos
Armadilhas Extracelulares/fisiologia , Hidrolases/fisiologia , Placa Aterosclerótica/etiologia , Trombose/etiologia , Animais , Transplante de Medula Óssea , Doenças das Artérias Carótidas/etiologia , Doenças das Artérias Carótidas/patologia , Morte Celular , Desoxirribonuclease I/farmacologia , Armadilhas Extracelulares/efeitos dos fármacos , Humanos , Hidrolases/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/fisiologia , Osteomielite/etiologia , Placa Aterosclerótica/patologia , Proteína-Arginina Desiminase do Tipo 4 , Trombose/prevenção & controle , Túnica Íntima/lesões
7.
Arterioscler Thromb Vasc Biol ; 39(8): 1588-1601, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31294624

RESUMO

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.


Assuntos
Aterosclerose/complicações , Células Endoteliais/fisiologia , Receptores de Mineralocorticoides/fisiologia , Vasculite/etiologia , Animais , Células Cultivadas , Selectina E/genética , Feminino , Molécula 1 de Adesão Intercelular/genética , Leucócitos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Caracteres Sexuais
8.
J Immunol ; 200(11): 3697-3710, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29669783

RESUMO

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.


Assuntos
Aterosclerose/imunologia , Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Inflamação/imunologia , Miocardite/imunologia , Peptídeo Intestinal Vasoativo/imunologia , Animais , Apolipoproteínas E/imunologia , Autoanticorpos/imunologia , Modelos Animais de Doenças , Feminino , Linfonodos/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Músculo Liso/imunologia , Miocárdio/imunologia , Neuropeptídeos/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia
9.
N Engl J Med ; 375(18): 1749-1755, 2016 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-27806233

RESUMO

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.).


Assuntos
Anticorpos Monoclonais/efeitos adversos , Imunoterapia/efeitos adversos , Miocardite/etiologia , Miocárdio/patologia , Idoso , Anticorpos Monoclonais/uso terapêutico , Arritmias Cardíacas/induzido quimicamente , Eletrocardiografia/efeitos dos fármacos , Evolução Fatal , Feminino , Glucocorticoides/uso terapêutico , Bloqueio Cardíaco/diagnóstico , Bloqueio Cardíaco/etiologia , Humanos , Ipilimumab , Masculino , Melanoma/complicações , Melanoma/tratamento farmacológico , Pessoa de Meia-Idade , Miocardite/tratamento farmacológico , Miocardite/patologia , Miosite/induzido quimicamente , Nivolumabe
10.
J Immunol ; 197(12): 4651-4662, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27837103

RESUMO

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.


Assuntos
Aterosclerose/imunologia , Células da Medula Óssea/fisiologia , Células Dendríticas/fisiologia , Fatores de Transcrição Kruppel-Like/metabolismo , Linfócitos T/imunologia , Animais , Transplante de Medula Óssea , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Imunidade Celular , Fatores de Transcrição Kruppel-Like/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de LDL/genética
11.
Circ Res ; 117(6): 502-512, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26199323

RESUMO

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.


Assuntos
Granzimas/metabolismo , Miocardite/enzimologia , Miocardite/imunologia , Animais , Linfócitos T CD8-Positivos/enzimologia , Linfócitos T CD8-Positivos/imunologia , Ativação Enzimática/fisiologia , Corantes Fluorescentes/análise , Granzimas/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocardite/patologia
12.
Arterioscler Thromb Vasc Biol ; 36(3): 456-65, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26821944

RESUMO

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.


Assuntos
Anticorpos Monoclonais/toxicidade , Doenças da Aorta/induzido quimicamente , Aterosclerose/induzido quimicamente , Linfócitos T CD4-Positivos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Proteínas de Membrana/antagonistas & inibidores , Receptores de LDL/deficiência , Animais , Doenças da Aorta/genética , Doenças da Aorta/imunologia , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apoptose/efeitos dos fármacos , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Receptor Celular 1 do Vírus da Hepatite A , Lipoproteínas LDL/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos Knockout , Fagocitose/efeitos dos fármacos , Placa Aterosclerótica , Receptores de LDL/genética , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th2/efeitos dos fármacos , Células Th2/imunologia
13.
J Immunol ; 194(4): 1467-79, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25595782

RESUMO

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.


Assuntos
Aterosclerose/etiologia , Fatores Reguladores de Interferon/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Síndrome Metabólica/etiologia , Animais , Aterosclerose/imunologia , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Fatores Reguladores de Interferon/deficiência , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/patologia , Masculino , Síndrome Metabólica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Arterioscler Thromb Vasc Biol ; 35(2): 280-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25414253

RESUMO

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.


Assuntos
Transferência Adotiva , Aterosclerose/terapia , Imunidade Celular/efeitos dos fármacos , Fatores Imunológicos/uso terapêutico , Inflamação/terapia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/transplante , Animais , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Fenótipo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 35(12): 2526-35, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26494229

RESUMO

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.


Assuntos
Anticorpos/farmacologia , Doenças da Aorta/prevenção & controle , Aterosclerose/prevenção & controle , Proliferação de Células/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Interleucina-2/farmacologia , Linfócitos/efeitos dos fármacos , Animais , Aorta/efeitos dos fármacos , Aorta/imunologia , Aorta/metabolismo , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/imunologia , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apolipoproteína B-100 , Apolipoproteínas B/sangue , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Biomarcadores/sangue , VLDL-Colesterol/sangue , Modelos Animais de Doenças , Eosinofilia/imunologia , Eosinofilia/metabolismo , Eosinofilia/prevenção & controle , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Subunidade alfa de Receptor de Interleucina-2/imunologia , Interleucina-5/antagonistas & inibidores , Interleucina-5/sangue , Interleucina-5/imunologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de LDL/deficiência , Receptores de LDL/genética , Triglicerídeos/sangue
16.
J Am Soc Nephrol ; 26(12): 3102-13, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25855773

RESUMO

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.


Assuntos
Aloenxertos/patologia , Moléculas de Adesão Celular/análise , Células Dendríticas/química , Sobrevivência de Enxerto , Transplante de Rim , Rim/patologia , Lectinas Tipo C/análise , Receptores de Superfície Celular/análise , Adulto , Aloenxertos/imunologia , Biópsia , Células Dendríticas/patologia , Células Dendríticas/ultraestrutura , Feminino , Sobrevivência de Enxerto/imunologia , Humanos , Rim/imunologia , Masculino , Microscopia Eletrônica , Pessoa de Meia-Idade , Nefrite/patologia , Valor Preditivo dos Testes , Linfócitos T/patologia , Linfócitos T/ultraestrutura
17.
Am J Pathol ; 182(1): 5-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23159526

RESUMO

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.


Assuntos
Aterosclerose/imunologia , Modelos Animais de Doenças , Imunidade Adaptativa , Animais , Aterosclerose/etiologia , Antígenos B7/imunologia , Citocinas/imunologia , Humanos , Hipercolesterolemia/complicações , Tolerância Imunológica , Camundongos , Camundongos Knockout , Linfócitos T Auxiliares-Indutores/imunologia
18.
J Immunol ; 188(10): 4876-84, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22491251

RESUMO

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.


Assuntos
Células Musculares/imunologia , Células Musculares/patologia , Miocardite/imunologia , Miocardite/patologia , Receptor de Morte Celular Programada 1/fisiologia , Subpopulações de Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Feminino , Inflamação/genética , Inflamação/patologia , Inflamação/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Miocardite/genética , Receptor de Morte Celular Programada 1/deficiência , Receptor de Morte Celular Programada 1/genética , Subpopulações de Linfócitos T/patologia
19.
J Immunol ; 188(3): 1421-30, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22219321

RESUMO

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.


Assuntos
Adesão Celular/imunologia , Endotélio Vascular/imunologia , Células Th1/fisiologia , Animais , Quimiocina CCL20/metabolismo , Selectina E/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiologia , Molécula 1 de Adesão Intercelular/imunologia , Camundongos , Células Th17
20.
J Immunol ; 189(7): 3714-23, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22933624

RESUMO

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.


Assuntos
Antígenos CD18/genética , Rim/patologia , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/imunologia , Antígeno de Macrófago 1/genética , Neutrófilos/imunologia , Neutrófilos/patologia , Soro/imunologia , Animais , Antígenos CD18/metabolismo , Humanos , Testes Intradérmicos , Células K562 , Rim/imunologia , Lúpus Eritematoso Sistêmico/patologia , Antígeno de Macrófago 1/fisiologia , Camundongos , Camundongos Knockout , Coelhos , Receptores de IgG/genética , Receptores de IgG/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa