Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 162.687
Filtrar
Mais filtros

Coleção SES
Intervalo de ano de publicação
1.
Annu Rev Immunol ; 39: 719-757, 2021 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-33646859

RESUMO

The enigmatic eosinophil has emerged as an exciting component of the immune system, involved in a plethora of homeostatic and inflammatory responses. Substantial progress has been achieved through experimental systems manipulating eosinophils in vivo, initially in mice and more recently in humans. Researchers using eosinophil knockout mice have identified a contributory role for eosinophils in basal and inflammatory processes and protective immunity. Primarily fueled by the purported proinflammatory role of eosinophils in eosinophil-associated diseases, a series of anti-eosinophil therapeutics have emerged as a new class of drugs. These agents, which dramatically deplete eosinophils, provide a valuable opportunity to characterize the consequences of eosinophil knockout humans. Herein, we comparatively describe mouse and human eosinophil knockouts. We put forth the view that human eosinophils negatively contribute to a variety of diseases and, unlike mouse eosinophils, do not yet have an identified role in physiological health; thus, clarifying all roles of eosinophils remains an ongoing pursuit.


Assuntos
Eosinófilos , Preparações Farmacêuticas , Animais , Terapia Biológica , Humanos , Camundongos , Camundongos Knockout
2.
Cell ; 185(3): 547-562.e22, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35051369

RESUMO

Hundreds of microbiota genes are associated with host biology/disease. Unraveling the causal contribution of a microbiota gene to host biology remains difficult because many are encoded by nonmodel gut commensals and not genetically targetable. A general approach to identify their gene transfer methodology and build their gene manipulation tools would enable mechanistic dissections of their impact on host physiology. We developed a pipeline that identifies the gene transfer methods for multiple nonmodel microbes spanning five phyla, and we demonstrated the utility of their genetic tools by modulating microbiome-derived short-chain fatty acids and bile acids in vitro and in the host. In a proof-of-principle study, by deleting a commensal gene for bile acid synthesis in a complex microbiome, we discovered an intriguing role of this gene in regulating colon inflammation. This technology will enable genetically engineering the nonmodel gut microbiome and facilitate mechanistic dissection of microbiota-host interactions.


Assuntos
Microbioma Gastrointestinal/genética , Genes Bacterianos , Animais , Ácidos e Sais Biliares/metabolismo , Sistemas CRISPR-Cas/genética , Clostridium/genética , Colite/induzido quimicamente , Colite/microbiologia , Colite/patologia , Sulfato de Dextrana , Resistência Microbiana a Medicamentos/genética , Feminino , Regulação Bacteriana da Expressão Gênica , Técnicas de Transferência de Genes , Vida Livre de Germes , Inflamação/patologia , Intestinos/patologia , Masculino , Metaboloma/genética , Metagenômica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese Insercional/genética , Mutação/genética , RNA Ribossômico 16S/genética , Transcrição Gênica
3.
Nat Immunol ; 25(1): 178-188, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38012416

RESUMO

Annotation of immunologic gene function in vivo typically requires the generation of knockout mice, which is time consuming and low throughput. We previously developed CHimeric IMmune Editing (CHIME), a CRISPR-Cas9 bone marrow delivery system for constitutive, ubiquitous deletion of single genes. Here we describe X-CHIME, four new CHIME-based systems for modular and rapid interrogation of gene function combinatorially (C-CHIME), inducibly (I-CHIME), lineage-specifically (L-CHIME) or sequentially (S-CHIME). We use C-CHIME and S-CHIME to assess the consequences of combined deletion of Ptpn1 and Ptpn2, an embryonic lethal gene pair, in adult mice. We find that constitutive deletion of both PTPN1 and PTPN2 leads to bone marrow hypoplasia and lethality, while inducible deletion after immune development leads to enteritis and lethality. These findings demonstrate that X-CHIME can be used for rapid mechanistic evaluation of genes in distinct in vivo contexts and that PTPN1 and PTPN2 have some functional redundancy important for viability in adult mice.


Assuntos
Sistemas CRISPR-Cas , Proteína Tirosina Fosfatase não Receptora Tipo 2 , Camundongos , Animais , Sistemas CRISPR-Cas/genética , Proteína Tirosina Fosfatase não Receptora Tipo 2/genética , Camundongos Knockout , Sistema Imunitário , Edição de Genes
4.
Nat Immunol ; 25(1): 77-87, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38049581

RESUMO

Intestinal intraepithelial lymphocytes (IELs) exhibit prompt innate-like responses to microenvironmental cues and require strict control of effector functions. Here we showed that Aiolos, an Ikaros zinc-finger family member encoded by Ikzf3, acted as a regulator of IEL activation. Ikzf3-/- CD8αα+ IELs had elevated expression of NK receptors, cytotoxic enzymes, cytokines and chemokines. Single-cell RNA sequencing of Ikzf3-/- and Ikzf3+/+ IELs showed an amplified effector machinery in Ikzf3-/- CD8αα+ IELs compared to Ikzf3+/+ counterparts. Ikzf3-/- CD8αα+ IELs had increased responsiveness to interleukin-15, which explained a substantial part, but not all, of the observed phenotypes. Aiolos binding sites were close to those for the transcription factors STAT5 and RUNX, which promote interleukin-15 signaling and cytolytic programs, and Ikzf3 deficiency partially increased chromatin accessibility and histone acetylation in these regions. Ikzf3 deficiency in mice enhanced susceptibility to colitis, underscoring the relevance of Aiolos in regulating the effector function in IELs.


Assuntos
Linfócitos Intraepiteliais , Fatores de Transcrição , Animais , Camundongos , Antígenos CD8/metabolismo , Interleucina-15/metabolismo , Mucosa Intestinal/metabolismo , Linfócitos Intraepiteliais/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Nat Immunol ; 25(7): 1172-1182, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38871999

RESUMO

Natural killer (NK) cells traffic through the blood and mount cytolytic and interferon-γ (IFNγ)-focused responses to intracellular pathogens and tumors. Type 1 innate lymphoid cells (ILC1s) also produce type 1 cytokines but reside in tissues and are not cytotoxic. Whether these differences reflect discrete lineages or distinct states of a common cell type is not understood. Using single-cell RNA sequencing and flow cytometry, we focused on populations of TCF7+ cells that contained precursors for NK cells and ILC1s and identified a subset of bone marrow lineage-negative NK receptor-negative cells that expressed the transcription factor Eomes, termed EomeshiNKneg cells. Transfer of EomeshiNKneg cells into Rag2-/-Il2rg-/- recipients generated functional NK cells capable of preventing metastatic disease. By contrast, transfer of PLZF+ ILC precursors generated a mixture of ILC1s, ILC2s and ILC3s that lacked cytotoxic potential. These findings identified EomeshiNKneg cells as the bone marrow precursor to classical NK cells and demonstrated that the NK and ILC1 lineages diverged early during development.


Assuntos
Células Matadoras Naturais , Proteínas com Domínio T , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Animais , Proteínas com Domínio T/metabolismo , Proteínas com Domínio T/genética , Camundongos , Camundongos Knockout , Linhagem da Célula/imunologia , Camundongos Endogâmicos C57BL , Imunidade Inata , Diferenciação Celular/imunologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Análise de Célula Única
6.
Nat Immunol ; 25(5): 778-789, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38589619

RESUMO

Natural killer (NK) cells are a critical first line of defense against viral infection. Rare mutations in a small subset of transcription factors can result in decreased NK cell numbers and function in humans, with an associated increased susceptibility to viral infection. However, our understanding of the specific transcription factors governing mature human NK cell function is limited. Here we use a non-viral CRISPR-Cas9 knockout screen targeting genes encoding 31 transcription factors differentially expressed during human NK cell development. We identify myocyte enhancer factor 2C (MEF2C) as a master regulator of human NK cell functionality ex vivo. MEF2C-haploinsufficient patients and mice displayed defects in NK cell development and effector function, with an increased susceptibility to viral infection. Mechanistically, MEF2C was required for an interleukin (IL)-2- and IL-15-mediated increase in lipid content through regulation of sterol regulatory element-binding protein (SREBP) pathways. Supplementation with oleic acid restored MEF2C-deficient and MEF2C-haploinsufficient patient NK cell cytotoxic function. Therefore, MEF2C is a critical orchestrator of NK cell antiviral immunity by regulating SREBP-mediated lipid metabolism.


Assuntos
Células Matadoras Naturais , Metabolismo dos Lipídeos , Fatores de Transcrição MEF2 , Fatores de Transcrição MEF2/metabolismo , Fatores de Transcrição MEF2/genética , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Animais , Humanos , Camundongos , Sistemas CRISPR-Cas , Camundongos Knockout , Interleucina-15/metabolismo , Camundongos Endogâmicos C57BL
7.
Nat Immunol ; 25(5): 886-901, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38609547

RESUMO

Intestinal immune responses to microbes are controlled by the cytokine IL-10 to avoid immune pathology. Here, we use single-cell RNA sequencing of colon lamina propria leukocytes (LPLs) along with RNA-seq and ATAC-seq of purified CD4+ T cells to show that the transcription factors Blimp-1 (encoded by Prdm1) and c-Maf co-dominantly regulate Il10 while negatively regulating proinflammatory cytokines in effector T cells. Double-deficient Prdm1fl/flMaffl/flCd4Cre mice infected with Helicobacter hepaticus developed severe colitis with an increase in TH1/NK/ILC1 effector genes in LPLs, while Prdm1fl/flCd4Cre and Maffl/flCd4Cre mice exhibited moderate pathology and a less-marked type 1 effector response. LPLs from infected Maffl/flCd4Cre mice had increased type 17 responses with increased Il17a and Il22 expression and an increase in granulocytes and myeloid cell numbers, resulting in increased T cell-myeloid-neutrophil interactions. Genes over-expressed in human inflammatory bowel disease showed differential expression in LPLs from infected mice in the absence of Prdm1 or Maf, revealing potential mechanisms of human disease.


Assuntos
Colite , Helicobacter hepaticus , Camundongos Knockout , Fator 1 de Ligação ao Domínio I Regulador Positivo , Proteínas Proto-Oncogênicas c-maf , Animais , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-maf/genética , Colite/imunologia , Colite/genética , Humanos , Helicobacter hepaticus/imunologia , Infecções por Helicobacter/imunologia , Camundongos Endogâmicos C57BL , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Mucosa Intestinal/microbiologia , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/genética , Regulação da Expressão Gênica , Modelos Animais de Doenças
8.
Nat Immunol ; 25(5): 790-801, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38664585

RESUMO

Innate immune cells generate a multifaceted antitumor immune response, including the conservation of essential nutrients such as iron. These cells can be modulated by commensal bacteria; however, identifying and understanding how this occurs is a challenge. Here we show that the food commensal Lactiplantibacillus plantarum IMB19 augments antitumor immunity in syngeneic and xenograft mouse tumor models. Its capsular heteropolysaccharide is the major effector molecule, functioning as a ligand for TLR2. In a two-pronged manner, it skews tumor-associated macrophages to a classically active phenotype, leading to generation of a sustained CD8+ T cell response, and triggers macrophage 'nutritional immunity' to deploy the high-affinity iron transporter lipocalin-2 for capturing and sequestering iron in the tumor microenvironment. This process induces a cycle of tumor cell death, epitope expansion and subsequent tumor clearance. Together these data indicate that food commensals might be identified and developed into 'oncobiotics' for a multi-layered approach to cancer therapy.


Assuntos
Ferro , Microambiente Tumoral , Animais , Ferro/metabolismo , Camundongos , Microambiente Tumoral/imunologia , Humanos , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Receptor 2 Toll-Like/metabolismo , Receptor 2 Toll-Like/imunologia , Camundongos Endogâmicos C57BL , Lipocalina-2/metabolismo , Lipocalina-2/imunologia , Feminino , Simbiose/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Ativação de Macrófagos/imunologia , Camundongos Knockout
9.
Nat Immunol ; 25(6): 1046-1058, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38816618

RESUMO

The durability of an antitumor immune response is mediated in part by the persistence of progenitor exhausted CD8+ T cells (Tpex). Tpex serve as a resource for replenishing effector T cells and preserve their quantity through self-renewal. However, it is unknown how T cell receptor (TCR) engagement affects the self-renewal capacity of Tpex in settings of continued antigen exposure. Here we use a Lewis lung carcinoma model that elicits either optimal or attenuated TCR signaling in CD8+ T cells to show that formation of Tpex in tumor-draining lymph nodes and their intratumoral persistence is dependent on optimal TCR engagement. Notably, attenuated TCR stimulation accelerates the terminal differentiation of optimally primed Tpex. This TCR-reinforced Tpex development and self-renewal is coupled to proximal positioning to dendritic cells and epigenetic imprinting involving increased chromatin accessibility at Egr2 and Tcf1 target loci. Collectively, this study highlights the critical function of TCR engagement in sustaining Tpex during tumor progression.


Assuntos
Linfócitos T CD8-Positivos , Carcinoma Pulmonar de Lewis , Fator 1-alfa Nuclear de Hepatócito , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T , Animais , Linfócitos T CD8-Positivos/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Camundongos , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Lewis/metabolismo , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Transdução de Sinais/imunologia , Camundongos Knockout , Ativação Linfocitária/imunologia , Autorrenovação Celular , Camundongos Transgênicos , Proteína 2 de Resposta de Crescimento Precoce
10.
Nat Immunol ; 25(6): 994-1006, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38671323

RESUMO

The lung is constantly exposed to the outside world and optimal adaptation of immune responses is crucial for efficient pathogen clearance. However, mechanisms that lead to lung-associated macrophages' functional and developmental adaptation remain elusive. To reveal such mechanisms, we developed a reductionist model of environmental intranasal ß-glucan exposure, allowing for the detailed interrogation of molecular mechanisms of pulmonary macrophage adaptation. Employing single-cell transcriptomics, high-dimensional imaging and flow cytometric characterization paired with in vivo and ex vivo challenge models, we reveal that pulmonary low-grade inflammation results in the development of apolipoprotein E (ApoE)-dependent monocyte-derived alveolar macrophages (ApoE+CD11b+ AMs). ApoE+CD11b+ AMs expressed high levels of CD11b, ApoE, Gpnmb and Ccl6, were glycolytic, highly phagocytic and produced large amounts of interleukin-6 upon restimulation. Functional differences were cell intrinsic, and myeloid cell-specific ApoE ablation inhibited Ly6c+ monocyte to ApoE+CD11b+ AM differentiation dependent on macrophage colony-stimulating factor secretion, promoting ApoE+CD11b+ AM cell death and thus impeding ApoE+CD11b+ AM maintenance. In vivo, ß-glucan-elicited ApoE+CD11b+ AMs limited the bacterial burden of Legionella pneumophilia after infection and improved the disease outcome in vivo and ex vivo in a murine lung fibrosis model. Collectively these data identify ApoE+CD11b+ AMs generated upon environmental cues, under the control of ApoE signaling, as an essential determinant for lung adaptation enhancing tissue resilience.


Assuntos
Apolipoproteínas E , Lectinas Tipo C , Macrófagos Alveolares , Camundongos Endogâmicos C57BL , beta-Glucanas , Animais , Camundongos , Adaptação Fisiológica/imunologia , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Antígeno CD11b/metabolismo , Diferenciação Celular , Lectinas Tipo C/metabolismo , Pulmão/imunologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo
11.
Nat Immunol ; 25(6): 1007-1019, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38816617

RESUMO

Rare multipotent stem cells replenish millions of blood cells per second through a time-consuming process, passing through multiple stages of increasingly lineage-restricted progenitors. Although insults to the blood-forming system highlight the need for more rapid blood replenishment from stem cells, established models of hematopoiesis implicate only one mandatory differentiation pathway for each blood cell lineage. Here, we establish a nonhierarchical relationship between distinct stem cells that replenish all blood cell lineages and stem cells that replenish almost exclusively platelets, a lineage essential for hemostasis and with important roles in both the innate and adaptive immune systems. These distinct stem cells use cellularly, molecularly and functionally separate pathways for the replenishment of molecularly distinct megakaryocyte-restricted progenitors: a slower steady-state multipotent pathway and a fast-track emergency-activated platelet-restricted pathway. These findings provide a framework for enhancing platelet replenishment in settings in which slow recovery of platelets remains a major clinical challenge.


Assuntos
Plaquetas , Diferenciação Celular , Células-Tronco Hematopoéticas , Megacariócitos , Plaquetas/imunologia , Plaquetas/metabolismo , Animais , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Diferenciação Celular/imunologia , Megacariócitos/citologia , Linhagem da Célula , Camundongos Endogâmicos C57BL , Hematopoese , Trombopoese , Camundongos Knockout , Humanos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Células-Tronco Multipotentes/imunologia
12.
Nat Immunol ; 25(7): 1283-1295, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38862796

RESUMO

While some infections elicit germinal centers, others produce only extrafollicular responses. The mechanisms controlling these dichotomous fates are poorly understood. We identify IL-12 as a cytokine switch, acting directly on B cells to promote extrafollicular and suppress germinal center responses. IL-12 initiates a B cell-intrinsic feed-forward loop between IL-12 and IFNγ, amplifying IFNγ production, which promotes proliferation and plasmablast differentiation from mouse and human B cells, in synergy with IL-12. IL-12 sustains the expression of a portion of IFNγ-inducible genes. Together, they also induce unique gene changes, reflecting both IFNγ amplification and cooperative effects between both cytokines. In vivo, cells lacking both IL-12 and IFNγ receptors are more impaired in plasmablast production than those lacking either receptor alone. Further, B cell-derived IL-12 enhances both plasmablast responses and T helper 1 cell commitment. Thus, B cell-derived IL-12, acting on T and B cells, determines the immune response mode, with implications for vaccines, pathogen protection and autoimmunity.


Assuntos
Linfócitos B , Diferenciação Celular , Centro Germinativo , Interferon gama , Interleucina-12 , Animais , Interleucina-12/imunologia , Interleucina-12/metabolismo , Camundongos , Interferon gama/metabolismo , Interferon gama/imunologia , Centro Germinativo/imunologia , Humanos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Diferenciação Celular/imunologia , Camundongos Knockout , Camundongos Endogâmicos C57BL , Plasmócitos/imunologia , Plasmócitos/metabolismo , Ativação Linfocitária/imunologia , Receptores de Interferon/metabolismo , Receptores de Interferon/genética , Células Cultivadas , Proliferação de Células
13.
Nat Immunol ; 25(7): 1193-1206, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38834865

RESUMO

Immune cells experience large cell shape changes during environmental patrolling because of the physical constraints that they encounter while migrating through tissues. These cells can adapt to such deformation events using dedicated shape-sensing pathways. However, how shape sensing affects immune cell function is mostly unknown. Here, we identify a shape-sensing mechanism that increases the expression of the chemokine receptor CCR7 and guides dendritic cell migration from peripheral tissues to lymph nodes at steady state. This mechanism relies on the lipid metabolism enzyme cPLA2, requires nuclear envelope tensioning and is finely tuned by the ARP2/3 actin nucleation complex. We also show that this shape-sensing axis reprograms dendritic cell transcription by activating an IKKß-NF-κB-dependent pathway known to control their tolerogenic potential. These results indicate that cell shape changes experienced by immune cells can define their migratory behavior and immunoregulatory properties and reveal a contribution of the physical properties of tissues to adaptive immunity.


Assuntos
Movimento Celular , Células Dendríticas , Homeostase , Linfonodos , Camundongos Endogâmicos C57BL , Receptores CCR7 , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Linfonodos/imunologia , Linfonodos/citologia , Receptores CCR7/metabolismo , Camundongos , Movimento Celular/imunologia , Forma Celular , NF-kappa B/metabolismo , Camundongos Knockout , Transdução de Sinais/imunologia , Quinase I-kappa B/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo
14.
Nat Immunol ; 25(7): 1183-1192, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38872000

RESUMO

Natural killer (NK) cells function by eliminating virus-infected or tumor cells. Here we identified an NK-lineage-biased progenitor population, referred to as early NK progenitors (ENKPs), which developed into NK cells independently of common precursors for innate lymphoid cells (ILCPs). ENKP-derived NK cells (ENKP_NK cells) and ILCP-derived NK cells (ILCP_NK cells) were transcriptionally different. We devised combinations of surface markers that identified highly enriched ENKP_NK and ILCP_NK cell populations in wild-type mice. Furthermore, Ly49H+ NK cells that responded to mouse cytomegalovirus infection primarily developed from ENKPs, whereas ILCP_NK cells were better IFNγ producers after infection with Salmonella and herpes simplex virus. Human CD56dim and CD56bright NK cells were transcriptionally similar to ENKP_NK cells and ILCP_NK cells, respectively. Our findings establish the existence of two pathways of NK cell development that generate functionally distinct NK cell subsets in mice and further suggest these pathways may be conserved in humans.


Assuntos
Diferenciação Celular , Células Matadoras Naturais , Células Matadoras Naturais/imunologia , Animais , Camundongos , Humanos , Diferenciação Celular/imunologia , Camundongos Endogâmicos C57BL , Imunidade Inata , Antígeno CD56/metabolismo , Muromegalovirus/imunologia , Linhagem da Célula/imunologia , Interferon gama/metabolismo , Interferon gama/imunologia , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Linfoides/citologia , Células Progenitoras Linfoides/imunologia , Camundongos Knockout , Células Cultivadas
15.
Nat Immunol ; 25(5): 860-872, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38632339

RESUMO

Adaptive immunity relies on specialized effector functions elicited by lymphocytes, yet how antigen recognition activates appropriate effector responses through nonspecific signaling intermediates is unclear. Here we examined the role of chromatin priming in specifying the functional outputs of effector T cells and found that most of the cis-regulatory landscape active in effector T cells was poised early in development before the expression of the T cell antigen receptor. We identified two principal mechanisms underpinning this poised landscape: the recruitment of the nucleosome remodeler mammalian SWItch/Sucrose Non-Fermentable (mSWI/SNF) by the transcription factors RUNX1 and PU.1 to establish chromatin accessibility at T effector loci; and a 'relay' whereby the transcription factor BCL11B succeeded PU.1 to maintain occupancy of the chromatin remodeling complex mSWI/SNF together with RUNX1, after PU.1 silencing during lineage commitment. These mechanisms define modes by which T cells acquire the potential to elicit specialized effector functions early in their ontogeny and underscore the importance of integrating extrinsic cues to the developmentally specified intrinsic program.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core , Proteínas Proto-Oncogênicas , Proteínas Repressoras , Transativadores , Fatores de Transcrição , Proteínas Supressoras de Tumor , Proteínas Proto-Oncogênicas/metabolismo , Animais , Transativadores/metabolismo , Transativadores/genética , Camundongos , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Proteínas Repressoras/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Camundongos Endogâmicos C57BL , Proteínas Cromossômicas não Histona/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Camundongos Knockout , Montagem e Desmontagem da Cromatina , Diferenciação Celular/imunologia
16.
Nat Immunol ; 25(6): 981-993, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38811816

RESUMO

Viral infection makes us feel sick as the immune system alters systemic metabolism to better fight the pathogen. The extent of these changes is relative to the severity of disease. Whether blood glucose is subject to infection-induced modulation is mostly unknown. Here we show that strong, nonlethal infection restricts systemic glucose availability, which promotes the antiviral type I interferon (IFN-I) response. Following viral infection, we find that IFNγ produced by γδ T cells stimulates pancreatic ß cells to increase glucose-induced insulin release. Subsequently, hyperinsulinemia lessens hepatic glucose output. Glucose restriction enhances IFN-I production by curtailing lactate-mediated inhibition of IRF3 and NF-κB signaling. Induced hyperglycemia constrained IFN-I production and increased mortality upon infection. Our findings identify glucose restriction as a physiological mechanism to bring the body into a heightened state of responsiveness to viral pathogens. This immune-endocrine circuit is disrupted in hyperglycemia, possibly explaining why patients with diabetes are more susceptible to viral infection.


Assuntos
Glicemia , Imunidade Inata , Interferon gama , Animais , Interferon gama/metabolismo , Interferon gama/imunologia , Camundongos , Glicemia/metabolismo , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia , Insulina/metabolismo , Insulina/imunologia , Camundongos Knockout , Hiperglicemia/imunologia , Fator Regulador 3 de Interferon/metabolismo , NF-kappa B/metabolismo , Humanos , Fígado/imunologia , Fígado/virologia , Fígado/metabolismo , Masculino
17.
Nat Immunol ; 25(6): 1020-1032, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38831106

RESUMO

The efficacy of T cell-based immunotherapies is limited by immunosuppressive pressures in the tumor microenvironment. Here we show a predominant role for the interaction between BTLA on effector T cells and HVEM (TNFRSF14) on immunosuppressive tumor microenvironment cells, namely regulatory T cells. High BTLA expression in chimeric antigen receptor (CAR) T cells correlated with poor clinical response to treatment. Therefore, we deleted BTLA in CAR T cells and show improved tumor control and persistence in models of lymphoma and solid malignancies. Mechanistically, BTLA inhibits CAR T cells via recruitment of tyrosine phosphatases SHP-1 and SHP-2, upon trans engagement with HVEM. BTLA knockout thus promotes CAR signaling and subsequently enhances effector function. Overall, these data indicate that the BTLA-HVEM axis is a crucial immune checkpoint in CAR T cell immunotherapy and warrants the use of strategies to overcome this barrier.


Assuntos
Imunoterapia Adotiva , Receptores de Antígenos Quiméricos , Receptores Imunológicos , Membro 14 de Receptores do Fator de Necrose Tumoral , Microambiente Tumoral , Animais , Humanos , Imunoterapia Adotiva/métodos , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/imunologia , Membro 14 de Receptores do Fator de Necrose Tumoral/genética , Camundongos , Microambiente Tumoral/imunologia , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Receptores de Antígenos Quiméricos/genética , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Linfócitos T Reguladores/imunologia , Transdução de Sinais , Linhagem Celular Tumoral , Neoplasias/imunologia , Neoplasias/terapia , Camundongos Knockout
18.
Nat Immunol ; 25(5): 847-859, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38658806

RESUMO

Immune cells need to sustain a state of constant alertness over a lifetime. Yet, little is known about the regulatory processes that control the fluent and fragile balance that is called homeostasis. Here we demonstrate that JAK-STAT signaling, beyond its role in immune responses, is a major regulator of immune cell homeostasis. We investigated JAK-STAT-mediated transcription and chromatin accessibility across 12 mouse models, including knockouts of all STAT transcription factors and of the TYK2 kinase. Baseline JAK-STAT signaling was detected in CD8+ T cells and macrophages of unperturbed mice-but abrogated in the knockouts and in unstimulated immune cells deprived of their normal tissue context. We observed diverse gene-regulatory programs, including effects of STAT2 and IRF9 that were independent of STAT1. In summary, our large-scale dataset and integrative analysis of JAK-STAT mutant and wild-type mice uncovered a crucial role of JAK-STAT signaling in unstimulated immune cells, where it contributes to a poised epigenetic and transcriptional state and helps prepare these cells for rapid response to immune stimuli.


Assuntos
Homeostase , Janus Quinases , Macrófagos , Camundongos Knockout , Fatores de Transcrição STAT , Transdução de Sinais , Animais , Camundongos , Macrófagos/imunologia , Macrófagos/metabolismo , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/genética , Camundongos Endogâmicos C57BL , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , TYK2 Quinase/metabolismo , TYK2 Quinase/genética , Regulação da Expressão Gênica
19.
Nat Immunol ; 25(7): 1231-1244, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38898157

RESUMO

To understand the role of T cells in the pathogenesis of ulcerative colitis (UC), we analyzed colonic T cells isolated from patients with UC and controls. Here we identified colonic CD4+ and CD8+ T lymphocyte subsets with gene expression profiles resembling stem-like progenitors, previously reported in several mouse models of autoimmune disease. Stem-like T cells were increased in inflamed areas compared to non-inflamed regions from the same patients. Furthermore, TCR sequence analysis indicated stem-like T cells were clonally related to proinflammatory T cells, suggesting their involvement in sustaining effectors that drive inflammation. Using an adoptive transfer colitis model in mice, we demonstrated that CD4+ T cells deficient in either BCL-6 or TCF1, transcription factors that promote T cell stemness, had decreased colon T cells and diminished pathogenicity. Our results establish a strong association between stem-like T cell populations and UC pathogenesis, highlighting the potential of targeting this population to improve clinical outcomes.


Assuntos
Colite Ulcerativa , Fator 1-alfa Nuclear de Hepatócito , Colite Ulcerativa/imunologia , Colite Ulcerativa/patologia , Humanos , Animais , Camundongos , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Fator 1-alfa Nuclear de Hepatócito/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD4-Positivos/imunologia , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6/genética , Células-Tronco/imunologia , Células-Tronco/metabolismo , Feminino , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Camundongos Knockout , Colo/imunologia , Colo/patologia , Masculino , Camundongos Endogâmicos C57BL , Transferência Adotiva , Modelos Animais de Doenças , Adulto , Pessoa de Meia-Idade
20.
Nat Immunol ; 25(7): 1207-1217, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38802512

RESUMO

The contribution of γδ T cells to immune responses is associated with rapid secretion of interferon-γ (IFN-γ). Here, we show a perinatal thymic wave of innate IFN-γ-producing γδ T cells that express CD8αß heterodimers and expand in preclinical models of infection and cancer. Optimal CD8αß+ γδ T cell development is directed by low T cell receptor signaling and through provision of interleukin (IL)-4 and IL-7. This population is pathologically relevant as overactive, or constitutive, IL-7R-STAT5B signaling promotes a supraphysiological accumulation of CD8αß+ γδ T cells in the thymus and peripheral lymphoid organs in two mouse models of T cell neoplasia. Likewise, CD8αß+ γδ T cells define a distinct subset of human T cell acute lymphoblastic leukemia pediatric patients. This work characterizes the normal and malignant development of CD8αß+ γδ T cells that are enriched in early life and contribute to innate IFN-γ responses to infection and cancer.


Assuntos
Imunidade Inata , Interferon gama , Receptores de Antígenos de Linfócitos T gama-delta , Receptores de Interleucina-7 , Fator de Transcrição STAT5 , Timo , Animais , Interferon gama/metabolismo , Interferon gama/imunologia , Camundongos , Humanos , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Timo/imunologia , Receptores de Interleucina-7/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/imunologia , Camundongos Endogâmicos C57BL , Linfócitos T CD8-Positivos/imunologia , Camundongos Knockout , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Antígenos CD8/metabolismo , Feminino , Linfócitos Intraepiteliais/imunologia , Linfócitos Intraepiteliais/metabolismo , Interleucina-7/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa