Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 55(11): 1978-1980, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36351372

RESUMO

RORγt+ regulatory T (Treg) cells are critical toward maintaining gut immune tolerance. In recent studies published in Nature, Kedmi et al., Lyu et al., and Akagbosu et al. describe MHCII+RORγt+ antigen-presenting cells that mediate RORγt+ Treg cell differentiation but propose disparate identities for these cells.


Assuntos
Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Tolerância Periférica , Constrangimento , Linfócitos T Reguladores , Células Apresentadoras de Antígenos , Células Th17 , Fatores de Transcrição Forkhead , Tolerância Imunológica
2.
Nat Immunol ; 18(7): 716-724, 2017 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-28632714

RESUMO

Efforts to understand autoimmunity have been pursued relentlessly for several decades. It has become apparent that the immune system evolved multiple mechanisms for controlling self-reactivity, and defects in one or more of these mechanisms can lead to a breakdown of tolerance. Among the multitude of lesions associated with disease, the most common seem to affect peripheral tolerance rather than central tolerance. The initial trigger for both systemic autoimmune disorders and organ-specific autoimmune disorders probably involves the recognition of self or foreign molecules, especially nucleic acids, by innate sensors. Such recognition, in turn, triggers inflammatory responses and the engagement of previously quiescent autoreactive T cells and B cells. Here we summarize the most prominent autoimmune pathways and identify key issues that require resolution for full understanding of pathogenic autoimmunity.


Assuntos
Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Linfócitos B/imunologia , Tolerância a Antígenos Próprios/imunologia , Linfócitos T/imunologia , Animais , Tolerância Central/imunologia , Humanos , Tolerância Periférica/imunologia
3.
Nat Immunol ; 17(6): 618-25, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27196520

RESUMO

The bidirectional interaction between the immune system and whole-body metabolism has been well recognized for many years. Via effects on adipocytes and hepatocytes, immune cells can modulate whole-body metabolism (in metabolic syndromes such as type 2 diabetes and obesity) and, reciprocally, host nutrition and commensal-microbiota-derived metabolites modulate immunological homeostasis. Studies demonstrating the metabolic similarities of proliferating immune cells and cancer cells have helped give birth to the new field of immunometabolism, which focuses on how the cell-intrinsic metabolic properties of lymphocytes and macrophages can themselves dictate the fate and function of the cells and eventually shape an immune response. We focus on this aspect here, particularly as it relates to regulatory T cells.


Assuntos
Adipócitos/imunologia , Diabetes Mellitus Tipo 2/imunologia , Macrófagos/metabolismo , Síndrome Metabólica/imunologia , Obesidade/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Homeostase , Humanos , Imunidade , Microbiota , Tolerância Periférica
4.
Nat Immunol ; 17(3): 304-14, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26829766

RESUMO

The role of anergy, an acquired state of T cell functional unresponsiveness, in natural peripheral tolerance remains unclear. In this study, we found that anergy was selectively induced in fetal antigen-specific maternal CD4(+) T cells during pregnancy. A naturally occurring subpopulation of anergic polyclonal CD4(+) T cells, enriched for self antigen-specific T cell antigen receptors, was also present in healthy hosts. Neuropilin-1 expression in anergic conventional CD4(+) T cells was associated with hypomethylation of genes related to thymic regulatory T cells (Treg cells), and this correlated with their ability to differentiate into Foxp3(+) Treg cells that suppressed immunopathology. Thus, our data suggest that not only is anergy induction important in preventing autoimmunity but also it generates the precursors for peripheral Treg cell differentiation.


Assuntos
Autoimunidade/imunologia , Diferenciação Celular/imunologia , Anergia Clonal/imunologia , Histocompatibilidade Materno-Fetal/imunologia , Tolerância Periférica/imunologia , Células Precursoras de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Artrite Experimental/imunologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Citocinas/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Fatores de Transcrição Forkhead/imunologia , Genes Codificadores da Cadeia alfa de Receptores de Linfócitos T , Immunoblotting , Masculino , Camundongos , Camundongos Knockout , Neuropilina-1/metabolismo , Gravidez , Receptores de Antígenos de Linfócitos T/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tolerância a Antígenos Próprios , Timócitos/imunologia
5.
Nat Immunol ; 17(3): 297-303, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26752376

RESUMO

Inflammation induced during infection can both promote and suppress immunity. This contradiction suggests that inflammatory cytokines affect the immune system in a context-dependent manner. Here we show that nonspecific bystander inflammation conditions naive CD4(+) T cells for enhanced peripheral Foxp3 induction and reduced effector differentiation. This results in inhibition of immune responses in vivo via a Foxp3-dependent effect on antigen-specific naive CD4(+) T cell precursors. Such conditioning may have evolved to allow immunity to infection while limiting subsequent autoimmunity caused by release of self-antigens in the wake of infection. Furthermore, this phenomenon suggests a mechanistic explanation for the idea that early tuning of the immune system by infection affects the long-term quality of immune regulation.


Assuntos
Asma/imunologia , Autoimunidade/imunologia , Efeito Espectador/imunologia , Linfócitos T CD4-Positivos/imunologia , Citocinas/imunologia , Diabetes Mellitus/imunologia , Fatores de Transcrição Forkhead/imunologia , Inflamação , Tolerância a Antígenos Próprios/imunologia , Animais , Autoantígenos , Efeito Espectador/efeitos dos fármacos , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linhagem Celular Tumoral , Citocinas/efeitos dos fármacos , Citocinas/farmacologia , Metilação de DNA , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Fatores de Transcrição Forkhead/genética , Perfilação da Expressão Gênica , Indutores de Interferon/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tolerância Periférica/imunologia , Poli I-C/farmacologia , Regiões Promotoras Genéticas , Linfopoietina do Estroma do Timo
6.
Nat Immunol ; 16(11): 1162-73, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26437242

RESUMO

Receptors of the Notch family direct the differentiation of helper T cell subsets, but their influence on regulatory T cell (T(reg) cell) responses is obscure. We found here that lineage-specific deletion of components of the Notch pathway enhanced T(reg) cell-mediated suppression of type 1 helper T cell (T(H)1 cell) responses and protected against their T(H)1 skewing and apoptosis. In contrast, expression in T(reg) cells of a gain-of-function transgene encoding the Notch1 intracellular domain resulted in lymphoproliferation, exacerbated T(H)1 responses and autoimmunity. Cell-intrinsic canonical Notch signaling impaired T(reg) cell fitness and promoted the acquisition by T(reg) cells of a T(H)1 cell-like phenotype, whereas non-canonical Notch signaling dependent on the adaptor Rictor activated the kinase AKT-transcription factor Foxo1 axis and impaired the epigenetic stability of Foxp3. Our findings establish a critical role for Notch signaling in controlling peripheral T(reg) cell function.


Assuntos
Tolerância Periférica , Receptor Notch1/imunologia , Linfócitos T Reguladores/imunologia , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Epigênese Genética , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação , Proteína Companheira de mTOR Insensível à Rapamicina , Receptor Notch1/deficiência , Receptor Notch1/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Transdução de Sinais/imunologia , Células Th1/imunologia , Transcriptoma
7.
Proc Natl Acad Sci U S A ; 121(11): e2318599121, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38446856

RESUMO

T cells help orchestrate immune responses to pathogens, and their aberrant regulation can trigger autoimmunity. Recent studies highlight that a threshold number of T cells (a quorum) must be activated in a tissue to mount a functional immune response. These collective effects allow the T cell repertoire to respond to pathogens while suppressing autoimmunity due to circulating autoreactive T cells. Our computational studies show that increasing numbers of pathogenic peptides targeted by T cells during persistent or severe viral infections increase the probability of activating T cells that are weakly reactive to self-antigens (molecular mimicry). These T cells are easily re-activated by the self-antigens and contribute to exceeding the quorum threshold required to mount autoimmune responses. Rare peptides that activate many T cells are sampled more readily during severe/persistent infections than in acute infections, which amplifies these effects. Experiments in mice to test predictions from these mechanistic insights are suggested.


Assuntos
Doenças Autoimunes , Infecção Persistente , Animais , Camundongos , Tolerância Periférica , Linfócitos T , Autoantígenos , Peptídeos
8.
Proc Natl Acad Sci U S A ; 120(16): e2300099120, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-37040412

RESUMO

B cells that bind soluble autoantigens receive chronic signaling via the B cell receptor (signal-1) in the absence of strong costimulatory signals (signal-2), and this leads to their elimination in peripheral tissues. The factors determining the extent of soluble autoantigen-binding B cell elimination are not fully understood. Here we demonstrate that the elimination of B cells chronically exposed to signal-1 is promoted by cathepsin B (Ctsb). Using hen egg lysozyme-specific (HEL-specific) immunoglobulin transgenic (MD4) B cells and mice harboring circulating HEL, we found improved survival and increased proliferation of HEL-binding B cells in Ctsb-deficient mice. Bone marrow chimera experiments established that both hematopoietic and nonhematopoietic sources of Ctsb were sufficient to promote peripheral B cell deletion. The depletion of CD4+ T cells overcame the survival and growth advantage provided by Ctsb deficiency, as did blocking CD40L or removing CD40 from the chronically antigen-engaged B cells. Thus, we suggest that Ctsb acts extracellularly to reduce soluble autoantigen-binding B cell survival and that its actions restrain CD40L-dependent pro-survival effects. These findings identify a role for cell-extrinsic protease activity in establishing a peripheral self-tolerance checkpoint.


Assuntos
Peptídeo Hidrolases , Tolerância Periférica , Camundongos , Animais , Camundongos Transgênicos , Ligante de CD40 , Catepsina B , Camundongos Endogâmicos C57BL , Autoantígenos
9.
Immunity ; 45(2): 305-18, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27533013

RESUMO

Dendritic cells (DCs) are instrumental in the initiation of T cell responses, but how thymic and peripheral tolerogenic DCs differ globally from Toll-like receptor (TLR)-induced immunogenic DCs remains unclear. Here, we show that thymic XCR1(+) DCs undergo a high rate of maturation, accompanied by profound gene-expression changes that are essential for central tolerance and also happen in germ-free mice. Those changes largely overlap those occurring during tolerogenic and, more unexpectedly, TLR-induced maturation of peripheral XCR1(+) DCs, arguing against the commonly held view that tolerogenic DCs undergo incomplete maturation. Interferon-stimulated gene (ISG) expression was among the few discriminators of immunogenic and tolerogenic XCR1(+) DCs. Tolerogenic XCR1(+) thymic DCs were, however, unique in expressing ISGs known to restrain virus replication. Therefore, a broad functional convergence characterizes tolerogenic and immunogenic XCR1(+) DC maturation in the thymus and periphery, maximizing antigen presentation and signal delivery to developing and to conventional and regulatory mature T cells.


Assuntos
Tolerância Central , Células Dendríticas/imunologia , Tolerância Periférica , Linfócitos T Reguladores/imunologia , Timo/imunologia , Animais , Apresentação de Antígeno , Diferenciação Celular , Células Cultivadas , Fatores Reguladores de Interferon/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Quimiocinas/metabolismo , Receptores Toll-Like/imunologia , Transcriptoma , Replicação Viral
10.
Proc Natl Acad Sci U S A ; 119(15): e2120149119, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35394861

RESUMO

Immunological tolerance is established and maintained by a diverse array of safeguards that work together to protect against autoimmunity. Despite the identification of numerous tolerogenic processes, the basis for cooperation among them remains poorly understood. We sought to identify synergy among several well-defined tolerance mediators that alone provide protection only from mild autoimmune symptoms in C57BL/6 mice: BIM, AIRE, CBL-B, and PD-1. Survey of a range of compound mutant mice revealed that the combined loss of the autoimmune regulator, AIRE, with PD-1 unleashed a spontaneous, lethal autoimmune disease. Pdcd1−/−Aire−/− mice succumbed to cachexia before adulthood, with near-complete destruction of the exocrine pancreas. Such fatal autoimmunity was not observed in Pdcd1−/−Bim−/−, Bim−/−Aire−/−, or Cblb−/−Bim−/− mice, suggesting that the cooperation between AIRE-mediated and PD-1­mediated tolerance was particularly potent. Immune profiling revealed largely normal development of FOXP3+ regulatory T (Treg) cells in Pdcd1−/−Aire−/− mice, yet excessive, early activation of effector T cells. Adoptive transfer experiments demonstrated that autoimmune exocrine pancreatitis was driven by conventional CD4+ T cells and could not be prevented by the cotransfer of Treg cells from wild-type mice. The development of autoimmunity in mixed bone marrow chimeras supported these observations, indicating that failure of recessive tolerance was responsible for disease. These findings reveal a potent tolerogenic axis between AIRE and PD-1 that has implications for our understanding of how immune checkpoint blockade might synergize with subclinical defects in central tolerance to elicit autoimmune disease.


Assuntos
Pancreatite Autoimune , Tolerância Imunológica , Tolerância Periférica , Receptor de Morte Celular Programada 1 , Fatores de Transcrição , Animais , Pancreatite Autoimune/genética , Pancreatite Autoimune/imunologia , Autoimunidade/genética , Tolerância Imunológica/genética , Camundongos , Camundongos Endogâmicos C57BL , Tolerância Periférica/genética , Tolerância Periférica/imunologia , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/fisiologia , Timo/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Proteína AIRE
11.
Immunity ; 42(3): 471-83, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25786177

RESUMO

Self-reactive T cells can escape thymic deletion and therefore some of these potentially autoaggressive T cells need to convert into regulatory T (Treg) cells to help control responses against self. However, it remains unknown how peripheral self-reactive T cells are specifically instructed to become Treg cells. We report that CD5, whose expression is upregulated in T cells by self and tolerizing antigens in the thymus and periphery, governed extrathymic Treg cell development. CD5 modified effector cell-differentiating signals that inhibit Treg cell induction. Treg cell conversion of Cd5(-/-) and CD5(lo) T cells was inhibited by even small amounts of interleukin-4 (IL-4), IL-6, and interferon-γ (IFN-γ) produced by bystander lymphocytes, while CD5(hi) T cells resisted this inhibition of Treg cell induction. Our findings further revealed that CD5 promoted Treg cell induction by blocking mechanistic target of rapamycin (mTOR) activation. Therefore CD5 instructs extrathymic Treg cell development in response to self and tolerizing antigens.


Assuntos
Autoantígenos/imunologia , Antígenos CD5/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Autoantígenos/genética , Efeito Espectador/imunologia , Antígenos CD5/genética , Diferenciação Celular , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/farmacologia , Interleucina-4/genética , Interleucina-4/imunologia , Interleucina-4/farmacologia , Interleucina-6/genética , Interleucina-6/imunologia , Interleucina-6/farmacologia , Camundongos , Camundongos Knockout , Tolerância Periférica , Transdução de Sinais , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/imunologia , Timo/citologia , Timo/imunologia
12.
Mol Ther ; 30(2): 745-762, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-34450252

RESUMO

The major challenge in the treatment of autoimmune diseases is the restoration of the impaired peripheral immune tolerance that always accompanies the development of such diseases. Here, we show that small splenic peptides (SSPs) of whole spleen extract efficiently suppress the development of psoriatic arthritis in vivo, even in the presence of sustained levels of pro-inflammatory cytokines. SSPs target dendritic cells (DCs) and convert them into tolerogenic cells, which in turn differentiate naive CD4+ cells into Foxp3-expressing T regulatory cells (Tregs). The latter requires direct contact between SSP-activated DCs and naive CD4+ T cells via PD-1 and CTLA4 immune checkpoint receptors of T cells. Finally, depletion of Foxp3+ Tregs in vivo abrogated the protective effect of SSPs on psoriatic arthritis development. We hypothesize that SSPs represent an intrinsic component of the adaptive immune system responsible for the physiological maintenance of peripheral tolerance and that therapeutically administered SSPs are able to restore imbalanced peripheral tolerance in autoimmune diseases.


Assuntos
Artrite Psoriásica , Tolerância Imunológica , Artrite Psoriásica/terapia , Citocinas , Células Dendríticas , Humanos , Tolerância Periférica , Baço , Linfócitos T Reguladores
13.
Eur J Immunol ; 51(1): 39-55, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33275279

RESUMO

CD4+ CD25high CD127low/- FOXP3+ T regulatory cells are responsible for maintaining immune tolerance and controlling excessive immune responses. Treg cell use in pre-clinical animal models showed the huge therapeutic potential of these cells in immune-mediated diseases and laid the foundations for their applications in therapy in humans. Currently, there are several clinical trials utilizing the adoptive transfer of Treg cells to reduce the morbidity in autoimmune disorders, allogeneic HSC transplantation, and solid organ transplantation. However, a large part of them utilizes total Treg cells without distinction of their biological variability. Many studies on the heterogeneity of Treg cell population revealed distinct subsets with different functions in the control of the immune response and induction of peripheral tolerance. Some of these subsets also showed a role in controlling the general homeostasis of non-lymphoid tissues. All these Treg cell subsets and their peculiar properties can be therefore exploited to develop novel therapeutic approaches. This review describes these functionally distinct subsets, their phenotype, homing properties and functions in lymphoid and non-lymphoid tissues. In addition, we also discuss the limitations in using Treg cells as a cellular therapy and the strategies to enhance their efficacy.


Assuntos
Imunoterapia Adotiva/métodos , Linfócitos T Reguladores/classificação , Linfócitos T Reguladores/imunologia , Tecido Adiposo/citologia , Tecido Adiposo/imunologia , Aloenxertos , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/terapia , Fatores de Transcrição Forkhead/imunologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Tolerância Imunológica , Camundongos , Modelos Imunológicos , Tolerância Periférica , Imunologia de Transplantes , Cicatrização/imunologia
14.
J Immunol ; 205(5): 1239-1247, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32709661

RESUMO

A series of layered peripheral checkpoints maintain self-reactive B cells in an unresponsive state. Autoantibody production occurs when these checkpoints are breached; however, when and how this occurs is largely unknown. In particular, how self-reactive B cells are restrained during bystander inflammation in otherwise healthy individuals is poorly understood. A weakness has been the unavailability of methods capable of dissecting physiologically relevant B cell responses without the use of an engineered BCR. Resolving this will provide insights that decipher how this process goes awry during autoimmunity or could be exploited for therapy. In this study, we use a strong adjuvant to provide bystander innate and adaptive signals that promote B cell responsiveness in conjunction with newly developed B cell detection tools to study in detail the ways that peripheral tolerance mechanisms limit the expansion and function of self-reactive B cells activated under these conditions. We show that although self-reactive B cells are recruited into the germinal center, their development does not proceed, possibly because of rapid counterselection. Consequently, differentiation of plasma cells is blunted, and Ab responses are transient and devoid of affinity maturation. We propose this approach, and these tools can be more widely applied to track Ag-specific B cell responses to more disease-relevant Ags, without the need for BCR transgenic mice, in settings where tolerance pathways are compromised or have been genetically manipulated to drive stronger insights into the biology underlying B cell-mediated autoimmunity.


Assuntos
Formação de Anticorpos/imunologia , Linfócitos B/imunologia , Tolerância Imunológica/imunologia , Tolerância Periférica/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Animais , Autoanticorpos/imunologia , Autoantígenos/imunologia , Autoimunidade/imunologia , Diferenciação Celular/imunologia , Feminino , Centro Germinativo/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasmócitos/imunologia
15.
J Immunol ; 205(7): 1920-1932, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32868410

RESUMO

Sialyl Lewis X (sLeX) regulates T cell trafficking from the vasculature into skin and sites of inflammation, thereby playing a critical role in immunity. In healthy persons, only a small proportion of human blood T cells express sLeX, and their function is not fully defined. Using a combination of biochemical and functional studies, we find that human blood sLeX+CD4+T cells comprise a subpopulation expressing high levels of Th2 and Th17 cytokines, chemokine receptors CCR4 and CCR6, and the transcription factors GATA-3 and RORγT. Additionally, sLeX+CD4+T cells exclusively contain the regulatory T cell population (CD127lowCD25high and FOXP3+) and characteristically display immune-suppressive molecules, including the coinhibitor receptors PD-1 and CTLA-4. Among CD8+T cells, sLeX expression distinguishes a subset displaying low expression of cytotoxic effector molecules, perforin and granzyme ß, with reduced degranulation and CD57 expression and, consistently, marginal cytolytic capacity after TCR engagement. Furthermore, sLeX+CD8+T cells present a pattern of features consistent with Th cell-like phenotype, including release of pertinent Tc2 cytokines and elevated expression of CD40L. Together, these findings reveal that sLeX display is associated with unique functional specialization of both CD4+ and CD8+T cells and indicate that circulating T cells that are primed to migrate to lesional sites at onset of inflammation are not poised for cytotoxic function.


Assuntos
Antígeno Sialil Lewis X/metabolismo , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Tolerância Central , Citotoxicidade Imunológica , Fator de Transcrição GATA3/metabolismo , Regulação da Expressão Gênica , Humanos , Tolerância Imunológica , Memória Imunológica , Ativação Linfocitária , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Tolerância Periférica , Receptor de Morte Celular Programada 1/metabolismo , Antígeno Sialil Lewis X/genética
16.
J Immunol ; 204(10): 2671-2684, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32238459

RESUMO

TCR signaling is required for homeostasis of naive αß T cells. However, whether such a signal is necessary for γδ T cell homeostasis in the periphery remains unknown. In this study, we present evidence that a portion of Vγ2+ γδ T cells, one of the major γδ T cell subsets in the secondary lymphoid organs, requires TCR signaling for homeostasis. To attenuate γδTCR signals, we generated mice lacking Eγ4 (Eγ4-/-), an enhancer located at the 3'-most end of the TCRγ locus. Overall, we found that in thymus, Eγ4 loss altered V-J rearrangement, chromatin accessibility, and transcription of the TCRγ locus in a distance-dependent manner. Vγ2+ γδ T cells in Eγ4-/- mice developed normally both fetal and adult mouse thymi but were relatively reduced in number in spleen and lymph nodes. Although Vγ2 TCR transcription decreased in all subpopulations of Eγ4-/- mice, the number of Vγ2+ γδ T cells decreased and TCR signaling was attenuated only in the innate-like CD27+CD45RBhigh subpopulation in peripheral lymphoid organs. Consistently, CD27+CD45RBhigh Vγ2+ γδ T cells from Eγ4-/- mice transferred into Rag2-deficient mice were not efficiently recovered, suggesting that continuous TCR signaling is required for their homeostasis. Finally, CD27+CD45RBhigh Vγ2+ γδ T cells from Eγ4-/- mice showed impaired TCR-induced activation and antitumor responses. These results suggest that normal homeostasis of innate-like CD27+CD45RBhigh Vγ2+ γδ T cells in peripheral lymphoid organs requires TCR signaling.


Assuntos
Centro Germinativo/imunologia , Linfonodos/imunologia , Tecido Linfoide/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Subpopulações de Linfócitos T/imunologia , Linfócitos T/imunologia , Animais , Células Cultivadas , Elementos Facilitadores Genéticos/genética , Homeostase , Imunidade Inata , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tolerância Periférica , Receptores de Antígenos de Linfócitos T gama-delta/genética , Transdução de Sinais , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
17.
Immunol Rev ; 285(1): 218-232, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30129206

RESUMO

Generating and maintaining a diverse repertoire of naive T cells is essential for protection against pathogens, and developing a mechanistic and quantitative description of the processes involved lies at the heart of our understanding of vertebrate immunity. Here, we review the biology of naive T cells from birth to maturity and outline how the integration of mathematical models and experiments has helped us to develop a full picture of their life histories.


Assuntos
Circulação Sanguínea/imunologia , Tolerância Periférica , Subpopulações de Linfócitos T/imunologia , Linfócitos T/imunologia , Timo/imunologia , Animais , Diferenciação Celular , Seleção Clonal Mediada por Antígeno , Humanos , Ativação Linfocitária
18.
Semin Cell Dev Biol ; 88: 138-146, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-29355606

RESUMO

The gut is the biggest immune organ in the body that encloses commensal microbiota which aids in food digestion. Paneth cells, positioned at the frontline of host-microbiota interphase, can modulate the composition of microbiota. Paneth cells achieve this via the delivery of microbicidal substances, among which enteric α-defensins play the primary role. If microbiota is dysregulated, it can impact the function of the local mucosal immune system. Importantly, this system is also exposed to an enormous number of antigens which are derived from the gut-resident microbiota and processed food, and may potentially trigger undesirable local inflammatory responses. To understand the intricate regulations and liaisons between Paneth cells, microbiota and the immune system in this intestinal-specific setting, one must consider their mode of interaction in a wider context of regulatory processes which impose immune tolerance not only to self, but also to microbiota and food-derived antigens. These include, but are not limited to, tolerogenic mechanisms of central tolerance in the thymus and peripheral tolerance in the secondary lymphoid organs, and the intestine itself. Defects in these processes can compromise homeostasis in the intestinal mucosal immunity. In this review, which is focused on tolerance to intestinal antigens and its relevance for the pathogenesis of gut immune diseases, we provide an outline of such multilayered immune control mechanisms and highlight functional links that underpin their cooperative nature.


Assuntos
Disbiose/prevenção & controle , Trato Gastrointestinal/imunologia , Celulas de Paneth/imunologia , Tolerância Periférica , alfa-Defensinas/imunologia , Animais , Tolerância Central , Disbiose/imunologia , Disbiose/microbiologia , Microbioma Gastrointestinal/imunologia , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Expressão Gênica/imunologia , Homeostase/imunologia , Humanos , Imunidade nas Mucosas/efeitos dos fármacos , Inflamação , Celulas de Paneth/efeitos dos fármacos , Celulas de Paneth/microbiologia , Simbiose/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/microbiologia , alfa-Defensinas/biossíntese , alfa-Defensinas/farmacologia
19.
Int Immunol ; 32(2): 117-131, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-31586207

RESUMO

Tissue-specific autoimmune diseases are assumed to arise through malfunction of two checkpoints for immune tolerance: defective elimination of autoreactive T cells in the thymus and activation of these T cells by corresponding autoantigens in the periphery. However, evidence for this model and the outcome of such alterations in each or both of the tolerance mechanisms have not been sufficiently investigated. We studied these issues by expressing human AIRE (huAIRE) as a modifier of tolerance function in NOD mice wherein the defects of thymic and peripheral tolerance together cause type I diabetes (T1D). Additive huAIRE expression in the thymic stroma had no major impact on the production of diabetogenic T cells in the thymus. In contrast, huAIRE expression in peripheral antigen-presenting cells (APCs) rendered the mice resistant to T1D, while maintaining other tissue-specific autoimmune responses and antibody production against an exogenous protein antigen, because of the loss of Xcr1+ dendritic cells, an essential component for activating diabetogenic T cells in the periphery. These results contrast with our recent demonstration that huAIRE expression in both the thymic stroma and peripheral APCs resulted in the paradoxical development of muscle-specific autoimmunity. Our results reveal that tissue-specific autoimmunity is differentially controlled by a combination of thymic function and peripheral tolerance, which can be manipulated by expression of huAIRE/Aire in each or both of the tolerance mechanisms.


Assuntos
Autoimunidade/imunologia , Tolerância Periférica/imunologia , Timo/imunologia , Fatores de Transcrição/imunologia , Animais , Autoantígenos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Modelos Animais de Doenças , Humanos , Ilhotas Pancreáticas/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Linfócitos T/imunologia , Fatores de Transcrição/genética , Proteína AIRE
20.
Immunity ; 37(3): 475-86, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22921379

RESUMO

The degree of T cell self-reactivity considered dangerous by the immune system, thereby requiring thymic selection processes to prevent autoimmunity, is unknown. Here, we analyzed a panel of T cell receptors (TCRs) with a broad range of reactivity to ovalbumin (OVA(323-339)) in the rat insulin promoter (RIP)-mOVA self-antigen model for their ability to trigger thymic self-tolerance mechanisms. Thymic regulatory T (Treg) cell generation in vivo was directly correlated with in vitro TCR reactivity to OVA-peptide in a broad ~1,000-fold range. Interestingly, higher TCR affinity was associated with a larger Treg cell developmental "niche" size, even though the amount of antigen should remain constant. The TCR-reactivity threshold to elicit thymic negative selection and peripheral T cell responses was ~100-fold higher than that of Treg cell differentiation. Thus, these data suggest that the broad range of self-reactivity that elicits thymic Treg cell generation is tuned to secure peripheral tolerance to self.


Assuntos
Autoantígenos/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Tolerância a Antígenos Próprios/imunologia , Linfócitos T Reguladores/imunologia , Sequência de Aminoácidos , Animais , Diferenciação Celular/imunologia , Feminino , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/química , Ovalbumina/imunologia , Peptídeos/imunologia , Tolerância Periférica/imunologia , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T Reguladores/metabolismo , Timo/imunologia , Timo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA