Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Science ; 366(6467): 881-886, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31727837

RESUMO

Myocarditis can develop into inflammatory cardiomyopathy through chronic stimulation of myosin heavy chain 6-specific T helper (TH)1 and TH17 cells. However, mechanisms governing the cardiotoxicity programming of heart-specific T cells have remained elusive. Using a mouse model of spontaneous autoimmune myocarditis, we show that progression of myocarditis to lethal heart disease depends on cardiac myosin-specific TH17 cells imprinted in the intestine by a commensal Bacteroides species peptide mimic. Both the successful prevention of lethal disease in mice by antibiotic therapy and the significantly elevated Bacteroides-specific CD4+ T cell and B cell responses observed in human myocarditis patients suggest that mimic peptides from commensal bacteria can promote inflammatory cardiomyopathy in genetically susceptible individuals. The ability to restrain cardiotoxic T cells through manipulation of the microbiome thereby transforms inflammatory cardiomyopathy into a targetable disease.


Assuntos
Doenças Autoimunes/complicações , Bacteroides/imunologia , Cardiomiopatia Dilatada/imunologia , Cardiomiopatia Dilatada/microbiologia , Microbioma Gastrointestinal/imunologia , Miocardite/complicações , Peptídeos/imunologia , beta-Galactosidase/imunologia , Animais , Doenças Autoimunes/imunologia , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Humanos , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Miocardite/imunologia , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/imunologia , Células Th17/imunologia
2.
Front Immunol ; 9: 2714, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30524444

RESUMO

Autoimmune myocarditis often leads to dilated cardiomyopathy (DCM). Although T cell reactivity to cardiac self-antigen is common in the disease, it is unknown which antigen presenting cell (APC) triggers autoimmunity. Experimental autoimmune myocarditis (EAM) was induced by immunizing mice with α-myosin loaded bone marrow APCs cultured in GM-CSF. APCs found in such cultures include conventional type 2 CD11b+ cDCs (GM-cDC2s) and monocyte-derived cells (GM-MCs). However, only α-myosin loaded GM-cDC2s could induce EAM. We also studied antigen presenting capacity of endogenous type 1 CD24+ cDCs (cDC1s), cDC2s, and MCs for α-myosin-specific TCR-transgenic TCR-M CD4+ T cells. After EAM induction, all cardiac APCs significantly increased and cDCs migrated to the heart-draining mediastinal lymph node (LN). Primarily cDC2s presented α-myosin to TCR-M cells and induced Th1/Th17 differentiation. Loss of IRF4 in Irf4fl/fl.Cd11cCre mice reduced MHCII expression on GM-cDC2s in vitro and cDC2 migration in vivo. However, partly defective cDC2 functions in Irf4fl/fl.Cd11cCre mice did not suppress EAM. MCs were the largest APC subset in the inflamed heart and produced pro-inflammatory cytokines. Targeting APC populations could be exploited in the design of new therapies for cardiac autoimmunity.


Assuntos
Apresentação de Antígeno , Autoantígenos , Doenças Autoimunes , Células Dendríticas/imunologia , Monócitos/imunologia , Miocardite , Animais , Autoantígenos/genética , Autoantígenos/imunologia , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/imunologia , Cardiomiopatia Dilatada/patologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Dendríticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Monócitos/patologia , Miocardite/genética , Miocardite/imunologia , Miocardite/patologia , Células Th1/imunologia , Células Th1/patologia , Células Th17/imunologia , Células Th17/patologia
3.
Cell Rep ; 18(12): 3005-3017, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28329691

RESUMO

Peripheral tolerance is crucial for avoiding activation of self-reactive T cells to tissue-restricted antigens. Sterile tissue injury can break peripheral tolerance, but it is unclear how autoreactive T cells get activated in response to self. An example of a sterile injury is myocardial infarction (MI). We hypothesized that tissue necrosis is an activator of dendritic cells (DCs), which control tolerance to self-antigens. DC subsets of a murine healthy heart consisted of IRF8-dependent conventional (c)DC1, IRF4-dependent cDC2, and monocyte-derived DCs. In steady state, cardiac self-antigen α-myosin was presented in the heart-draining mediastinal lymph node (mLN) by cDC1s, driving the proliferation of antigen-specific CD4+ TCR-M T cells and their differentiation into regulatory cells (Tregs). Following MI, all DC subsets infiltrated the heart, whereas only cDCs migrated to the mLN. Here, cDC2s induced TCR-M proliferation and differentiation into interleukin-(IL)-17/interferon-(IFN)γ-producing effector cells. Thus, cardiac-specific autoreactive T cells get activated by mature DCs following myocardial infarction.


Assuntos
Células Dendríticas/imunologia , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Linfócitos T/imunologia , Animais , Antígeno CD11c/metabolismo , Linfócitos T CD4-Positivos/imunologia , Movimento Celular , Fatores Reguladores de Interferon/metabolismo , Linfonodos/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/patologia , Miocárdio/patologia , Miosinas/metabolismo , Fenótipo , Fatores de Transcrição/metabolismo
4.
Eur J Immunol ; 45(8): 2218-31, 2015 08.
Artigo em Inglês | MEDLINE | ID: mdl-25973789

RESUMO

The thymic epithelium forms specialized niches to enable thymocyte differentiation. While the common epithelial progenitor of medullary and cortical thymic epithelial cells (mTECs and cTECs) is well defined, early stages of mTEC lineage specification have remained elusive. Here, we utilized in vivo targeting of mTECs to resolve their differentiation pathways and to determine whether mTEC progenitors participate in thymocyte education. We found that mTECs descend from a lineage committed, podoplanin (PDPN)-expressing progenitor located at the cortico-medullary junction. PDPN(+) junctional TECs (jTECs) represent a distinct TEC population that builds the thymic medulla, but only partially supports negative selection and thymocyte differentiation. Moreover, conditional gene targeting revealed that abrogation of alternative NF-κB pathway signaling in the jTEC stage completely blocked mTEC development. Taken together, this study identifies jTECs as lineage-committed mTEC progenitors and shows that NF-κB-dependent progression of jTECs to mTECs is critical to secure central tolerance.


Assuntos
Diferenciação Celular/imunologia , Células Epiteliais/imunologia , Glicoproteínas de Membrana/imunologia , NF-kappa B/imunologia , Transdução de Sinais/imunologia , Células-Tronco/imunologia , Timo/imunologia , Animais , Diferenciação Celular/genética , Células Epiteliais/citologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , NF-kappa B/genética , Transdução de Sinais/genética , Células-Tronco/citologia , Timo/citologia
5.
J Exp Med ; 209(13): 2485-99, 2012 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-23230000

RESUMO

The inhibitory programmed death 1 (PD-1)-programmed death ligand 1 (PD-L1) pathway contributes to the functional down-regulation of T cell responses during persistent systemic and local virus infections. The blockade of PD-1-PD-L1-mediated inhibition is considered as a therapeutic approach to reinvigorate antiviral T cell responses. Yet previous studies reported that PD-L1-deficient mice develop fatal pathology during early systemic lymphocytic choriomeningitis virus (LCMV) infection, suggesting a host protective role of T cell down-regulation. As the exact mechanisms of pathology development remained unclear, we set out to delineate in detail the underlying pathogenesis. Mice deficient in PD-1-PD-L1 signaling or lacking PD-1 signaling in CD8 T cells succumbed to fatal CD8 T cell-mediated immunopathology early after systemic LCMV infection. In the absence of regulation via PD-1, CD8 T cells killed infected vascular endothelial cells via perforin-mediated cytolysis, thereby severely compromising vascular integrity. This resulted in systemic vascular leakage and a consequential collapse of the circulatory system. Our results indicate that the PD-1-PD-L1 pathway protects the vascular system from severe CD8 T cell-mediated damage during early systemic LCMV infection, highlighting a pivotal physiological role of T cell down-regulation and suggesting the potential development of immunopathological side effects when interfering with the PD-1-PD-L1 pathway during systemic virus infections.


Assuntos
Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/fisiopatologia , Receptor de Morte Celular Programada 1/fisiologia , Choque/fisiopatologia , Animais , Antígeno B7-H1/deficiência , Antígeno B7-H1/genética , Antígeno B7-H1/fisiologia , Linfócitos T CD8-Positivos/imunologia , Permeabilidade Capilar/fisiologia , Modelos Animais de Doenças , Endotélio Vascular/patologia , Antígenos H-2/genética , Antígenos H-2/fisiologia , Antígeno de Histocompatibilidade H-2D , Hipotensão/etiologia , Hipotensão/fisiopatologia , Coriomeningite Linfocítica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Citotóxicas Formadoras de Poros/deficiência , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Receptor de Morte Celular Programada 1/deficiência , Receptor de Morte Celular Programada 1/genética , Edema Pulmonar/etiologia , Edema Pulmonar/fisiopatologia , Choque/imunologia , Choque/prevenção & controle , Transdução de Sinais
6.
Front Immunol ; 3: 279, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22969771

RESUMO

Vascular endothelial cells (ECs) form the inner layer of blood vessels and exert crucial functions during immune reactions including coagulation, inflammation, and regulation of innate immunity. Importantly, ECs can interact with T cells in an antigen-specific, i.e., T cell receptor-dependent manner. In this review, we will discuss EC actions and reactions during acute inflammation and focus on the interaction of T cells with ECs at two vascular sites: the high endothelial venule (HEV) of lymph nodes, and the vascular lesion during transplant vasculopathy (TV). HEVs are characterized by a highly active endothelium that produces chemoattracting factors and expresses adhesion molecules to facilitate transit of lymphocytes into the lymph node (LN) parenchyma. Yet, T cell-EC interaction at this anatomical location results neither in T cell activation nor tolerization. In contrast, the endothelium at sites of chronic inflammation, such as solid organ transplants, can promote T cell activation by upregulation of major histocompatibility complex (MHC) and costimulatory molecules. Importantly, a major function of ECs in inflamed tissues must be the maintenance of vascular integrity including the efficient attenuation of effector T cells that may damage the vascular bed. Thus, antigen-specific T cell-EC interaction is characterized by a tightly controlled balance between immunological ignorance, immune activation, and tolerization.

7.
Eur J Immunol ; 42(9): 2311-21, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22730043

RESUMO

Myocarditis is a potentially lethal inflammatory heart disease of children and young adults that frequently leads to dilated cardiomyopathy (DCM). Since diagnostic procedures and efficient therapies are lacking, it is important to characterize the critical immune effector pathways underlying the initial cardiac inflammation and the transition from myocarditis to DCM. We describe here a T-cell receptor (TCR) transgenic mouse model with spontaneously developing autoimmune myocarditis that progresses to lethal DCM. Cardiac magnetic resonance imaging revealed early inflammation-associated changes in the ventricle wall including transient thickening of the left ventricle wall. Furthermore, we found that IFN-γ was a major effector cytokine driving the initial inflammatory process and that the cooperation of IFN-γ and IL-17A was essential for the development of the progressive disease. This novel TCR transgenic mouse model permits the identification of the central pathophysiological and immunological processes involved in the transition from autoimmune myocarditis to DCM.


Assuntos
Doenças Autoimunes/imunologia , Cardiomiopatia Dilatada/imunologia , Cardiomiopatia Dilatada/patologia , Miocardite/imunologia , Miocardite/patologia , Células Th1/imunologia , Células Th17/imunologia , Animais , Autoantígenos/imunologia , Doenças Autoimunes/patologia , Modelos Animais de Doenças , Ventrículos do Coração/imunologia , Ventrículos do Coração/patologia , Inflamação/imunologia , Inflamação/patologia , Interferon gama/imunologia , Interleucina-17/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/imunologia , Remodelação Ventricular/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...