Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cell ; 175(2): 458-471.e19, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30173917

RESUMO

Inflammatory disorders of the CNS are frequently accompanied by synaptic loss, which is thought to involve phagocytic microglia and complement components. However, the mechanisms accounting for aberrant synaptic connectivity in the context of CD8+ T cell-driven neuronal damage are poorly understood. Here, we profiled the neuronal translatome in a murine model of encephalitis caused by CD8+ T cells targeting antigenic neurons. Neuronal STAT1 signaling and downstream CCL2 expression were essential for apposition of phagocytes, ensuing synaptic loss and neurological disease. Analogous observations were made in the brains of Rasmussen's encephalitis patients. In this devastating CD8+ T cell-driven autoimmune disease, neuronal STAT1 phosphorylation and CCL2 expression co-clustered with infiltrating CD8+ T cells as well as phagocytes. Taken together, our findings uncover an active role of neurons in coordinating phagocyte-mediated synaptic loss and highlight neuronal STAT1 and CCL2 as critical steps in this process that are amenable to pharmacological interventions.


Assuntos
Neurônios/metabolismo , Fagocitose/fisiologia , Sinapses/fisiologia , Animais , Encéfalo/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/fisiologia , Modelos Animais de Doenças , Encefalite/genética , Encefalite/imunologia , Encefalite/fisiopatologia , Feminino , Humanos , Inflamação/imunologia , Inflamação/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Doenças do Sistema Nervoso/metabolismo , Neurônios/fisiologia , Fagócitos/imunologia , Fagócitos/metabolismo , Fagocitose/imunologia , Fosforilação , Fator de Transcrição STAT1/fisiologia , Transcriptoma/genética
2.
Immunity ; 48(5): 937-950.e8, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29768177

RESUMO

Infections are thought to trigger CD8+ cytotoxic T lymphocyte (CTL) responses during autoimmunity. However, the transcriptional programs governing the tissue-destructive potential of CTLs remain poorly defined. In a model of central nervous system (CNS) inflammation, we found that infection with lymphocytic choriomeningitis virus (LCMV), but not Listeria monocytogenes (Lm), drove autoimmunity. The DNA-binding factor TOX was induced in CTLs during LCMV infection and was essential for their encephalitogenic properties, and its expression was inhibited by interleukin-12 during Lm infection. TOX repressed the activity of several transcription factors (including Id2, TCF-1, and Notch) that are known to drive CTL differentiation. TOX also reduced immune checkpoint sensitivity by restraining the expression of the inhibitory checkpoint receptor CD244 on the surface of CTLs, leading to increased CTL-mediated damage in the CNS. Our results identify TOX as a transcriptional regulator of tissue-destructive CTLs in autoimmunity, offering a potential mechanistic link to microbial triggers.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proteínas de Homeodomínio/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Adulto , Idoso , Animais , Autoimunidade/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Feminino , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Família de Moléculas de Sinalização da Ativação Linfocitária/imunologia , Linfócitos T Citotóxicos/imunologia
4.
J Autoimmun ; 67: 8-18, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26341385

RESUMO

Plasmacytoid dendritic cells (pDCs) exhibit both innate and adaptive functions. In particular they are the main source of type I IFNs and directly impact T cell responses through antigen presentation. We have previously demonstrated that during experimental autoimmune encephalomyelitis (EAE) initiation, myelin-antigen presentation by pDCs is associated with suppressive Treg development and results in attenuated EAE. Here, we show that pDCs transferred during acute disease phase confer recovery from EAE. Clinical improvement is associated with migration of injected pDCs into inflamed CNS and is dependent on the subsequent and selective chemerin-mediated recruitment of endogenous pDCs to the CNS. The protective effect requires pDC pre-loading with myelin antigen, and is associated with the modulation of CNS-infiltrating pDC phenotype and inhibition of CNS encephalitogenic T cells. This study may pave the way for novel pDC-based cell therapies in autoimmune diseases, aiming at specifically modulating pathogenic cells that induce and sustain autoimmune inflammation.


Assuntos
Transferência Adotiva , Quimiotaxia/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Animais , Autoantígenos/imunologia , Terapia Baseada em Transplante de Células e Tecidos , Quimiocinas/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/terapia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos Knockout , Bainha de Mielina/imunologia , Receptores de Quimiocinas , Receptores Acoplados a Proteínas G/metabolismo , Medula Espinal/imunologia , Medula Espinal/metabolismo , Medula Espinal/patologia
5.
Brain ; 138(Pt 11): 3263-74, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26359290

RESUMO

Recent association studies have linked numerous genetic variants with an increased risk for multiple sclerosis, although their functional relevance remains largely unknown. Here we investigated phenotypical and functional consequences of a genetic variant in the CD226 gene that, among other autoimmune diseases, predisposes to multiple sclerosis. Phenotypically, effector and regulatory CD4(+) memory T cells of healthy individuals carrying the predisposing CD226 genetic variant showed, in comparison to carriers of the protective variant, reduced surface expression of CD226 and an impaired induction of CD226 after stimulation. This haplotype-dependent reduction in CD226 expression on memory T cells was abrogated in patients with multiple sclerosis, as CD226 expression was comparable to healthy risk haplotype carriers irrespective of genetic variant. Functionally, FOXP3-positive regulatory T cells from healthy carriers of the genetic protective variant showed superior suppressive capacity, which was again abrogated in multiple sclerosis patients. Mimicking the phenotype of human CD226 genetic risk variant carriers, regulatory T cells derived from Cd226-deficient mice showed similarly reduced inhibitory activity, eventually resulting in an exacerbated disease course of experimental autoimmune encephalomyelitis, the animal model of multiple sclerosis. Therefore, by combining human and mouse analyses we show that CD226 exhibits an important role in the activation of regulatory T cells, with its genetically imposed dysregulation impairing regulatory T cell function.


Assuntos
Antígenos de Diferenciação de Linfócitos T/genética , Esclerose Múltipla/genética , RNA Mensageiro/metabolismo , Linfócitos T Reguladores/imunologia , Adulto , Animais , Antígenos de Diferenciação de Linfócitos T/imunologia , Antígenos de Diferenciação de Linfócitos T/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Encefalomielite Autoimune Experimental/genética , Feminino , Fatores de Transcrição Forkhead/metabolismo , Predisposição Genética para Doença , Haplótipos , Heterozigoto , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Fenótipo , Polimorfismo de Nucleotídeo Único , Linfócitos T Reguladores/metabolismo
6.
Eur J Immunol ; 44(10): 3119-28, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25043505

RESUMO

Recent findings indicate a pathogenic involvement of IL-17-producing CD8(+) T cells in multiple sclerosis (MS). IL-17 production has been attributed to a subset of CD8(+) T cells that belong to the mucosal-associated invariant T (MAIT) cell population. Here, we report a reduction of CD8(+) MAIT cells in the blood of MS patients compared with healthy individuals, which significantly correlated with IL-18 serum levels in MS patients. In vitro stimulation of peripheral blood mononuclear cells from healthy individuals and MS patients with IL-18 specifically activated CD8(+) MAIT cells. Moreover, IL-18 together with T-cell receptor stimulation induced, specifically on CD8(+) MAIT cells, an upregulation of the integrin very late antigen-4 that is essential for the infiltration of CD8(+) T cells into the CNS. Notably, we were able to identify CD8(+) MAIT cells in MS brain lesions by immunohistochemistry while they were almost absent in the cerebrospinal fluid (CSF). In summary, our findings indicate that an IL-18-driven activation of CD8(+) MAIT cells contributes to their CNS infiltration in MS, in turn leading to reduced CD8(+) MAIT-cell frequencies in the blood. Therefore, CD8(+) MAIT cells seem to play a role in the innate arm of immunopathology in MS.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Interleucina-18/sangue , Esclerose Múltipla/imunologia , Subpopulações de Linfócitos T/imunologia , Quimiotaxia de Leucócito , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Ativação Linfocitária/imunologia , Esclerose Múltipla/sangue , Esclerose Múltipla/patologia
7.
J Immunol ; 191(9): 4531-9, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24062488

RESUMO

Multiple sclerosis is considered to be initiated by a deregulated, myelin-specific T cell response. However, the formation of inflammatory CNS lesions and the contribution of different leukocyte subsets in setting up these lesions are still incompletely understood. In this study, we show that, in the mouse model of multiple sclerosis, experimental autoimmune encephalomyelitis, neutrophil granulocytes are important contributors in preparing CNS inflammation. Preclinical single-dose Ab-mediated depletion of neutrophils delayed the onset and continuous depletion attenuated the development of experimental autoimmune encephalomyelitis, whereas the generation of a myelin-specific T cell response remained unaffected. Neutrophil-related enzymes such as myeloperoxidase and neutrophil elastase did not contribute in mounting CNS inflammation, as analyzed by using respective knockout mice and inhibitors. CNS-infiltrating neutrophils secreted proinflammatory molecules and matured bone marrow-derived dendritic cells in vitro, which in turn enhanced their ability to restimulate myelin-specific T cells. This was mirrored in vivo, in which depletion of neutrophils specifically impaired maturation of microglia and macrophages into professional APCs, resulting in a diminished amplification of early CNS inflammation. Therefore, inside the CNS neutrophils provide local cofactors that are required for the maturation of myeloid cells into professional APCs representing an essential step for the local restimulation of myelin-specific T cells and the development of autoimmune disease.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Sistema Nervoso Central/imunologia , Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Neutrófilos/imunologia , Linfócitos T/imunologia , Animais , Apresentação de Antígeno/imunologia , Autoimunidade/imunologia , Células Cultivadas , Citocinas/metabolismo , Inflamação/imunologia , Elastase de Leucócito/genética , Elastase de Leucócito/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/imunologia , Microglia/metabolismo , Esclerose Múltipla/imunologia , Peroxidase/genética , Peroxidase/metabolismo
8.
Blood ; 120(18): 3793-802, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22976954

RESUMO

The devastating effect of ischemic stroke is attenuated in mice lacking conventional and unconventional T cells, suggesting that inflammation enhances tissue damage in cerebral ischemia. We explored the functional role of αß and γδ T cells in a murine model of stroke and distinguished 2 different T cell-dependent proinflammatory pathways in ischemia-reperfusion injury. IFN-γ produced by CD4(+) T cells induced TNF-α production in macrophages, whereas IL-17A secreted by γδ T cells led to neutrophil recruitment. The synergistic effect of TNF-α and IL-17A on astrocytes resulted in enhanced secretion of CXCL-1, a neutrophil chemoattractant. Application of an IL-17A-blocking antibody within 3 hours after stroke induction decreased infarct size and improved neurologic outcome in the murine model. In autoptic brain tissue of patients who had a stroke, we detected IL-17A-positive lymphocytes, suggesting that this aspect of the inflammatory cascade is also relevant in the human brain. We propose that selective targeting of IL-17A signaling might provide a new therapeutic option for the treatment of stroke.


Assuntos
Interleucina-17/imunologia , Infiltração de Neutrófilos/imunologia , Transdução de Sinais/imunologia , Acidente Vascular Cerebral/imunologia , Linfócitos T/imunologia , Animais , Isquemia Encefálica/imunologia , Isquemia Encefálica/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neutrófilos/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Acidente Vascular Cerebral/metabolismo , Linfócitos T/metabolismo
9.
Elife ; 112022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35578890

RESUMO

Thermal adaptation is an extensively used intervention for enhancing or suppressing thermogenic and mitochondrial activity in adipose tissues. As such, it has been suggested as a potential lifestyle intervention for body weight maintenance. While the metabolic consequences of thermal acclimation are not limited to the adipose tissues, the impact on the rest of the tissues in context of their gene expression profile remains unclear. Here, we provide a systematic characterization of the effects in a comparative multi-tissue RNA sequencing approach following exposure of mice to 10 °C, 22 °C, or 34 °C in a panel of organs consisting of spleen, bone marrow, spinal cord, brain, hypothalamus, ileum, liver, quadriceps, subcutaneous-, visceral- and brown adipose tissues. We highlight that transcriptional responses to temperature alterations exhibit a high degree of tissue-specificity both at the gene level and at GO enrichment gene sets, and show that the tissue-specificity is not directed by the distinct basic gene expression pattern exhibited by the various organs. Our study places the adaptation of individual tissues to different temperatures in a whole-organism framework and provides integrative transcriptional analysis necessary for understanding the temperature-mediated biological programming.


Humans, mice and most other mammals are constantly exposed to fluctuations in the temperature of their environment. These fluctuations cause striking metabolic effects in the body, for example, exposure to cold promotes burning of calories to generate heat, thereby reducing how much fat accumulates in the body. On the other hand, warmer temperatures strengthen the bones and protect against a bone disease known as osteoporosis. As such, it has been suggested that exposure to alternating warm or cold temperatures could be a potential lifestyle intervention that conveys various benefits to our health. Our body stores fat in tissues known as adipose tissues, which are found all over the body including under the skin and around our major organs and muscles. Exposure to cold triggers changes in the activities of some genes in the adipose tissues to burn more calories. But it remains unclear how temperature affects the activities of other organs with respect to their expression of genes in the whole-body context. Hadadi, Spiljar et al. used an RNA sequencing approach to study the activities of genes in various tissues of mice exposed to cold (10°C), room temperature (22°C), or mild warm (34°C). The experiments revealed numerous genes whose levels were different in the various organs and temperatures tested. Overall, adipose tissues experienced the biggest changes in gene levels between different temperatures, followed by tissues involved in immune responses, and the brain and spinal cord tissues. Each organ changed gene expression levels in its own way. , and this was not due to the different intimate gene expression profile between the various organs. These findings improve our understanding of how changes in temperature affect mammals by putting the responses of individual tissues into the context of the whole body. Hadadi, Spiljar et al. also generated a web-based, free-to-use application to allow others to view and further analyze the data collected in this work for gene levels in the various organs of interest.


Assuntos
Temperatura Baixa , Transcriptoma , Aclimatação/genética , Tecido Adiposo Marrom/metabolismo , Animais , Camundongos , Termogênese
10.
Sci Transl Med ; 14(640): eabl6058, 2022 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-35417190

RESUMO

In chronic inflammatory diseases of the central nervous system (CNS), immune cells persisting behind the blood-brain barrier are supposed to promulgate local tissue destruction. The drivers of such compartmentalized inflammation remain unclear, but tissue-resident memory T cells (TRM) represent a potentially important cellular player in this process. Here, we investigated whether resting CD8+ TRM persisting after cleared infection with attenuated lymphocytic choriomeningitis virus (LCMV) can initiate immune responses directed against cognate self-antigen in the CNS. We demonstrated that time-delayed conditional expression of the LCMV glycoprotein as neo-self-antigen by glia cells reactivated CD8+ TRM. Subsequently, CD8+ TRM expanded and initiated CNS inflammation and immunopathology in an organ-autonomous manner independently of circulating CD8+ T cells. However, in the absence of CD4+ T cells, TCF-1+ CD8+ TRM failed to expand and differentiate into terminal effectors. Similarly, in human demyelinating CNS autoimmune lesions, we found CD8+ T cells expressing TCF-1 that predominantly exhibited a TRM-like phenotype. Together, our study provides evidence for CD8+ TRM-driven CNS immunopathology and sheds light on why inflammatory processes may evade current immunomodulatory treatments in chronic autoimmune CNS conditions.


Assuntos
Linfócitos T CD8-Positivos , Memória Imunológica , Autoantígenos , Linfócitos T CD4-Positivos , Sistema Nervoso Central , Humanos , Inflamação , Vírus da Coriomeningite Linfocítica
11.
J Neuroinflammation ; 8: 113, 2011 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-21906273

RESUMO

BACKGROUND: Nogo-66 receptor NgR1 and its structural homologue NgR2 are binding proteins for a number of myelin-associated inhibitory factors. After neuronal injury, these inhibitory factors are responsible for preventing axonal outgrowth via their interactions with NgR1 and NgR2 expressed on neurons. In vitro, cells expressing NgR1/2 are inhibited from adhering to and spreading on a myelin substrate. Neuronal injury also results in the presence of dendritic cells (DCs) in the central nervous system, where they can come into contact with myelin debris. The exact mechanisms of interaction of immune cells with CNS myelin are, however, poorly understood. METHODS: Human DCs were differentiated from peripheral blood monocytes and mouse DCs were differentiated from wild type and NgR1/NgR2 double knockout bone marrow precursors. NgR1 and NgR2 expression were determined with quantitative real time PCR and immunoblot, and adhesion of cells to myelin was quantified. RESULTS: We demonstrate that human immature myeloid DCs express NgR1 and NgR2, which are then down-regulated upon maturation. Human mature DCs also adhere to a much higher extent to a myelin substrate than immature DCs. We observe the same effect when the cells are plated on Nogo-66-His (binding peptide for NgR1), but not on control proteins. Mature DCs taken from Ngr1/2 knockout mice adhere to a much higher extent to myelin compared to wild type mouse DCs. In addition, Ngr1/2 knockout had no effect on in vitro DC differentiation or phenotype. CONCLUSIONS: These results indicate that a lack of NgR1/2 expression promotes the adhesion of DCs to myelin. This interaction could be important in neuroinflammatory disorders such as multiple sclerosis in which peripheral immune cells come into contact with myelin debris.


Assuntos
Adesão Celular/fisiologia , Células Dendríticas/metabolismo , Proteínas da Mielina/metabolismo , Bainha de Mielina/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Diferenciação Celular , Citocinas/metabolismo , Células Dendríticas/citologia , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Humanos , Subpopulações de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/citologia , Monócitos/fisiologia , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , Proteínas da Mielina/genética , Bainha de Mielina/genética , Proteínas Nogo , Receptor Nogo 1 , Receptor Nogo 2 , Receptores Nogo , Isoformas de Proteínas/genética , Receptores de Superfície Celular/genética
12.
J Immunol ; 183(9): 5494-501, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19843935

RESUMO

Th17 cells are involved in the defense against bacteria and fungi and play a prominent role in the pathogenesis of autoimmune diseases, but research on human Th17 cells is hindered due to the lack of a surface marker. In this study, we report that a subset of human and mouse CD4(+) T cells as well as human Th17 T cell clones express IL-17A on their surface upon stimulation. Correlation of surface IL-17A expression with intracellular IL-17A production and with RORgammat mRNA expression identified surface IL-17A as a specific marker for human and mouse Th17 cells. Phenotype characterization of ex vivo CD4(+) IL-17A(+) cells showed that the chemokines CCR6 and CCR4, costimulatory molecules, as well as CD2 and CD49d were more prominently expressed on these cells than in surface IL-17A(-) cells, supporting the concept of Th17 cells as a potent inflammatory effector subtype. In addition, we generated human Th1, Th1/17 (producing both IFN-gamma and IL-17A), and Th17 T cell clones based on single cell sorting of surface IL-17A(-), IL-17A(int), and IL-17A(high) CD4(+) T cells, respectively, and showed the plasticity of the double producing clones to the cytokine milieu. The identification of surface IL-17A as a marker for Th17 cells should facilitate research on this subset.


Assuntos
Imunofenotipagem , Interleucina-17/biossíntese , Proteínas de Membrana/biossíntese , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Sequência de Aminoácidos , Animais , Biomarcadores/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células Clonais , Citocinas/fisiologia , Humanos , Imunofenotipagem/métodos , Interleucina-1/biossíntese , Ativação Linfocitária/imunologia , Camundongos , Dados de Sequência Molecular
13.
Cell Metab ; 33(11): 2231-2246.e8, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34687652

RESUMO

Autoimmunity is energetically costly, but the impact of a metabolically active state on immunity and immune-mediated diseases is unclear. Ly6Chi monocytes are key effectors in CNS autoimmunity with an elusive role in priming naive autoreactive T cells. Here, we provide unbiased analysis of the immune changes in various compartments during cold exposure and show that this energetically costly stimulus markedly ameliorates active experimental autoimmune encephalomyelitis (EAE). Cold exposure decreases MHCII on monocytes at steady state and in various inflammatory mouse models and suppresses T cell priming and pathogenicity through the modulation of monocytes. Genetic or antibody-mediated monocyte depletion or adoptive transfer of Th1- or Th17-polarized cells for EAE abolishes the cold-induced effects on T cells or EAE, respectively. These findings provide a mechanistic link between environmental temperature and neuroinflammation and suggest competition between cold-induced metabolic adaptations and autoimmunity as energetic trade-off beneficial for the immune-mediated diseases.


Assuntos
Encefalomielite Autoimune Experimental , Doenças Neuroinflamatórias , Transferência Adotiva , Animais , Autoimunidade , Camundongos , Camundongos Endogâmicos C57BL , Células Th17
14.
Stroke ; 40(5): 1849-57, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19265055

RESUMO

BACKGROUND AND PURPOSE: Ischemic stroke leads to significant morbidity and mortality in the Western world. Early reperfusion strategies remain the treatment of choice but can initiate and augment an inflammatory response causing secondary brain damage. The understanding of postischemic inflammation is very limited. The objectives of this study were to define the temporal and spatial infiltration of immune cell populations and their activation patterns in a murine cerebral ischemia-reperfusion injury model. METHODS: Transient middle cerebral artery occlusion was induced for 1 hour followed by 12-hour to 7-day reperfusion in C57/BL6 mice. Immunohistochemistry and flow cytometry were used to quantify the infiltrating immune cell subsets. RESULTS: Accumulation of microglia and infiltration of the ischemic hemisphere by macrophages, lymphocytes, and dendritic cells (DCs) preceded the neutrophilic influx. DCs were found to increase 20-fold and constituted a substantial proportion of infiltrating cells. DCs exhibited a significant upregulation of major histocompatibility complex II and major histocompatibility complex II high-expressing DCs were found 100 times more abundant than in sham conditions. Upregulation of the costimulatory molecule CD80 was observed in DCs and microglial cells but did not further increase in major histocompatibility complex II high-expressing DCs. No lymphocyte activation was observed. Additionally, regulatory immune cells (natural killer T-cells, CD4(-)/CD8(-)T lymphocytes) cumulated in the ischemic hemisphere. CONCLUSIONS: This study provides a detailed analysis of the temporal dynamics of immune cell accumulation in a rodent stroke model. The peculiar activation pattern and massive increase of antigen-presenting cells in temporal conjunction with regulatory cells might provide additional insight into poststroke immune regulation.


Assuntos
Imunidade Celular/imunologia , Acidente Vascular Cerebral/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Dendríticas/imunologia , Citometria de Fluxo , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/imunologia , Infarto da Artéria Cerebral Média/patologia , Inflamação/patologia , Células Matadoras Naturais/imunologia , Cinética , Linfócitos/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Neutrófilos/imunologia , Traumatismo por Reperfusão/imunologia , Acidente Vascular Cerebral/patologia
15.
Brain Behav Immun ; 23(6): 861-7, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19362139

RESUMO

Inflammation within the Central Nervous System (CNS) is largely controlled by the balance between CNS-specific effector and regulatory T lymphocytes. To suppress CNS-inflammation in an antigen-specific manner, CNS-specific effector and regulatory T cells thus have to be differentially regulated. We employed recombinant peptide/MHC class II tetramers to assess CNS-specific effector and regulatory T cells during the specific suppression of myelin proteolipid protein aa139-151 (PLP139-151)-induced experimental autoimmune encephalomyelitis (EAE) by intravenous injection of recombinant invariant chains (Ii) in which the CLIP region has been replaced by the PLP139-151 epitope (Ii-PLP139-151). Injection of Ii-PLP139-151 induced apoptosis in CNS-specific effector T cells. In contrast, the proportion of specific regulatory T cells was increased and these cells expressed larger amounts of molecules that mediate regulatory T cell function including transforming growth factor beta and the inducible costimulator (ICOS). Consequently, regulatory T cells from Ii-treated mice were more potent than regulatory T cells from control-treated animals in suppressing effector T cell proliferation. These data demonstrate that effector T cells and regulatory T cells directed against the same CNS-antigen can be differentially regulated in vivo to suppress CNS-autoimmunity. Recombinant Ii induce apoptosis in CNS-specific effector T cells and provoke qualitative changes in specific regulatory T cells that enhance their immunosuppressive properties.


Assuntos
Sistema Nervoso Central/citologia , Sistema Nervoso Central/imunologia , Tolerância Imunológica/fisiologia , Proteínas Recombinantes/farmacologia , Linfócitos T Reguladores/imunologia , Linfócitos T/imunologia , Transferência Adotiva , Animais , Apoptose/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Caspase 3/biossíntese , Caspase 3/genética , Encefalomielite Autoimune Experimental/imunologia , Citometria de Fluxo , Genes MHC da Classe II/imunologia , Linfonodos/citologia , Linfonodos/efeitos dos fármacos , Camundongos , Proteínas Recombinantes/química , Células Th2/imunologia
16.
Sci Transl Med ; 11(498)2019 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-31243152

RESUMO

Epidemiological studies associate viral infections during childhood with the risk of developing autoimmune disease during adulthood. However, the mechanistic link between these events remains elusive. We report that transient viral infection of the brain in early life, but not at a later age, precipitates brain autoimmune disease elicited by adoptive transfer of myelin-specific CD4+ T cells at sites of previous infection in adult mice. Early-life infection of mouse brains imprinted a chronic inflammatory signature that consisted of brain-resident memory T cells expressing the chemokine (C-C motif) ligand 5 (CCL5). Blockade of CCL5 signaling via C-C chemokine receptor type 5 prevented the formation of brain lesions in a mouse model of autoimmune disease. In mouse and human brain, CCL5+ TRM were located predominantly to sites of microglial activation. This study uncovers how transient brain viral infections in a critical window in life might leave persisting chemotactic cues and create a long-lived permissive environment for autoimmunity.


Assuntos
Doenças Autoimunes/imunologia , Encéfalo/imunologia , Memória Imunológica , Linfócitos T/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Quimiocina CCL5/metabolismo , Suscetibilidade a Doenças , Antígenos HLA-DR/metabolismo , Humanos , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia
17.
Front Immunol ; 9: 2827, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30555489

RESUMO

Tissue-resident-memory CD8+ T cells (TRM) have been described as a non-circulating memory T cell subset that persists at sites of previous infection. While TRM in all non-lymphoid organs probably share a core signature differentiation pathway, certain aspects of their maintenance and effector functions may vary. It is well-established that TRM provide long-lived protective immunity through immediate effector function and accelerated recruitment of circulating immune cells. Besides immune defense against pathogens, other immunological roles of TRM are less well-studied. Likewise, evidence of a putative detrimental role of TRM for inflammatory diseases is only beginning to emerge. In this review, we discuss the protective and harmful role of TRM in organ-specific immunity and immunopathology as well as prospective implications for immunomodulatory therapy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica , Animais , Humanos , Inflamação/imunologia , Especificidade de Órgãos/imunologia
18.
Int J Dev Neurosci ; 24(1): 65-72, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16338115

RESUMO

During the development of the central nervous system (CNS), the correct wiring of outgrowing neurites is mediated by antagonistic mechanisms. Aberrant growth is prevented by repulsive factors such as semaphorins. Expression of the ligands Sema3A and -3E and the receptors neuropilin Npn-1, -2a and -2b in the chick visual system were analyzed by RT-PCR. Whereas Sema3A and its major receptor Npn-1 were abundant, Sema3E and Npn-2 isoform expression was highly restricted and developmentally regulated. Peak expression occurred during retinal axon innervation of the tectum. Functional in vitro assays with recombinant proteins revealed a topography-specific growth cone collapsing activity of Sema3A for tectal axons. Interestingly, whereas tectal axons collapsed in a topographic-specific manner only in the presence of Sema3A, retinal axons responded only to Sema3E. The collapsing activity was intracellularly mediated by cGMP. For a detailed analysis of neuronal responses to sempahorins, time lapse video recording was performed. When tectal and retinal axons were pre-exposed to brain-derived neurotrophic factor (BDNF), a protective effect was evident only in the case of retinal axons. Our results suggest a molecular mechanism whereby ingrowth of retinal axons into the tectum can be regulated by Sema3E/BDNF modulation without disturbing tectal axon growth out of the tectum mediated by Sema3A.


Assuntos
Proteínas Aviárias/metabolismo , Axônios/metabolismo , Sistema Nervoso Central/citologia , Semaforina-3A/metabolismo , Semaforinas/metabolismo , Animais , Proteínas Aviárias/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Embrião de Galinha , Vias Neurais/citologia , Vias Neurais/metabolismo , Neuropilina-1/genética , Neuropilina-1/metabolismo , Neuropilina-2/genética , Neuropilina-2/metabolismo , Nucleotídeos Cíclicos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/metabolismo , Semaforina-3A/genética , Semaforinas/genética , Transdução de Sinais/fisiologia , Colículos Superiores/citologia , Colículos Superiores/metabolismo
19.
Methods Mol Biol ; 1304: 189-209, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25146304

RESUMO

Neuropathological techniques such as conventional and immunohistochemical staining of paraffin-embedded tissue sections are instrumental for identification and characterization of aberrations of organ architecture during human inflammatory disorders of the central nervous system (CNS) as in their animal models. Here we describe step-by-step protocols for tissue processing, sectioning, and conventional and immunohistochemical stainings to display as well as quantify CNS inflammation, demyelination, and neuronal damage in experimental autoimmune encephalomyelitis (EAE), an animal model of Multiple Sclerosis (MS).


Assuntos
Encefalomielite Autoimune Experimental/patologia , Animais , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Imuno-Histoquímica/métodos , Camundongos , Esclerose Múltipla/patologia
20.
J Exp Med ; 213(8): 1571-87, 2016 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-27377586

RESUMO

Tissue-resident memory T cells (TRM) persist at sites of prior infection and have been shown to enhance pathogen clearance by recruiting circulating immune cells and providing bystander activation. Here, we characterize the functioning of brain-resident memory T cells (bTRM) in an animal model of viral infection. bTRM were subject to spontaneous homeostatic proliferation and were largely refractory to systemic immune cell depletion. After viral reinfection in mice, bTRM rapidly acquired cytotoxic effector function and prevented fatal brain infection, even in the absence of circulating CD8(+) memory T cells. Presentation of cognate antigen on MHC-I was essential for bTRM-mediated protective immunity, which involved perforin- and IFN-γ-dependent effector mechanisms. These findings identify bTRM as an organ-autonomous defense system serving as a paradigm for TRM functioning as a self-sufficient first line of adaptive immunity.


Assuntos
Encéfalo/imunologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Memória Imunológica , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Linfócitos T CD8-Positivos/patologia , Interferon gama/genética , Interferon gama/imunologia , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/patologia , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA