Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mucosal Immunol ; 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38570140

RESUMO

Sublingual allergen immunotherapy (SLIT) is an emerging treatment option for allergic asthma and a potential disease-modifying strategy for asthma prevention. The key cellular events leading to such long-term tolerance remain to be fully elucidated. We administered prophylactic SLIT in a mouse model of house dust mite (HDM)-driven allergic asthma. HDM extract was sublingually administered over 3 weeks followed by intratracheal sensitization and intranasal challenges with HDM. Prophylactic SLIT prevented allergic airway inflammation and hyperreactivity with a low lab-to-lab variation. The HDM-specific T helper (Th)2 (cluster of differentiation 4 Th) response was shifted by SLIT toward a regulatory and Th17 response in the lung and mediastinal lymph node. By using Derp1-specific cluster of differentiation 4+ T cells (1-DER), we found that SLIT blocked 1-DER T cell recruitment to the mediastinal lymph node and dampened IL-4 secretion following intratracheal HDM sensitization. Sublingually administered Derp1 protein activated 1-DER T cells in the cervical lymph node via chemokine receptor7+ migratory dendritic cells (DC). DCs migrating from the oral submucosa to the cervical lymph node after SLIT-induced Foxp3+ regulatory T cells. When mice were sensitized with HDM, prior prophylactic SLIT increased Derp1 specific regulatory T cells (Tregs) and lowered Th2 recruitment in the lung. By using Foxp3-diphtheria toxin receptor mice, Tregs were found to contribute to the immunoregulatory prophylactic effect of SLIT on type 2 immunity. These findings in a mouse model suggest that DC-mediated functional Treg induction in oral mucosa draining lymph nodes is one of the driving mechanisms behind the disease-modifying effect of prophylactic SLIT.

2.
Cytometry A ; 105(6): 464-473, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38456613

RESUMO

Over the past decade, the flow cytometry field has witnessed significant advancements in the number of fluorochromes that can be detected. This enables researchers to analyze more than 40 markers simultaneously on thousands of cells per second. However, with this increased complexity and multiplicity of markers, the manual dispensing of antibodies for flow cytometry experiments has become laborious, time-consuming, and prone to errors. An automated antibody dispensing system could provide a potential solution by enhancing the efficiency, and by improving data quality by faithfully dispensing the fluorochrome-conjugated antibodies and by enabling the easy addition of extra controls. In this study, a comprehensive comparison of different liquid handlers for dispensing fluorochrome-labeled antibodies was conducted for the preparation of flow cytometry stainings. The evaluation focused on key criteria including dispensing time, dead volume, and reliability of dispensing. After benchmarking, the I.DOT, a non-contact liquid handler, was selected and optimized in more detail. In the end, the I.DOT was able to prepare a 25-marker panel in 20 min, including the full stain, all FMOs and all single stain controls for cells and beads. Having all these controls improved the validation of the panel, visualization, and analysis of the data. Thus, automated antibody dispensing by dispensers such as the I.DOT reduces time and errors, enhances data quality, and can be easily integrated in an automated workflow to prepare samples for flow cytometry.


Assuntos
Anticorpos , Citometria de Fluxo , Corantes Fluorescentes , Citometria de Fluxo/métodos , Humanos , Anticorpos/imunologia , Corantes Fluorescentes/química , Coloração e Rotulagem/métodos , Ensaios de Triagem em Larga Escala/métodos , Automação , Reprodutibilidade dos Testes
3.
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
4.
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
5.
Cell Immunol ; 330: 105-113, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29650244

RESUMO

Heart disease is the major cause of death and it is broadly recognized that the immune system plays a central role in healthy and injured heart. Here, we focus on the contribution of various subsets of mononuclear phagocytes in the cardiac system. Macrophages and dendritic cells reside in the healthy myocardium to fulfill homeostatic functions and rapidly increase in numbers in diseases like myocardial ischemia and myocarditis to contribute to disease or resolve it. Recent experiments have revealed the extraordinary heterogeneity of cardiac mononuclear phagocytes that differ in origin, lifespan, phenotype and function. Although many studies described cardiac phagocytes in the mouse, subsets of cardiac mononuclear phagocytes can also be broadly found in the human heart, opening up the potential of selective targeting of these cells in a therapeutic setting. Before this goal can be achieved we need better understanding not only of the detrimental but also beneficial functions of these highly diverse cells in the heart.


Assuntos
Células Dendríticas/imunologia , Macrófagos/imunologia , Isquemia Miocárdica/imunologia , Miocardite/imunologia , Miocárdio/imunologia , Animais , Humanos , Modelos Cardiovasculares , Modelos Imunológicos , Fagócitos/imunologia
6.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...