Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 134(1)2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-37934601

RESUMO

Although most CD8+ T cells are equipped to kill infected or transformed cells, a subset may regulate immune responses and preserve self-tolerance. Here, we describe a CD8 lineage that is instructed to differentiate into CD8 T regulatory cells (Tregs) by a surprisingly restricted set of T cell receptors (TCRs) that recognize MHC-E (mouse Qa-1) and several dominant self-peptides. Recognition and elimination of pathogenic target cells that express these Qa-1-self-peptide complexes selectively inhibits pathogenic antibody responses without generalized immune suppression. Immunization with synthetic agonist peptides that mobilize CD8 Tregs in vivo efficiently inhibit antigraft antibody responses and markedly prolong heart and kidney organ graft survival. Definition of TCR-dependent differentiation and target recognition by this lineage of CD8 Tregs may open the way to new therapeutic approaches to inhibit pathogenic antibody responses.


Assuntos
Linfócitos T CD8-Positivos , Linfócitos T Reguladores , Camundongos , Animais , Receptores de Antígenos de Linfócitos T , Peptídeos , Tolerância Imunológica , Antígenos de Histocompatibilidade Classe I
2.
Proc Natl Acad Sci U S A ; 119(18): e2200757119, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35482921

RESUMO

Regulatory T cells (Treg) can impede antitumor immunity and currently represent a major obstacle to effective cancer immunotherapy. Targeting tumor-infiltrating regulatory Treg while sparing systemic Treg represents an optimal approach to this problem. Here, we provide evidence that the interleukin 23 receptor (IL23R) expressed by tumor-infiltrating Treg promotes suppressive activity. Disruption of the IL23R results in increased responsiveness of destabilized Treg to the IL12 cytokine, the production of γ-interferon, and the recruitment of CD8 T cells that inhibit tumor growth. Since the Treg destabilization pathway that is initiated by IL23R blockade is distinct and independent from the destabilization pathway coupled to glucocorticoid-induced TNFR-related protein (GITR) activation, we examined the impact of the coordinate induction of the two destabilization pathways on antitumor immune responses. Combined GITR and IL23R antibody treatment of mice inoculated with MC38 tumors resulted in robust and synergistic antitumor responses. These findings indicate that the delineation of independent Treg destabilization pathways may allow improved approaches to the development of combination immunotherapy for cancers.


Assuntos
Neoplasias , Linfócitos T Reguladores , Anticorpos Bloqueadores , Humanos , Fatores Imunológicos/metabolismo , Imunoterapia , Interleucina-23/metabolismo , Neoplasias/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35177477

RESUMO

Expression of Itgax (encoding the CD11c surface protein) and Spp1 (encoding osteopontin; OPN) has been associated with activated microglia that can develop in healthy brains and some neuroinflammatory disorders. However, whether CD11c and OPN expression is a consequence of microglial activation or represents a portion of the genetic program expressed by a stable microglial subset is unknown. Here, we show that OPN production in the brain is confined to a small CD11c+ microglial subset that differentiates from CD11c- precursors in perinatal life after uptake of apoptotic neurons. Our analysis suggests that coexpression of OPN and CD11c marks a microglial subset that is expressed at birth and persists into late adult life, independent of environmental activation stimuli. Analysis of the contribution of OPN to the intrinsic functions of this CD11c+ microglial subset indicates that OPN is required for subset stability and the execution of phagocytic and proinflammatory responses, in part through OPN-dependent engagement of the αVß3-integrin receptor. Definition of OPN-producing CD11c+ microglia as a functional microglial subset provides insight into microglial differentiation in health and disease.


Assuntos
Antígenos CD11/metabolismo , Microglia/fisiologia , Osteopontina/metabolismo , Animais , Encéfalo/metabolismo , Antígenos CD11/genética , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Inflamação/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese , Osteopontina/genética , Fagócitos/metabolismo , Transcriptoma/genética
4.
Nature ; 590(7844): 157-162, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33361812

RESUMO

Tumour-associated antigens (TAAs) comprise a large set of non-mutated cellular antigens recognized by T cells in human and murine cancers. Their potential as targets for immunotherapy has been explored for more than two decades1, yet the origins of TAA-specific T cells remain unclear. While tumour cells may be an important source of TAAs for T cell priming2, several recent studies suggest that infection with some viruses, including Epstein-Barr virus and influenza virus can elicit T cell responses against abnormally expressed cellular antigens that function as TAAs3,4. However, the cellular and molecular basis of such responses remains undefined. Here we show that expression of the Epstein-Barr virus signalling protein LMP1 in B cells provokes T cell responses to multiple TAAs. LMP1 signalling leads to overexpression of many cellular antigens previously shown to be TAAs, their presentation on major histocompatibility complex classes I (MHC-I) and II (MHC-II) (mainly through the endogenous pathway) and the upregulation of costimulatory ligands CD70 and OX40L, thereby inducing potent cytotoxic CD4+ and CD8+ T cell responses. These findings delineate a mechanism of infection-induced anti-tumour immunity. Furthermore, by ectopically expressing LMP1 in tumour B cells from patients with cancer and thereby enabling them to prime T cells, we develop a general approach for rapid production of autologous cytotoxic CD4+ T cells against a wide range of endogenous tumour antigens, such as TAAs and neoantigens, for treating B cell malignancies. This work stresses the need to revisit classical concepts concerning viral and tumour immunity, which will be critical to fully understand the impact of common infections on human health and to improve the rational design of immune approaches to treatment of cancers.


Assuntos
Linfócitos B/imunologia , Linfócitos B/virologia , Linfócitos T CD4-Positivos/imunologia , Herpesvirus Humano 4/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T Citotóxicos/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Antígenos de Neoplasias/imunologia , Ligante CD27/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Ligante OX40/imunologia
5.
Trends Immunol ; 41(10): 948-963, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32976740

RESUMO

Recent progress in cancer immunotherapy highlights the power of the immune system to control tumors, although a small patient subset responds to current immunotherapies. Additional approaches to mobilize antitumor immunity are required to overcome primary and acquired resistance to immunotherapy such as immune checkpoint blockade (ICB). Emerging evidence shows that targeting epigenetic elements that promote tumor progression and inhibit immune cell activity can enhance antitumor immunity by reshaping the tumor microenvironment (TME). Here, we review the pleiotropic functions in tumor and immune cells of enhancer of zeste homolog 2 (EZH2), the catalytic subunit of polycomb repressive complex 2 (PRC2), with a focus on EZH2 inhibition as a potentially promising approach to enhance current immunotherapies and improve patient outcomes for certain cancers.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste , Imunoterapia , Neoplasias , Microambiente Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias/tratamento farmacológico , Microambiente Tumoral/imunologia
6.
Artigo em Inglês | MEDLINE | ID: mdl-32117809

RESUMO

Elucidating key factors that regulate immune-mediated pathology in vivo is critical for developing improved strategies to treat autoimmune disease and cancer. NK cells can exhibit regulatory functions against CD8+ T cells following viral infection. Here we show that while low doses of lymphocytic choriomeningitis virus (LCMV-WE) can readily induce strong CD8+ T cell responses and diabetes in mice expressing the LCMV glycoprotein on ß-islet cells (RIP-GP mice), hyperglycemia does not occur after infection with higher doses of LCMV. High-dose LCMV infection induced an impaired CD8+ T cell response, which coincided with increased NK cell activity during early time points following infection. Notably, we observed increased NKp46 expression on NK cells during infection with higher doses, which resulted in an NK cell dependent suppression of T cells. Accordingly, depletion with antibodies specific for NK1.1 as well as NKp46 deficiency (Ncr1gfp/gfp mice) could restore CD8+ T cell immunity and permitted the induction of diabetes even following infection of RIP-GP mice with high-dose LCMV. Therefore, we identify conditions where innate lymphoid cells can play a regulatory role and interfere with CD8+ T cell mediated tissue specific pathology using an NKp46 dependent mechanism.


Assuntos
Coriomeningite Linfocítica , Animais , Autoimunidade , Linfócitos T CD8-Positivos , Imunidade Inata , Células Matadoras Naturais , Camundongos , Camundongos Endogâmicos C57BL
7.
Cell Rep ; 29(7): 1848-1861.e6, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31722202

RESUMO

Follicular regulatory T (TFR) cells are a specialized suppressive subset that controls the germinal center (GC) response and maintains humoral self-tolerance. The mechanisms that maintain TFR lineage identity and suppressive activity remain largely unknown. Here, we show that expression of Blimp1 by FoxP3+ TFR cells is essential for TFR lineage stability, entry into the GC, and expression of regulatory activity. Deletion of Blimp1 in TFR cells reduced FoxP3 and CTLA-4 expression and increased pro-inflammatory cytokines and spontaneous production of autoantibodies, including elevated IgE. Maintenance of TFR stability reflected Blimp1-dependent repression of the IL-23R-STAT3 axis and activation of the CD25-STAT5 pathway, while silenced IL-23R-STAT3 or increased STAT5 activation rescued the Blimp1-deficient TFR phenotype. Blimp1-dependent control of CXCR5/CCR7 expression also regulated TFR homing into the GC. These findings uncover a Blimp1-dependent TFR checkpoint that enforces suppressive activity and acts as a gatekeeper of GC entry.


Assuntos
Regulação da Expressão Gênica/imunologia , Centro Germinativo/imunologia , Fator 1 de Ligação ao Domínio I Regulador Positivo/imunologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linhagem Celular , Centro Germinativo/citologia , Humanos , Subunidade alfa de Receptor de Interleucina-2/genética , Subunidade alfa de Receptor de Interleucina-2/imunologia , Camundongos , Camundongos Transgênicos , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Receptores CCR7/genética , Receptores CCR7/imunologia , Receptores CXCR5/genética , Receptores CXCR5/imunologia , Receptores de Interleucina/genética , Receptores de Interleucina/imunologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/imunologia , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/imunologia , Transdução de Sinais/genética , Linfócitos T Reguladores/citologia
8.
Adv Immunol ; 140: 1-20, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30366517

RESUMO

Regulatory T cells are central mediators of immune regulation and play an essential role in the maintenance of immune homeostasis in the steady state and under pathophysiological conditions. Disruption of CD8 Treg-dependent recognition of Qa-1-restricted self-antigens can result in dysregulated immune responses, tissue damage, autoimmune disease and cancer. Recent progress in studies on regulatory T cells of the CD8 lineage has provided new biological insight into this specialized regulatory T cell subpopulation. Identification of the Helios transcription factor as an essential control element for the differentiation and function of CD8 regulatory T cells has led to a better understanding of the unique genetic program of these cells. Recent analyses of T-cell receptor usage and antigen recognition by Qa-1-restricted CD8 Treg have provided additional insight into the unusual biological function of this regulatory CD8 lineage. Here we summarize recent advances in our understanding of CD8 regulatory T cells with emphasis on lineage commitment, differentiation and stability.


Assuntos
Doenças Autoimunes/imunologia , Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Animais , Autoantígenos , Antígenos CD8/metabolismo , Diferenciação Celular , Movimento Celular , Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/metabolismo , Homeostase , Humanos , Fator de Transcrição Ikaros/genética , Ativação Linfocitária , Receptores de Antígenos de Linfócitos T/metabolismo
9.
Proc Natl Acad Sci U S A ; 115(26): 6780-6785, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29891681

RESUMO

Lineage commitment and differentiation into CD4+ T cell subsets reflect an interplay between chromatin regulators and transcription factors (TF). Follicular T cell development is regulated by the Bcl6 TF, which helps determine the phenotype and follicular localization of both CD4+ follicular helper T cells (TFH) and follicular regulatory T cells (TFR). Here we show that Bcl6-dependent control of follicular T cells is mediated by a complex formed between Bcl6 and the Mi-2ß-nucleosome-remodeling deacetylase complex (Mi-2ß-NuRD). Formation of this complex reflects the contribution of the intracellular isoform of osteopontin (OPN-i), which acts as a scaffold to stabilize binding between Bcl6 and the NuRD complex that together regulate the genetic program of both TFH and TFR cells. Defective assembly of the Bcl6-NuRD complex distorts follicular T cell differentiation, resulting in impaired TFR development and skewing of the TFH lineage toward a TH1-like program that includes expression of Blimp1, Tbet, granzyme B, and IFNγ. These findings define a core Bcl6-directed transcriptional complex that enables CD4+ follicular T cells to regulate the germinal center response.


Assuntos
Montagem e Desmontagem da Cromatina , Centro Germinativo/citologia , Linfopoese/fisiologia , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/fisiologia , Proteínas Proto-Oncogênicas c-bcl-6/fisiologia , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Reguladores/citologia , Animais , Linhagem da Célula , Regulação da Expressão Gênica/imunologia , Regulação da Expressão Gênica/fisiologia , Técnicas de Introdução de Genes , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/fisiologia , Osteopontina/biossíntese , Osteopontina/genética , Transcrição Gênica
10.
Proc Natl Acad Sci U S A ; 115(9): 2162-2167, 2018 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-29440380

RESUMO

Regulatory T cells (Tregs) are key modulators of immune tolerance, capable of suppressing inflammatory immune responses and promoting nonlymphoid tissue homeostasis. Helios, a transcription factor (TF) that is selectively expressed by Tregs, has been shown to be essential for the maintenance of Treg lineage stability in the face of inflammatory conditions that include autoimmune disease and cancer. Helios-deficient Tregs within tumors acquire effector T cell function and contribute to immune responses against cancer. However, the underlying genetic basis of this Treg reprogramming is not well understood. Here, we report that Helios-deficient Tregs within the chronic inflammatory tumor microenvironment (TME) derepress genetic programs associated with T helper (Th) cell differentiation by up-regulating Th cell-associated TFs and effector cytokines. These genetic changes of Helios-deficient Tregs are most apparent in a Treg subpopulation with high affinity for self-antigens, as detected by both increased GITR/PD-1 expression and increased responsiveness to self-antigens. Their combined effects may promote a phenotype conversion of Tregs into effector T cells within the TME, where TCR engagement and costimulatory receptor expression by Tregs are increased. These data provide a genetic basis for the unstable phenotype of Helios-deficient Tregs within the inflammatory environment of tumors and suggest that immune milieu-dependent alterations in gene expression are a central feature of Treg conversion.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Melanoma/metabolismo , Neoplasias Experimentais/metabolismo , Linfócitos T Reguladores/fisiologia , Fatores de Transcrição/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/genética , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição/genética
11.
Proc Natl Acad Sci U S A ; 115(4): E686-E695, 2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29311309

RESUMO

The B-lymphotropic Epstein-Barr virus (EBV), pandemic in humans, is rapidly controlled on initial infection by T cell surveillance; thereafter, the virus establishes a lifelong latent infection in the host. If surveillance fails, fatal lymphoproliferation and lymphomagenesis ensue. The initial T cell response consists of predominantly CD8+ cytotoxic T cells and a smaller expansion of CD4+ cells. A major approach to treating EBV-associated lymphomas is adoptive transfer of autologous or allogeneic T cells that are stimulated/expanded on EBV-transformed B cells. Strikingly, the clinical response correlates with the frequency of CD4 cells in the infused T cells. Although in vitro studies suggested that EBV-specific CD4 cells develop cytotoxicity, they have not been comprehensively characterized and the molecular mechanism underlying their formation remains unknown. Our recent work, using a transgenic approach in mice, has revealed a central role for the EBV signaling molecule LMP1 in immune surveillance and transformation of EBV-infected B cells. The mouse model offers a unique tool for uncovering basic features of EBV immunity. Here, we show that LMP1 expression in B cells induces potent cytotoxic CD4 and CD8 T cell responses, by enhancing antigen presentation and costimulation by CD70, OX40 ligand, and 4-1BB ligand. Our data further suggest that cytotoxic CD4 cells hold superior therapeutic value for LMP1 (EBV)-driven lymphomas. These findings provide insights into EBV immunity, demonstrating that LMP1 signaling alone is sufficient to induce a prominent cytotoxic CD4 response, and suggest strategies for immunotherapy in EBV-related and other cancers.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/fisiologia , Herpesvirus Humano 4/imunologia , Linfoma/imunologia , Proteínas da Matriz Viral/imunologia , Ligante 4-1BB/metabolismo , Animais , Linfócitos B/metabolismo , Ligante CD27/metabolismo , Linfoma/virologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ligante OX40/metabolismo , Proteínas com Domínio T/metabolismo
12.
Am J Pathol ; 188(3): 757-767, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29248460

RESUMO

Brain injury in premature infants, especially periventricular leukomalacia, is an important cause of neurologic disabilities. Inflammation contributes to perinatal brain injury development, but the essential mediators that lead to early-life brain injury remain largely unknown. Neonates have reduced capacity for mounting conventional αßT-cell responses. However, γδT cells are already functionally competent during early development and are important in early-life immunity. We investigated the potential contribution of γδT cells to preterm brain injury using postmortem brains from human preterm infants with periventricular leukomalacia and two animal models of preterm brain injury-the hypoxic-ischemic mouse model and a fetal sheep asphyxia model. Large numbers of γδT cells were observed in the brains of mice, sheep, and postmortem preterm infants after injury, and depletion of γδT cells provided protection in the mouse model. The common γδT-cell-associated cytokines interferon-γ and IL-17A were not detectable in the brain. Although there were increased mRNA levels of Il17f and Il22 in the mouse brains after injury, neither IL-17F nor IL-22 cytokines contributed to preterm brain injury. These findings highlight unique features of injury in the developing brain, where, unlike injury in the mature brain, γδT cells function as initiators of injury independently of common γδT-cell-associated cytokines. This finding will help to identify therapeutic targets for preventing or treating preterm infants with brain injury.


Assuntos
Encéfalo/patologia , Hipóxia-Isquemia Encefálica/patologia , Linfócitos Intraepiteliais/patologia , Leucomalácia Periventricular/patologia , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Hipóxia-Isquemia Encefálica/metabolismo , Interferon gama/metabolismo , Interleucina-17/metabolismo , Linfócitos Intraepiteliais/metabolismo , Leucomalácia Periventricular/metabolismo , Masculino , Camundongos , Ovinos
13.
PLoS Pathog ; 13(5): e1006384, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28475642

RESUMO

A number of nonclassical MHC Ib molecules recognizing distinct microbial antigens have been implicated in the immune response to Mycobacterium tuberculosis (Mtb). HLA-E has been identified to present numerous Mtb peptides to CD8+ T cells, with multiple HLA-E-restricted cytotoxic T lymphocyte (CTL) and regulatory T cell lines isolated from patients with active and latent tuberculosis (TB). In other disease models, HLA-E and its mouse homolog Qa-1 can act as antigen presenting molecules as well as regulators of the immune response. However, it is unclear what precise role(s) HLA-E/Qa-1 play in the immune response to Mtb. In this study, we found that murine Qa-1 can bind and present Mtb peptide antigens to CD8+ T effector cells during aerosol Mtb infection. Further, mice lacking Qa-1 (Qa-1-/-) were more susceptible to high-dose Mtb infection compared to wild-type controls, with higher bacterial burdens and increased mortality. The increased susceptibility of Qa-1-/- mice was associated with dysregulated T cells that were more activated and produced higher levels of pro-inflammatory cytokines. T cells from Qa-1-/- mice also had increased expression of inhibitory and apoptosis-associated cell surface markers such as CD94/NKG2A, KLRG1, PD-1, Fas-L, and CTLA-4. As such, they were more prone to cell death and had decreased capacity in promoting the killing of Mtb in infected macrophages. Lastly, comparing the immune responses of Qa-1 mutant knock-in mice deficient in either Qa-1-restricted CD8+ Tregs (Qa-1 D227K) or the inhibitory Qa-1-CD94/NKG2A interaction (Qa-1 R72A) with Qa-1-/- and wild-type controls indicated that both of these Qa-1-mediated mechanisms were involved in suppression of the immune response in Mtb infection. Our findings reveal that Qa-1 participates in the immune response to Mtb infection by presenting peptide antigens as well as regulating immune responses, resulting in more effective anti-Mtb immunity.


Assuntos
Antígenos de Bactérias/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/microbiologia , Animais , Apresentação de Antígeno/imunologia , Citocinas/imunologia , Humanos , Macrófagos/imunologia , Camundongos , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Tuberculose/imunologia
14.
J Exp Med ; 214(5): 1453-1469, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28400474

RESUMO

T helper type 17 cells (Th17 cells) are major contributors to many autoimmune diseases. In this study, we demonstrate that the germinal center kinase family member MINK1 (misshapen/NIK-related kinase 1) negatively regulates Th17 cell differentiation. The suppressive effect of MINK1 on induction of Th17 cells is mediated by the inhibition of SMAD2 activation through direct phosphorylation of SMAD2 at the T324 residue. The importance of MINK1 to Th17 cell differentiation was strengthened in the animal model of experimental autoimmune encephalomyelitis (EAE). Moreover, we show that the reactive oxygen species (ROS) scavenger N-acetyl cysteine boosts Th17 cell differentiation in a MINK1-dependent manner and exacerbates the severity of EAE. Thus, we have not only established MINK1 as a critical regulator of Th17 cell differentiation, but also clarified that accumulation of ROS may limit the generation of Th17 cells. The contribution of MINK1 to ROS-regulated Th17 cell differentiation may suggest an important mechanism for the development of autoimmune diseases influenced by antioxidant dietary supplements.


Assuntos
Proteínas Serina-Treonina Quinases/fisiologia , Células Th17/fisiologia , Animais , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo , Proteína Smad2/fisiologia
15.
Proc Natl Acad Sci U S A ; 113(22): 6248-53, 2016 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-27185917

RESUMO

Expression of the transcription factor Helios by Tregs ensures stable expression of a suppressive and anergic phenotype in the face of intense inflammatory responses, whereas Helios-deficient Tregs display diminished lineage stability, reduced FoxP3 expression, and production of proinflammatory cytokines. Here we report that selective Helios deficiency within CD4 Tregs leads to enhanced antitumor immunity through induction of an unstable phenotype and conversion of intratumoral Tregs into T effector cells within the tumor microenvironment. Induction of an unstable Treg phenotype is associated with enhanced production of proinflammatory cytokines by tumor-infiltrating but not systemic Tregs and significantly delayed tumor growth. Ab-dependent engagement of Treg surface receptors that result in Helios down-regulation also promotes conversion of intratumoral but not systemic Tregs into T effector cells and leads to enhanced antitumor immunity. These findings suggest that selective instability and conversion of intratumoral CD4 Tregs through genetic or Ab-based targeting of Helios may represent an effective approach to immunotherapy.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/prevenção & controle , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Fatores de Transcrição/fisiologia , Animais , Linfócitos do Interstício Tumoral/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Knockout , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Reguladores/metabolismo
16.
Proc Natl Acad Sci U S A ; 112(52): 15988-93, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26668377

RESUMO

Changes of histone modification status at critical lineage-specifying gene loci in multipotent precursors can influence cell fate commitment. The contribution of these epigenetic mechanisms to natural killer (NK) cell lineage determination from common lymphoid precursors is not understood. Here we investigate the impact of histone methylation repressive marks (H3 Lys27 trimethylation; H3K27(me3)) on early NK cell differentiation. We demonstrate that selective loss of the histone-lysine N-methyltransferase Ezh2 (enhancer of zeste homolog 2) or inhibition of its enzymatic activity with small molecules unexpectedly increased generation of the IL-15 receptor (IL-15R) CD122(+) NK precursors and mature NK progeny from both mouse and human hematopoietic stem and progenitor cells. Mechanistic studies revealed that enhanced NK cell expansion and cytotoxicity against tumor cells were associated with up-regulation of CD122 and the C-type lectin receptor NKG2D. Moreover, NKG2D deficiency diminished the positive effects of Ezh2 inhibitors on NK cell commitment. Identification of the contribution of Ezh2 to NK lineage specification and function reveals an epigenetic-based mechanism that regulates NK cell development and provides insight into the clinical application of Ezh2 inhibitors in NK-based cancer immunotherapies.


Assuntos
Diferenciação Celular/genética , Histona-Lisina N-Metiltransferase/genética , Células Matadoras Naturais/metabolismo , Complexo Repressor Polycomb 2/genética , Animais , Linhagem Celular Tumoral , Linhagem da Célula/genética , Proliferação de Células/genética , Sobrevivência Celular/genética , Células Cultivadas , Proteína Potenciadora do Homólogo 2 de Zeste , Citometria de Fluxo , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Immunoblotting , Subunidade beta de Receptor de Interleucina-2/genética , Subunidade beta de Receptor de Interleucina-2/metabolismo , Células K562 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Proc Natl Acad Sci U S A ; 112(2): 494-9, 2015 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-25550515

RESUMO

Natural killer (NK) cells play an essential role in the immune response to infection and cancer. After infection or during homeostatic expansion NK cells express a developmental program that includes a contraction phase followed by the formation of long-lived mature memory-like cells. Although this NK cell response pattern is well established, the underlying mechanisms that ensure efficient transition to long-lived NK cells remain largely undefined. Here we report that deficient expression of intracellular osteopontin (OPN-i) by NK cells results in defective responses to IL-15 associated with a substantial increase in the NK cell contraction phase of homeostatic expansion, defective expression of the Eomes transcription factor, and diminished responses to metastatic tumors. The OPN-i-deficient phenotype is accompanied by increased NK cell apoptosis, impaired transition from immature to mature NK cells, and diminished ability to develop memory-like NK cells that respond to mouse cytomegalovirus. Gene pathway analysis of OPN-i-deficient NK cells suggests that the mechanistic target of rapamycin pathway may connect OPN-i to Eomes and T-bet expression by mature NK cells following up-regulation of OPN-i after IL-15 stimulation. Identification of OPN-i as an essential molecular component for maintenance of functional NK cell expansion provides insight into the NK cell response and may provide the basis for improved approaches to immunotherapy for infectious disease and cancer.


Assuntos
Células Matadoras Naturais/imunologia , Osteopontina/imunologia , Animais , Diferenciação Celular/imunologia , Sobrevivência Celular/imunologia , Homeostase/imunologia , Interleucina-15/imunologia , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Melanoma Experimental/imunologia , Melanoma Experimental/secundário , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteopontina/deficiência , Osteopontina/genética , Transdução de Sinais/imunologia , Proteínas com Domínio T/metabolismo , Serina-Treonina Quinases TOR/imunologia , Serina-Treonina Quinases TOR/metabolismo
18.
Nat Immunol ; 16(1): 96-106, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25436971

RESUMO

Follicular helper T cells (TFH cells) and follicular regulatory T cells (TFR cells) regulate the quantity and quality of humoral immunity. Although both cell types express the costimulatory receptor ICOS and require the transcription factor Bcl-6 for their differentiation, the ICOS-dependent pathways that coordinate their responses are not well understood. Here we report that activation of ICOS in CD4(+) T cells promoted interaction of the p85α regulatory subunit of the signaling kinase PI(3)K and intracellular osteopontin (OPN-i), followed by translocation of OPN-i to the nucleus, its interaction with Bcl-6 and protection of Bcl-6 from ubiquitin-dependent proteasome degradation. Post-translational protection of Bcl-6 by OPN-i was essential for sustained responses of TFH cells and TFR cells and regulation of the germinal center B cell response to antigen. Thus, the p85α-OPN-i axis represents a molecular bridge that couples activation of ICOS to Bcl-6-dependent functional differentiation of TFH cells and TFR cells; this suggests new therapeutic avenues to manipulate the responses of these cells.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/imunologia , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Osteopontina/imunologia , Proteínas Proto-Oncogênicas c-bcl-6/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular/imunologia , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Feminino , Citometria de Fluxo , Centro Germinativo/imunologia , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteopontina/genética , Isoformas de Proteínas , Proteínas Proto-Oncogênicas c-bcl-6/genética , RNA Mensageiro/química , RNA Mensageiro/genética , Distribuição Aleatória , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Organismos Livres de Patógenos Específicos
19.
Cancer Immunol Res ; 2(10): 926-36, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25281320

RESUMO

Cancer immunology has recently made major therapeutic inroads that represent clinical application of basic insights into mechanisms that govern immunity against tumors. Research into fundamental elements of T-cell and natural killer-cell biology, including the basis of antigen recognition, activation, proliferation, and survival, has informed the design of new therapeutic approaches to augment the body's natural anticancer immune response. Here, we describe some of the key steps that have provided the foundation for current strategies of immunotherapy.


Assuntos
Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Humanos , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Subpopulações de Linfócitos T/imunologia , Evasão Tumoral/imunologia
20.
Cancer Immunol Res ; 2(2): 91-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24778273

RESUMO

Research over the past decade has revealed the increasingly complex biologic features of the CD4(+) T-cell lineage. This T-cell subset, which was originally defined on the basis of helper activity in antibody responses, expresses receptors that recognize peptides that have been processed and presented by specialized antigen-presenting cells. At the core of the adaptive immune response, CD4 T cells display a large degree of plasticity and the ability to differentiate into multiple sublineages in response to developmental and environmental cues. These differentiated sublineages can orchestrate a broad range of effector activities during the initiation, expansion, and memory phase of an immune response. The contribution of CD4 cells to host defense against pathogenic invasion and regulation of autoimmunity is now well established. Emerging evidence suggests that CD4 cells also actively participate in shaping antitumor immunity. Here, we outline the biologic properties of CD4 T-cell subsets with an emphasis on their contribution to the antitumor response.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Neoplasias/imunologia , Subpopulações de Linfócitos T/imunologia , Imunidade Adaptativa/imunologia , Diferenciação Celular/imunologia , Humanos , Imunoterapia/métodos , Neoplasias/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA