Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
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
2.
Clin Immunol ; 212: 108361, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32058071

RESUMO

Until recently, autoimmune disease research has primarily been focused on elucidating the role of the adaptive immune system. In the past decade or so, the role of the innate immune system in the pathogenesis of autoimmunity has increasingly been realized. Recent findings have elucidated paradigm-shifting concepts, for example, the implications of "trained immunity" and a dysbiotic microbiome in the susceptibility of predisposed individuals to clinical autoimmunity. In addition, the application of modern technologies such as the quantum dot (Qdot) system and 'Omics' (e.g., genomics, proteomics, and metabolomics) data-processing tools has proven fruitful in revisiting mechanisms underlying autoimmune pathogenesis and in identifying novel therapeutic targets. This review highlights recent findings discussed at the American Autoimmune Related Disease Association (AARDA) 2019 colloquium. The findings covering autoimmune diseases and autoinflammatory diseases illustrate how new developments in common innate immune pathways can contribute to the better understanding and management of these immune-mediated disorders.


Assuntos
Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Imunidade Inata/imunologia , Memória Imunológica/imunologia , Inflamação/imunologia , Alarminas/imunologia , Animais , Disbiose/imunologia , Doenças Hereditárias Autoinflamatórias/imunologia , Humanos , Microbiota/imunologia , Moléculas com Motivos Associados a Patógenos/imunologia
3.
Clin Immunol ; 181: 1-8, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28531376

RESUMO

On September 20, 2016 the American Autoimmune Related Disease Association convened a selected group of 16 experts in Washington DC to discuss current issues and future directions of research in autoimmune diseases from the cell signaling perspective. The major topics considered included new findings on the follicular T cell cells in mice, their metabolic patterns and their control by the newly discovered T follicular regulatory cells. Current work on the metabolic pathways deemed important for the function of T cells was presented. The broader implications of these studies on our understanding of human and murine T regulatory cell and current aspects of B cell biology, innate immunity and organ damage were discussed. This document should provide the reader with a quick update on these topics and help instigate novel studies.


Assuntos
Doenças Autoimunes/imunologia , Linfócitos B/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T/metabolismo , Animais , Congressos como Assunto , Humanos , Imunidade Inata/imunologia , Tecido Linfoide/citologia , Redes e Vias Metabólicas , Camundongos , Transdução de Sinais
4.
Clin Immunol ; 175: 26-33, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27894980

RESUMO

Despite of a multitude of excellent studies, the regulatory role of natural killer (NK) cells in the pathogenesis of inflammatory cardiac disease is greatly underappreciated. Clinical abnormalities in the numbers and functions of NK cells are observed in myocarditis and inflammatory dilated cardiomyopathy (DCMi) as well as in cardiac transplant rejection [1-6]. Because treatment of these disorders remains largely symptomatic in nature, patients have little options for targeted therapies [7,8]. However, blockade of NK cells and their receptors can protect against inflammation and damage in animal models of cardiac injury and inflammation. In these models, NK cells suppress the maturation and trafficking of inflammatory cells, alter the local cytokine and chemokine environments, and induce apoptosis in nearby resident and hematopoietic cells [1,9,10]. This review will dissect each protective mechanism employed by NK cells and explore how their properties might be exploited for their therapeutic potential.


Assuntos
Cardiopatias/imunologia , Inflamação/imunologia , Células Matadoras Naturais/imunologia , Animais , Cardiomiopatia Dilatada/imunologia , Citocinas/imunologia , Coração/fisiopatologia , Humanos , Miocardite/imunologia
5.
Eur J Immunol ; 46(3): 582-92, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26660726

RESUMO

Using a mouse model of experimental autoimmune myocarditis (EAM), we showed for the first time that IL-23 stimulation of CD4(+) T cells is required only briefly at the initiation of GM-CFS-dependent cardiac autoimmunity. IL-23 signal, acting as a switch, turns on pathogenicity of CD4(+) T cells, and becomes dispensable once autoreactivity is established. Il23a(-/-) mice failed to mount an efficient Th17 response to immunization, and were protected from myocarditis. However, remarkably, transient IL-23 stimulation ex vivo fully restored pathogenicity in otherwise nonpathogenic CD4(+) T cells raised from Il23a(-/-) donors. Thus, IL-23 may no longer be necessary to uphold inflammation in established autoimmune diseases. In addition, we demonstrated that IL-23-induced GM-CSF mediates the pathogenicity of CD4(+) T cells in EAM. The neutralization of GM-CSF abrogated cardiac inflammation. However, sustained IL-23 signaling is required to maintain IL-17A production in CD4(+) T cells. Despite inducing inflammation in Il23a(-/-) recipients comparable to wild-type (WT), autoreactive CD4(+) T cells downregulated IL-17A production without persistent IL-23 signaling. This divergence on the controls of GM-CSF-dependent pathogenicity on one side and IL-17A production on the other side may contribute to the discrepant efficacies of anti-IL-23 therapy in different autoimmune diseases.


Assuntos
Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Interleucina-23/metabolismo , Miocardite/imunologia , Transdução de Sinais , Animais , Modelos Animais de Doenças , Feminino , Interleucina-17/biossíntese , Interleucina-17/genética , Interleucina-23/deficiência , Interleucina-23/genética , Interleucina-23/farmacologia , Camundongos , Miocardite/fisiopatologia , Baço/citologia , Baço/efeitos dos fármacos , Baço/metabolismo , Células Th17/imunologia
6.
Eur J Immunol ; 46(12): 2749-2760, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27621211

RESUMO

Cardiac manifestations are a major cause of morbidity and mortality in patients with eosinophil-associated diseases. Eosinophils are thought to play a pathogenic role in myocarditis. We investigated the pathways that recruit eosinophils to the heart using a model of eosinophilic myocarditis, in which experimental autoimmune myocarditis (EAM) is induced in IFNγ-/- IL-17A-/- mice. Two conditions are necessary for efficient eosinophil trafficking to the heart: high eotaxin (CCL11, CCL24) expression in the heart and expression of the eotaxin receptor CCR3 by eosinophils. We identified cardiac fibroblasts as the source of CCL11 in the heart interstitium. CCL24 is produced by F4/80+ macrophages localized at inflammatory foci in the heart. Expression of CCL11 and CCL24 is controlled by Th2 cytokines, IL-4 and IL-13. To determine the relevance of this pathway in humans, we analyzed endomyocardial biopsy samples from myocarditis patients. Expression of CCL11 and CCL26 was significantly increased in eosinophilic myocarditis compared to chronic lymphocytic myocarditis and positively correlated with the number of eosinophils. Thus, eosinophil trafficking to the heart is dependent on the eotaxin-CCR3 pathway in a mouse model of EAM and associated with cardiac eotaxin expression in patients with eosinophilic myocarditis. Blocking this pathway may prevent eosinophil-mediated cardiac damage.


Assuntos
Quimiocina CCL11/metabolismo , Quimiocina CCL24/metabolismo , Eosinófilos/imunologia , Fibroblastos/imunologia , Macrófagos/imunologia , Miocardite/imunologia , Miocárdio/imunologia , Doença Autoimune do Sistema Nervoso Experimental/imunologia , Adulto , Idoso , Animais , Miosinas Cardíacas/imunologia , Movimento Celular , Células Cultivadas , Feminino , Humanos , Interferon gama/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Miocárdio/patologia , Receptores CCR3/genética , Equilíbrio Th1-Th2
7.
Am J Pathol ; 186(9): 2337-52, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27470712

RESUMO

Infections with Staphylococcus aureus are a continuing and growing problem in community and hospital settings. Preclinical animal modeling of S. aureus relies on experimental infection, which carries some limitations. We describe here a novel, spontaneous model of oral staphylococcal infection in double knockout mice, deficient in the receptors for IL-17 (IL-17RA) and interferon (IFN)-γ (IFNγRI), beginning at 6 to 8 weeks of age. IFNγRI(-/-)IL17RA(-/-) (GRAKO) mice developed progressive oral abscesses. Cytometric methods revealed extensive neutrophilic infiltration of oral tissues in GRAKO mice; further investigation evidenced that IL-17 predominated neutrophil defects in these mice. To investigate the contribution of IFN-γ signaling to this native host defense to S. aureus, we observed perturbations of monocyte recruitment and macrophage differentiation in the oral tissues of GRAKO mice, and CXCL9/chemokine ligand receptor (CXCR)3-driven recruitment of T-cell oral tissues and draining lymph nodes. To address the former finding, we depleted macrophages and monocytes in vivo from IL17RA(-/-) mice using liposomes loaded with clodronate. This treatment elicited oral abscesses, recapitulating the phenotype of GRAKO mice. From these findings, we propose novel collaborative functions of IL-17 and IFN-γ, acting through neutrophils and macrophages, respectively, in native mucocutaneous host defenses to S. aureus.


Assuntos
Interferon gama/imunologia , Interleucina-17/imunologia , Mucosa Bucal/imunologia , Mucosa Bucal/microbiologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Transdução de Sinais/imunologia
8.
Exp Mol Pathol ; 103(2): 141-152, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28822770

RESUMO

The extensive, diverse communities that constitute the microbiome are increasingly appreciated as important regulators of human health and disease through inflammatory, immune, and metabolic pathways. We sought to elucidate pathways by which microbiota contribute to inflammatory, autoimmune cardiac disease. We employed an animal model of experimental autoimmune myocarditis (EAM), which results in inflammatory and autoimmune pathophysiology and subsequent maladaptive cardiac remodeling and heart failure. Antibiotic dysbiosis protected mice from EAM and fibrotic cardiac dysfunction. Additionally, mice derived from different sources with different microbiome colonization profiles demonstrated variable susceptibility to disease. Unexpectedly, it did not track with segmented filamentous bacteria (SFB)-driven Th17 programming of CD4+ T cells in the steady-state gut. Instead, we found disease susceptibility to track with presence of type 3 innate lymphoid cells (ILC3s). Ablating ILCs by antibody depletion or genetic tools in adoptive transfer variants of the EAM model demonstrated that ILCs and microbiome profiles contributed to the induction of CCL20/CCR6-mediated inflammatory chemotaxis to the diseased heart. From these data, we conclude that sensing of the microbiome by ILCs is an important checkpoint in the development of inflammatory cardiac disease processes through their ability to elicit cardiotropic chemotaxis.


Assuntos
Antibacterianos/farmacologia , Doenças Autoimunes/imunologia , Coração/fisiopatologia , Linfócitos/imunologia , Microbiota , Miocardite/imunologia , Animais , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/metabolismo , Modelos Animais de Doenças , Disbiose/prevenção & controle , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Miocardite/tratamento farmacológico , Miocardite/metabolismo
9.
Am J Epidemiol ; 183(5): 403-6, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26888748

RESUMO

Autoimmune diseases represent a family of at least 80 illnesses that share a common pathogenesis: an immune-mediated attack on the body's own organs. Collectively, autoimmune diseases affect some 20 million Americans, predominantly women. Many of these diseases are increasing in frequency in industrialized countries. Treatment of autoimmune diseases improved greatly during the second half of the 20th century but has been hampered because the diseases often progress before a clinical diagnosis is possible. Research in the 21st century is focusing on prospective prediction using the tools of systems biology.


Assuntos
Doenças Autoimunes/prevenção & controle , Previsões , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/epidemiologia , Feminino , História do Século XX , História do Século XXI , Humanos , Masculino , Estados Unidos/epidemiologia
10.
Curr Opin Rheumatol ; 28(4): 383-9, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27166925

RESUMO

PURPOSE OF REVIEW: The article traces the pathways leading from viral infection of the heart by coxsackievirus B3 to autoimmune myocarditis in its various manifestations. RECENT FINDINGS: Myocarditis can be induced by a number of different infectious agents and represents a significant cause of death especially in young individuals. Following infection, patients may develop lymphocytic, eosinophilic, or giant cell/granulomatous myocardial inflammation. It can lead to infectious dilated cardiomyopathy, a disease frequently requiring cardiac transplantation. Although acute viral myocarditis is frequently subclinical and recovery may be spontaneous, treatment of chronic myocarditis is currently unsatisfactory. Ongoing disease may be because of persistent virus in the heart or to immunopathic attack. Depending on the cause, treatment may be antiviral or immunosuppressive. Endomyocardial biopsy is proving of value in determining cause and deciding future therapy. A great deal of information about the pathogenesis of myocarditis has been gained from experimental models in rodents using heart disease induced by infection using coxsackievirus B3 or by immunization with cardiac myosin. SUMMARY: Treatment of myocarditis is still problematic and may depend on etiologic diagnosis to distinguish infectious from immune-mediated disease. Both pathogenic mechanisms may co-occur in individual patients. In the future, treatment may depend upon endomyocardial biopsy, immunohistologic testing, improved imaging, and molecular genetic analysis for providing more precise diagnoses.


Assuntos
Infecções por Coxsackievirus/diagnóstico , Miocardite/virologia , Animais , Antivirais/uso terapêutico , Doenças Autoimunes/virologia , Biópsia/efeitos adversos , Cardiomiopatia Dilatada/diagnóstico , Cardiomiopatia Dilatada/imunologia , Cardiomiopatia Dilatada/virologia , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/tratamento farmacológico , Infecções por Coxsackievirus/imunologia , Modelos Animais de Doenças , Enterovirus Humano B , Humanos , Miocardite/diagnóstico , Miocardite/imunologia , Viroses/complicações , Viroses/diagnóstico , Viroses/tratamento farmacológico
11.
Am J Pathol ; 185(3): 847-61, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25622543

RESUMO

Myocarditis is a leading cause of sudden cardiac failure in young adults. Natural killer (NK) cells, a subset of the innate lymphoid cell compartment, are protective in viral myocarditis. Herein, we demonstrated that these protective qualities extend to suppressing autoimmune inflammation. Experimental autoimmune myocarditis (EAM) was initiated in BALB/c mice by immunization with myocarditogenic peptide. During EAM, activated cardiac NK cells secreted interferon γ, perforin, and granzyme B, and expressed CD69, tumor necrosis factor-related apoptosis-inducing ligand treatment, and CD27 on their cell surfaces. The depletion of NK cells during EAM with anti-asialo GM1 antibody significantly increased myocarditis severity, and was accompanied by elevated fibrosis and a 10-fold increase in the percentage of cardiac-infiltrating eosinophils. The resultant influx of eosinophils to the heart was directly responsible for the increased disease severity in the absence of NK cells, because treatment with polyclonal antibody asialogangloside GM-1 did not augment myocarditis severity in eosinophil-deficient ΔdoubleGATA1 mice. We demonstrate that NK cells limit eosinophilic infiltration both indirectly, through altering eosinophil-related chemokine production by cardiac fibroblasts, and directly, by inducing eosinophil apoptosis in vitro. Altogether, we define a new pathway of eosinophilic regulation through interactions with NK cells.


Assuntos
Eosinófilos/imunologia , Células Matadoras Naturais/imunologia , Miocardite/imunologia , Miocárdio/imunologia , Animais , Apoptose/imunologia , Eosinófilos/patologia , Fibrose/imunologia , Fibrose/patologia , Inflamação/patologia , Células Matadoras Naturais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Miocardite/patologia , Miocárdio/patologia
12.
J Autoimmun ; 75: 20-29, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27545842

RESUMO

In the broad field of autoimmunity and clinical immunology, experimental evidence over the past few years have demonstrated several connections between the immune system and the nervous system, both central and peripheral, leading to the definition of neuroimmunology and of an immune-brain axis. Indeed, the central nervous system as an immune-privileged site, thanks to the blood-brain barrier, is no longer a dogma as the barrier may be altered during chronic inflammation with disruptive changes of endothelial cells and tight junctions, largely mediated by adenosine receptors and the expression of CD39/CD73. The diseases that encompass the neuroimmunology field vary from primary nervous diseases such as multiple sclerosis to systemic conditions with neuropsychiatric complications, such as systemic lupus erythematosus or vasculitidies. Despite potentially similar clinical manifestations, the pathogenesis of each condition is different, but the interaction between the ultra-specialized structure that is the nervous system and inflammation mediators are crucial. Two examples come from anti-dsDNA cross-reacting with anti-N-Methyl-d-Aspartate receptor (NMDAR) antibodies in neuropsychiatric lupus or the new family of antibody-associated neuronal autoimmune diseases including classic paraneoplastic syndromes with antibodies directed to intracellular antigens (Hu, Yo, Ri) and autoimmune encephalitis. In the case of multiple sclerosis, the T cell paradigm is now complicated by the growing evidence of a B cell involvement, particularly via aquaporin antibodies, and their influence on Th1 and Th17 lineages. Inspired by a productive AARDA-sponsored colloquium among experts we provide a critical review of the literature on the pathogenesis of different immune-mediated diseases with neurologic manifestations and we discuss the basic immunology of the central nervous system and the interaction between immune cells and the peripheral nervous system.


Assuntos
Autoimunidade/imunologia , Encéfalo/imunologia , Sistema Nervoso Central/imunologia , Sistema Imunitário/imunologia , Sistema Nervoso Periférico/imunologia , Autoanticorpos/imunologia , Encefalite/imunologia , Doença de Hashimoto/imunologia , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Esclerose Múltipla/imunologia
13.
Cytokine ; 74(1): 62-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25649043

RESUMO

IL6 is a pleiotropic cytokine that is made in response to perturbations in homeostasis. IL6 becomes elevated in the acute response to host injury and can activate immune cells, direct immune cell trafficking, signal protective responses in local tissue, initial the acute phase response or initiate wound healing. In the short term this proinflammatory response is protective and limits host damage. It is when this acute response remains chronically activated that IL6 becomes pathogenic to the host. Chronically elevated IL6 levels lead to chronic inflammation and fibrotic disorders. The heart is a tissue where this temporal regulation of IL6 is very apparent. Studies from myocardial infarction show how short-term IL6 signaling can protect and preserve the heart tissue in response to acute damage, where long term IL6 signaling or an over-production of IL6R protein plays a causal role in cardiovascular disease. Thus, IL6 can be both protective and pathogenic, depending on the kinetics of the host response.


Assuntos
Cardiopatias/imunologia , Cardiopatias/metabolismo , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/metabolismo , Interleucina-6/metabolismo , Miocárdio/metabolismo , Animais , Receptor gp130 de Citocina/imunologia , Receptor gp130 de Citocina/metabolismo , Humanos , Inflamação/imunologia , Interleucina-6/biossíntese , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/imunologia , Miocardite/imunologia , Miocárdio/imunologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/metabolismo , Receptores de Interleucina-6/metabolismo , Transdução de Sinais
14.
J Theor Biol ; 375: 71-76, 2015 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-25172771

RESUMO

In this article, I trace the historic background of clonal deletion and molecular mimicry, two major pillars underlying our present understanding of autoimmunity and autoimmune disease. Clonal deletion originated as a critical element of the clonal selection theory of antibody formation in order to explain tolerance of self. If we did have complete clonal deletion, there would be major voids, the infamous "black holes", in our immune repertoire. For comprehensive, protective adaptive immunity, full deletion is necessarily a rare event. Molecular mimicry, the sharing of epitopes among self and non-self antigens, is extraordinary common and provides the evidence that complete deletion of self-reactive clones is rare. If molecular mimicry were not common, protective adaptive immunity could not be all-encompassing. By taking a fresh look at these two processes together we can envision their evolutionary basis and understand the need for regulatory devices to prevent molecular mimicry from progressing to autoimmune disease.


Assuntos
Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Deleção Clonal , Tolerância Imunológica/imunologia , Mimetismo Molecular , Animais , Autoantígenos/imunologia , Epitopos/imunologia , Homeostase , Humanos , Linfócitos/imunologia , Modelos Biológicos
15.
Inflamm Res ; 64(1): 31-40, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25376339

RESUMO

OBJECTIVE: Mercury is a ubiquitous environmental contaminant with toxic outcomes over a range of exposures. In this study, we investigated the effects of mercury exposure on early immune responses to coxsackievirus B3 (CVB3) infection in a murine model of autoimmune heart disease. MATERIALS AND METHODS: Female BALB/c mice, susceptible to CVB3-induced autoimmune myocarditis, were treated with mercuric chloride (200 µg/kg body weight every other day for 2 weeks) prior to infection with CVB3. Six hours post-infection, immune cells were isolated from the spleen and peritoneum for flow cytometry, gene expression, and cytokine profiling. Thirty-five days post-infection, hearts were collected for histological examination of immune cell infiltration. RESULTS: As for male mice, mercury exposure significantly increased autoimmune myocarditis and immune infiltration into the heart. During the innate response 6 h post-infection, mercury increased expression of co-stimulatory molecules and innate immune receptors on peritoneal macrophages. At the same time point, the alternatively activated macrophage gene, arginase, was increased while the classically activated macrophage gene, inducible nitric oxide synthase, was unaffected. Expression of activation markers were decreased on peritoneal B cells with mercury exposure while T cells were unaffected. Mercury increased production of pro-inflammatory mediators in the spleen. Macrophage-recruiting chemokines and activating cytokines, such as CCL2, CCL4, and IL-6, were increased with mercury following CVB3 infection. CONCLUSIONS: Thus, mercury treatment exacerbates autoimmune myocarditis in female mice and alters early innate signaling on peritoneal macrophages. Mercury also modulates the cytokine profile in the spleen toward a macrophage-activating milieu, and upregulates alternatively activated macrophage genes, providing evidence that mercury exposure promotes inflammation in the context of infection.


Assuntos
Doenças Autoimunes/etiologia , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/imunologia , Imunidade Inata/efeitos dos fármacos , Cloreto de Mercúrio/farmacologia , Miocardite/etiologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/patologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Relação Dose-Resposta a Droga , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Miocardite/imunologia , Miocardite/patologia , Baço/efeitos dos fármacos , Baço/metabolismo , Baço/patologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia
16.
J Immunol ; 191(8): 4038-47, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24048893

RESUMO

CD4(+) T cells play a central role in inflammatory heart disease, implicating a cytokine product associated with Th cell effector function as a necessary mediator of this pathophysiology. IFN-γ-deficient mice developed severe experimental autoimmune myocarditis (EAM), in which mice are immunized with cardiac myosin peptide, whereas IL-17A-deficient mice were protected from progression to dilated cardiomyopathy. We generated IFN-γ(-/-)IL-17A(-/-) mice to assess whether IL-17 signaling was responsible for the severe EAM of IFN-γ(-/-) mice. Surprisingly, IFN-γ(-/-)IL-17A(-/-) mice developed a rapidly fatal EAM. Eosinophils constituted a third of infiltrating leukocytes, qualifying this disease as eosinophilic myocarditis. We found increased cardiac production of CCL11/eotaxin, as well as Th2 deviation, among heart-infiltrating CD4(+) cells. Ablation of eosinophil development improved survival of IFN-γ(-/-)IL-17A(-/-) mice, demonstrating the necessity of eosinophils in fatal heart failure. The severe and rapidly fatal autoimmune inflammation that developed in the combined absence of IFN-γ and IL-17A constitutes a novel model of eosinophilic heart disease in humans. This is also, to our knowledge, the first demonstration that eosinophils have the capacity to act as necessary mediators of morbidity in an autoimmune process.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Eosinófilos/imunologia , Interferon gama/deficiência , Interleucina-17/deficiência , Miocardite/imunologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/prevenção & controle , Biomarcadores , Miosinas Cardíacas/imunologia , Cardiomiopatias/imunologia , Quimiocina CCL11/biossíntese , Inflamação , Interferon gama/genética , Interferon gama/imunologia , Interleucina-17/genética , Interleucina-17/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Miocardite/genética , Miocardite/prevenção & controle , Miocárdio/imunologia , Miosite
17.
Int J Mol Sci ; 15(8): 14269-97, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25196523

RESUMO

Autoimmune diseases include 80 or more complex disorders characterized by self-reactive, pathologic immune responses in which genetic susceptibility is largely insufficient to determine disease onset. In September 2010, the National Institute of Environmental Health Sciences (NIEHS) organized an expert panel workshop to evaluate the role of environmental factors in autoimmune diseases, and the state of the science regarding relevant mechanisms, animal models, and human studies. The objective of the workshop was to analyze the existing data to identify conclusions that could be drawn regarding environmental exposures and autoimmunity and to identify critical knowledge gaps and areas of uncertainty for future study. This consensus document summarizes key findings from published workshop monographs on areas in which "confident" and "likely" assessments were made, with recommendations for further research. Transcribed notes and slides were reviewed to synthesize an overview on exposure assessment and questions addressed by interdisciplinary panels. Critical advances in the field of autoimmune disease research have been made in the past decade. Collaborative translational and interdisciplinary research is needed to elucidate the role of environmental factors in autoimmune diseases. A focus on exposure assessment methodology is needed to improve the effectiveness of human studies, and more experimental studies are needed to focus on causal mechanisms underlying observed associations of environmental factors with autoimmune disease in humans.


Assuntos
Doenças Autoimunes/epidemiologia , Exposição Ambiental/efeitos adversos , Doenças Autoimunes/etiologia , Humanos
18.
Eur J Immunol ; 42(3): 726-36, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22161142

RESUMO

The involvement of macrophages (MΦs) in Th17-cell responses is still poorly understood. While neutrophils are thought to be the predominant effector of Th17-cell responses, IL-17 is also known to induce myelotropic chemokines and growth factors. Other T-cell-derived cytokines induce non-classical functions, suggesting that IL-17 sigxnaling may similarly elicit unique MΦ functions. Here, we characterized the expression of subunits of the IL-17 receptor on primary murine MΦs from different anatomical compartments. The greatest expression of IL-17 receptors was observed on mucosal Ly6C(hi) "inflammatory" MΦs. We further observed upregulation of IL-17 receptors in vitro on bone marrow-derived macrophages (BMMΦs) in response to peptidoglycan or CpG oligonucleotide stimuli, and in vivo, upon CFA administration. Macrophages expressing IL-17 receptors were observed infiltrating the hearts of mice with myocarditis, and genetic ablation of IL-17RA altered MΦ recruitment. Treating primary MΦs from a wide variety of different anatomic sources (as well as cell lines) with IL-17A induced the production of unique profiles of cytokines and chemokines, including GM-CSF, IL-3, IL-9, CCL4/MIP-1ß and CCL5/RANTES. IL-17A also induced production of IL-12p70; IL-17-signaling-deficient MΦs elicited diminished IFN-γ production by responding DO11.10 CD4(+) T cells when used as APCs. These data indicate that MΦs from different anatomic locations direct IL-17-mediated responses.


Assuntos
Interleucina-17/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Miocardite/imunologia , Animais , Quimiocinas/imunologia , Feminino , Citometria de Fluxo , Inflamação/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de Interleucina-17/imunologia , Organismos Livres de Patógenos Específicos , Regulação para Cima/imunologia
19.
Camb Prism Precis Med ; 1: e25, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38550937

RESUMO

Precision Medicine is an emerging approach for disease treatment and prevention that takes into account individual variability in genes, environment, and lifestyle. Autoimmune diseases are those in which the body's natural defense system loses discriminating power between its own cells and foreign cells, causing the body to mistakenly attack healthy tissues. These conditions are very heterogeneous in their presentation and therefore difficult to diagnose and treat. Achieving precision medicine in autoimmune diseases has been challenging due to the complex etiologies of these conditions, involving an interplay between genetic, epigenetic, and environmental factors. However, recent technological and computational advances in molecular profiling have helped identify patient subtypes and molecular pathways which can be used to improve diagnostics and therapeutics. This review discusses the current understanding of the disease mechanisms, heterogeneity, and pathogenic autoantigens in autoimmune diseases gained from genomic and transcriptomic studies and highlights how these findings can be applied to better understand disease heterogeneity in the context of disease diagnostics and therapeutics.

20.
Clin Immunol ; 144(3): 237-49, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22854287

RESUMO

Enteroviruses like coxsackievirus B3 (CVB3) are common suspects in myocarditis/dilated cardiomyopathy patients. Autoimmunity has been proposed as an underlying mechanism, but direct evidence of its role is lacking. To delineate autoimmune response in CVB3 myocarditis, we used IA(k) dextramers for cardiac myosin heavy chain (Myhc)-α 334-352. We have demonstrated that myocarditis-susceptible A/J mice infected with CVB3 generate Myhc-α-reactive CD4 T cells and such a repertoire was absent in naïve mice as measured by proliferative response to Myhc-α 334-352 and IA(k) dextramer staining. We also detected Myhc-α 334-352 dextramer(+) cells in the hearts of CVB3-infected mice. The autoreactive T cell repertoire derived from infected mice contained a high frequency of interleukin-17-producing cells capable of inducing myocarditis in naïve recipients. The data suggest that CVB3, a bona fide pathogen of cardiovascular system that primarily infects the heart can lead to the secondary generation of autoreactive T cells and contribute to cardiac pathology.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Miosinas Cardíacas/imunologia , Infecções por Coxsackievirus/imunologia , Enterovirus Humano B/imunologia , Cadeias Pesadas de Miosina/imunologia , Animais , Autoimunidade/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/virologia , Cardiomiopatia Dilatada/imunologia , Cardiomiopatia Dilatada/patologia , Chlorocebus aethiops , Infecções por Coxsackievirus/patologia , Coração/virologia , Interleucina-17/imunologia , Camundongos , Miocardite/imunologia , Miocardite/patologia , Miocardite/virologia , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA