Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Acta Neuropathol ; 145(3): 335-355, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36695896

RESUMO

B cells contribute to the pathogenesis of both cellular- and humoral-mediated central nervous system (CNS) inflammatory diseases through a variety of mechanisms. In such conditions, B cells may enter the CNS parenchyma and contribute to local tissue destruction. It remains unexplored, however, how infection and autoimmunity drive transcriptional phenotypes, repertoire features, and antibody functionality. Here, we profiled B cells from the CNS of murine models of intracranial (i.c.) viral infections and autoimmunity. We identified a population of clonally expanded, antibody-secreting cells (ASCs) that had undergone class-switch recombination and extensive somatic hypermutation following i.c. infection with attenuated lymphocytic choriomeningitis virus (rLCMV). Recombinant expression and characterisation of these antibodies revealed specificity to viral antigens (LCMV glycoprotein GP), correlating with ASC persistence in the brain weeks after resolved infection. Furthermore, these virus-specific ASCs upregulated proliferation and expansion programs in response to the conditional and transient induction of the LCMV GP as a neo-self antigen by astrocytes. This class-switched, clonally expanded, and mutated population persisted and was even more pronounced when peripheral B cells were depleted prior to autoantigen induction in the CNS. In contrast, the most expanded B cell clones in mice with persistent expression of LCMV GP in the CNS did not exhibit neo-self antigen specificity, potentially a consequence of local tolerance induction. Finally, a comparable population of clonally expanded, class-switched, and proliferating ASCs was detected in the cerebrospinal fluid of relapsing multiple sclerosis (RMS) patients. Taken together, our findings support the existence of B cells that populate the CNS and are capable of responding to locally encountered autoantigens.


Assuntos
Células Produtoras de Anticorpos , Autoantígenos , Camundongos , Animais , Linfócitos B , Vírus da Coriomeningite Linfocítica , Encéfalo
2.
Genes Immun ; 23(6): 183-195, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36028771

RESUMO

Adaptive immune repertoires are composed by the ensemble of B and T-cell receptors within an individual, reflecting both past and current immune responses. Recent advances in single-cell sequencing enable recovery of the complete adaptive immune receptor sequences in addition to transcriptional information. Here, we recovered transcriptome and immune repertoire information for polyclonal T follicular helper cells following lymphocytic choriomeningitis virus (LCMV) infection, CD8+ T cells with binding specificity restricted to two distinct LCMV peptides, and B and T cells isolated from the nervous system in the context of experimental autoimmune encephalomyelitis. We could relate clonal expansion, germline gene usage, and clonal convergence to cell phenotypes spanning activation, memory, naive, antibody secretion, T-cell inflation, and regulation. Together, this dataset provides a resource for immunologists that can be integrated with future single-cell immune repertoire and transcriptome sequencing datasets.


Assuntos
Autoimunidade , Coriomeningite Linfocítica , Animais , Linfócitos T CD8-Positivos , Modelos Animais de Doenças , Coriomeningite Linfocítica/genética , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos , Receptores de Antígenos de Linfócitos T/genética
3.
Curr Opin Immunol ; 77: 102211, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35644112

RESUMO

Tissue-resident memory T cells (TRM) represent a subset of antigen-experienced T cells that are constantly retained in a given tissue with limited trafficking through the circulation. These cells are characterized by expression of molecules enabling their tissue anchoring and downregulation of molecules promoting tissue egress. They reside at sites of previous antigen encounter and their number increases with age. TRM have been shown to provide rapid and efficient protection against tissue reinfection and TRM density correlates with efficient antitumor responses. Intriguingly, the density of CD8 TRM is increased in the central nervous system (CNS) of patients with neuroinflammatory diseases such as multiple sclerosis, or suffering from neurodegenerative diseases. In this review, we discuss current knowledge regarding the diversity of CNS-resident CD8 T cells and their role in CNS autoimmunity. Given their likely contribution to the protracted course of several inflammatory diseases of the CNS, their therapeutic targeting becomes an important challenge.


Assuntos
Linfócitos T CD8-Positivos , Memória Imunológica , Antígenos/metabolismo , Sistema Nervoso Central , Crime , Humanos
4.
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
5.
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
6.
Cell Rep Med ; 2(3): 100209, 2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33763654

RESUMO

Therapeutic vaccination regimens inducing clinically effective tumor-specific CD8+ T lymphocyte (CTL) responses are an unmet medical need. We engineer two distantly related arenaviruses, Pichinde virus and lymphocytic choriomeningitis virus, for therapeutic cancer vaccination. In mice, life-replicating vector formats of these two viruses delivering a self-antigen in a heterologous prime-boost regimen induce tumor-specific CTL responses up to 50% of the circulating CD8 T cell pool. This CTL attack eliminates established solid tumors in a significant proportion of animals, accompanied by protection against tumor rechallenge. The magnitude of CTL responses is alarmin driven and requires combining two genealogically distantly related arenaviruses. Vector-neutralizing antibodies do not inhibit booster immunizations by the same vector or by closely related vectors. Rather, CTL immunodominance hierarchies favor vector backbone-targeted responses at the expense of self-reactive CTLs. These findings establish an arenavirus-based immunotherapy regimen that allows reshuffling of immunodominance hierarchies and breaking self-directed tolerance for efficient tumor control.


Assuntos
Vacinas Anticâncer/administração & dosagem , Imunoterapia/métodos , Vírus da Coriomeningite Linfocítica/imunologia , Mastocitoma/terapia , Vírus Pichinde/imunologia , Linfócitos T Citotóxicos/imunologia , Alarminas/genética , Alarminas/imunologia , Animais , Anticorpos Neutralizantes/farmacologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Feminino , Expressão Gênica , Engenharia Genética/métodos , Vetores Genéticos/classificação , Vetores Genéticos/imunologia , Cobaias , Imunização Secundária , Vírus da Coriomeningite Linfocítica/classificação , Vírus da Coriomeningite Linfocítica/genética , Mastocitoma/genética , Mastocitoma/imunologia , Mastocitoma/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Filogenia , Vírus Pichinde/classificação , Vírus Pichinde/genética , Tolerância a Antígenos Próprios , Análise de Sobrevida , Vacinação/métodos
7.
Curr Opin Virol ; 47: 68-78, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33636592

RESUMO

Protective immune responses in the central nervous system (CNS) must act efficiently but need to be tightly controlled to avoid excessive damage to this vital organ. Under homeostatic conditions, the immune surveillance of the CNS is mediated by innate immune cells together with subsets of memory lymphocytes accumulating over lifetime. Accordingly, a wide range of immune responses can be triggered upon pathogen infection that can be associated with devastating clinical outcomes, and which most frequently are due to neurotropic viruses. Here, we discuss recent advances about our understanding of anti-viral immune responses with special emphasis on mechanisms operating in the various anatomical compartments of the CNS.


Assuntos
Sistema Nervoso Central/imunologia , Sistema Nervoso Central/virologia , Imunidade Adaptativa , Animais , Linfócitos B/imunologia , Sistema Nervoso Central/anatomia & histologia , Sistema Nervoso Central/patologia , Humanos , Imunidade Inata , Vigilância Imunológica , Linfócitos T/imunologia , Vírus/classificação , Vírus/patogenicidade
8.
Nat Commun ; 12(1): 1009, 2021 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-33579927

RESUMO

Self-reactive CD8+ T cells are important mediators of progressive tissue damage in autoimmune diseases, but the molecular program underlying these cells' functional adaptation is unclear. Here we characterize the transcriptional and epigenetic landscape of self-reactive CD8+ T cells in a mouse model of protracted central nervous system (CNS) autoimmunity and compare it to populations of CNS-resident memory CD8+ T cells emerging from acute viral infection. We find that autoimmune CD8+ T cells persisting at sites of self-antigen exhibit characteristic transcriptional regulation together with distinct epigenetic remodeling. This self-reactive CD8+ T cell fate depends on the transcriptional regulation by the DNA-binding HMG-box protein TOX which remodels more than 400 genomic regions including loci such as Tcf7, which is central to stemness of CD8+ T cells. Continuous exposure to CNS self-antigen sustains TOX levels in self-reactive CD8+ T cells, whereas genetic ablation of TOX in CD8+ T cells results in shortened persistence of self-reactive CD8+ T cells in the inflamed CNS. Our study establishes and characterizes the genetic differentiation program enabling chronic T cell-driven immunopathology in CNS autoimmunity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Sistema Nervoso Central/metabolismo , Montagem e Desmontagem da Cromatina , Animais , Autoimunidade/imunologia , Feminino , Regulação da Expressão Gênica , Memória Imunológica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
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
11.
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
12.
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
13.
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
14.
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
15.
J Mol Biol ; 426(4): 936-44, 2014 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-24374182

RESUMO

The gene encoding the p53 tumor suppressor protein, a sequence-specific DNA binding transcription factor, is the most frequently mutated gene in human cancer. Crystal structures of homo-oligomerizing p53 polypeptides with specific DNA suggest that DNA binding is associated with a conformational switch. Specifically, in the absence of DNA, loop L1 of the p53 DNA binding domain adopts an extended conformation, whereas two p53 subunits switch to a recessed loop L1 conformation when bound to DNA as a tetramer. We previously designed a p53 protein, p53FG, with amino substitutions S121F and V122G targeting loop L1. These two substitutions enhanced the affinity of p53 for specific DNA yet, counterintuitively, decreased the residency time of p53 on DNA. Here, we confirmed these DNA binding properties of p53FG using a different method. We also determined by crystallography the structure of p53FG in its free state and bound to DNA as a tetramer. In the free state, loop L1 adopted a recessed conformation, whereas upon DNA binding, two subunits switched to the extended loop L1 conformation, resulting in a final structure that was very similar to that of wild-type p53 bound to DNA. Thus, altering the apo structure of p53 changed its DNA binding properties, even though the DNA-bound structure was not altered.


Assuntos
Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/metabolismo , Substituição de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Inibidor de Quinase Dependente de Ciclina p21/genética , DNA/química , DNA/metabolismo , Meia-Vida , Humanos , Modelos Moleculares , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Elementos de Resposta , Ressonância de Plasmônio de Superfície , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...