Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43.136
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 42(1): 647-677, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38424658

ABSTRACT

Lymphocytes spanning the entire innate-adaptive spectrum can stably reside in tissues and constitute an integral component of the local defense network against immunological challenges. In tight interactions with the epithelium and endothelium, tissue-resident lymphocytes sense antigens and alarmins elicited by infectious microbes and abiotic stresses at barrier sites and mount effector responses to restore tissue homeostasis. Of note, such a host cell-directed immune defense system has been recently demonstrated to surveil epithelial cell transformation and carcinoma development, as well as cancer cell metastasis at selected distant organs, and thus represents a primordial cancer immune defense module. Here we review how distinct lineages of tissue-resident innate lymphoid cells, innate-like T cells, and adaptive T cells participate in a form of multilayered cancer immunity in murine models and patients, and how their convergent effector programs may be targeted through both shared and private regulatory pathways for cancer immunotherapy.


Subject(s)
Immunity, Innate , Neoplasms , Humans , Animals , Neoplasms/immunology , Neoplasms/therapy , Lymphocytes/immunology , Lymphocytes/metabolism , Tumor Microenvironment/immunology , Adaptive Immunity , Immunotherapy/methods
2.
Annu Rev Immunol ; 38: 365-395, 2020 04 26.
Article in English | MEDLINE | ID: mdl-31986070

ABSTRACT

Sialic acid-binding immunoglobulin-type lectins (Siglecs) are expressed on the majority of white blood cells of the immune system and play critical roles in immune cell signaling. Through recognition of sialic acid-containing glycans as ligands, they help the immune system distinguish between self and nonself. Because of their restricted cell type expression and roles as checkpoints in immune cell responses in human diseases such as cancer, asthma, allergy, neurodegeneration, and autoimmune diseases they have gained attention as targets for therapeutic interventions. In this review we describe the Siglec family, its roles in regulation of immune cell signaling, current efforts to define its roles in disease processes, and approaches to target Siglecs for treatment of human disease.


Subject(s)
Disease Susceptibility , Immune Checkpoint Proteins/genetics , Immune Checkpoint Proteins/metabolism , Immunomodulation , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Signal Transduction , Animals , Biomarkers , Humans , Immune System/immunology , Immune System/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism
3.
Annu Rev Immunol ; 38: 759-784, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340572

ABSTRACT

The signaling lipid sphingosine 1-phosphate (S1P) plays critical roles in an immune response. Drugs targeting S1P signaling have been remarkably successful in treatment of multiple sclerosis, and they have shown promise in clinical trials for colitis and psoriasis. One mechanism of these drugs is to block lymphocyte exit from lymph nodes, where lymphocytes are initially activated, into circulation, from which lymphocytes can reach sites of inflammation. Indeed, S1P can be considered a circulation marker, signaling to immune cells to help them find blood and lymphatic vessels, and to endothelial cells to stabilize the vasculature. That said, S1P plays pleiotropic roles in the immune response, and it will be important to build an integrated view of how S1P shapes inflammation. S1P can function so effectively because its distribution is exquisitely tightly controlled. Here we review how S1P gradients regulate immune cell exit from tissues, with particular attention to key outstanding questions in the field.


Subject(s)
Cell Movement/immunology , Immune System/immunology , Immune System/metabolism , Lysophospholipids/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Animals , Biomarkers , Humans , Immune System/cytology , Lymphocyte Activation/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Sphingosine/metabolism
4.
Annu Rev Immunol ; 34: 203-42, 2016 05 20.
Article in English | MEDLINE | ID: mdl-26907216

ABSTRACT

The continuous migration of immune cells between lymphoid and nonlymphoid organs is a key feature of the immune system, facilitating the distribution of effector cells within nearly all compartments of the body. Furthermore, reaching their correct position within primary, secondary, or tertiary lymphoid organs is a prerequisite to ensure immune cells' unimpaired differentiation, maturation, and selection, as well as their activation or functional silencing. The superfamilies of chemokines and chemokine receptors are of major importance in guiding immune cells to and within lymphoid and nonlymphoid tissues. In this review we focus on the role of the chemokine system in the migration dynamics of immune cells within lymphoid organs at the steady state and on how these dynamics are affected by infectious and inflammatory processes.


Subject(s)
Chemokines/immunology , Immune System , Infections/immunology , Inflammation/immunology , Lymphocytes/immunology , Lymphoid Tissue/immunology , Receptors, Chemokine/immunology , Animals , Cell Communication , Cell Movement , Humans , Lymphocyte Activation
5.
Annu Rev Immunol ; 34: 299-316, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27168240

ABSTRACT

The discovery of tissue-resident innate lymphoid cell populations effecting different forms of type 1, 2, and 3 immunity; tissue repair; and immune regulation has transformed our understanding of mucosal immunity and allergy. The emerging complexity of these populations along with compounding issues of redundancy and plasticity raise intriguing questions about their precise lineage relationship. Here we review advances in mapping the emergence of these lineages from early lymphoid precursors. We discuss the identification of a common innate lymphoid cell precursor characterized by transient expression of the transcription factor PLZF, and the lineage relationships of innate lymphoid cells with conventional natural killer cells and lymphoid tissue inducer cells. We also review the rapidly growing understanding of the network of transcription factors that direct the development of these lineages.


Subject(s)
Cell Differentiation , Hypersensitivity/immunology , Immunity, Innate , Lymphocytes/immunology , Lymphoid Progenitor Cells/immunology , Animals , Cell Lineage , Cytokines/metabolism , Gene Expression Regulation/immunology , Gene Regulatory Networks , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Promyelocytic Leukemia Zinc Finger Protein , Th1 Cells/immunology , Th2 Cells/immunology
6.
Annu Rev Immunol ; 33: 505-38, 2015.
Article in English | MEDLINE | ID: mdl-25650177

ABSTRACT

Mammalian lymphoid immunity is mediated by fast and slow responders to pathogens. Fast innate lymphocytes are active within hours after infections in mucosal tissues. Slow adaptive lymphocytes are conventional T and B cells with clonal antigen receptors that function days after pathogen exposure. A transcription factor (TF) regulatory network guiding early T cell development is at the core of effector function diversification in all innate lymphocytes, and the kinetics of immune responses is set by developmental programming. Operational units within the innate lymphoid system are not classified by the types of pathogen-sensing machineries but rather by discrete effector functions programmed by regulatory TF networks. Based on the evolutionary history of TFs of the regulatory networks, fast effectors likely arose earlier in the evolution of animals to fortify body barriers, and in mammals they often develop in fetal ontogeny prior to the establishment of fully competent adaptive immunity.


Subject(s)
Immunity, Innate/physiology , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphopoiesis , Transcription Factors/metabolism , Animals , Biological Evolution , Humans , Immunity , Protein Binding/immunology , Signal Transduction
7.
Annu Rev Immunol ; 33: 747-85, 2015.
Article in English | MEDLINE | ID: mdl-25706098

ABSTRACT

Interleukin-22 (IL-22) is a recently described IL-10 family cytokine that is produced by T helper (Th) 17 cells, γδ T cells, NKT cells, and newly described innate lymphoid cells (ILCs). Knowledge of IL-22 biology has evolved rapidly since its discovery in 2000, and a role for IL-22 has been identified in numerous tissues, including the intestines, lung, liver, kidney, thymus, pancreas, and skin. IL-22 primarily targets nonhematopoietic epithelial and stromal cells, where it can promote proliferation and play a role in tissue regeneration. In addition, IL-22 regulates host defense at barrier surfaces. However, IL-22 has also been linked to several conditions involving inflammatory tissue pathology. In this review, we assess the current understanding of this cytokine, including its physiologic and pathologic effects on epithelial cell function.


Subject(s)
Interleukins/genetics , Interleukins/metabolism , Animals , Disease Susceptibility , Gene Expression Regulation , Humans , Interleukins/chemistry , Lymphocytes/immunology , Lymphocytes/metabolism , Organ Specificity/genetics , Organ Specificity/immunology , Signal Transduction , Interleukin-22
8.
Annu Rev Immunol ; 33: 563-606, 2015.
Article in English | MEDLINE | ID: mdl-25665078

ABSTRACT

In the 40 years since their discovery, dendritic cells (DCs) have been recognized as central players in immune regulation. DCs sense microbial stimuli through pathogen-recognition receptors (PRRs) and decode, integrate, and present information derived from such stimuli to T cells, thus stimulating immune responses. DCs can also regulate the quality of immune responses. Several functionally specialized subsets of DCs exist, but DCs also display functional plasticity in response to diverse stimuli. In addition to sensing pathogens via PRRs, emerging evidence suggests that DCs can also sense stress signals, such as amino acid starvation, through ancient stress and nutrient sensing pathways, to stimulate adaptive immunity. Here, I discuss these exciting advances in the context of a historic perspective on the discovery of DCs and their role in immune regulation. I conclude with a discussion of emerging areas in DC biology in the systems immunology era and suggest that the impact of DCs on immunity can be usefully contextualized in a hierarchy-of-organization model in which DCs, their receptors and signaling networks, cell-cell interactions, tissue microenvironment, and the host macroenvironment represent different levels of the hierarchy. Immunity or tolerance can then be represented as a complex function of each of these hierarchies.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Cell Communication/immunology , Cell Differentiation/immunology , Clonal Selection, Antigen-Mediated , Disease Resistance , Disease Susceptibility , Host-Pathogen Interactions/immunology , Humans , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Stress, Physiological
9.
Annu Rev Immunol ; 33: 607-42, 2015.
Article in English | MEDLINE | ID: mdl-25665079

ABSTRACT

The lymphocyte family has expanded significantly in recent years to include not only the adaptive lymphocytes (T cells, B cells) and NK cells, but also several additional innate lymphoid cell (ILC) types. ILCs lack clonally distributed antigen receptors characteristic of adaptive lymphocytes and instead respond exclusively to signaling via germline-encoded receptors. ILCs resemble T cells more closely than any other leukocyte lineage at the transcriptome level and express many elements of the core T cell transcriptional program, including Notch, Gata3, Tcf7, and Bcl11b. We present our current understanding of the shared and distinct transcriptional regulatory mechanisms involved in the development of adaptive T lymphocytes and closely related ILCs. We discuss the possibility that a core set of transcriptional regulators common to ILCs and T cells establish enhancers that enable implementation of closely aligned effector pathways. Studies of the transcriptional regulation of lymphopoiesis will support the development of novel therapeutic approaches to correct early lymphoid developmental defects and aberrant lymphocyte function.


Subject(s)
Adaptive Immunity/genetics , Cell Lineage/genetics , Gene Expression Regulation , Immunity, Innate/genetics , Lymphocytes/immunology , Lymphocytes/metabolism , Transcription, Genetic , Animals , Cell Differentiation , Humans , Lymphocytes/cytology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism
10.
Annu Rev Immunol ; 33: 677-713, 2015.
Article in English | MEDLINE | ID: mdl-25665077

ABSTRACT

Dynamic tuning of cellular responsiveness as a result of repeated stimuli improves the ability of cells to distinguish physiologically meaningful signals from each other and from noise. In particular, lymphocyte activation thresholds are subject to tuning, which contributes to maintaining tolerance to self-antigens and persisting foreign antigens, averting autoimmunity and immune pathogenesis, but allowing responses to strong, structured perturbations that are typically associated with acute infection. Such tuning is also implicated in conferring flexibility to positive selection in the thymus, in controlling the magnitude of the immune response, and in generating memory cells. Additional functional properties are dynamically and differentially tuned in parallel via subthreshold contact interactions between developing or mature lymphocytes and self-antigen-presenting cells. These interactions facilitate and regulate lymphocyte viability, maintain their functional integrity, and influence their responses to foreign antigens and accessory signals, qualitatively and quantitatively. Bidirectional tuning of T cells and antigen-presenting cells leads to the definition of homeostatic set points, thus maximizing clonal diversity.


Subject(s)
Lymphocytes/immunology , Lymphocytes/metabolism , Animals , Cell Survival/immunology , Homeostasis , Humans , Immunologic Memory , Infections/immunology , Infections/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes/cytology , Phenotype , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism
11.
Annu Rev Immunol ; 33: 291-353, 2015.
Article in English | MEDLINE | ID: mdl-25861976

ABSTRACT

Ion channels and transporters mediate the transport of charged ions across hydrophobic lipid membranes. In immune cells, divalent cations such as calcium, magnesium, and zinc have important roles as second messengers to regulate intracellular signaling pathways. By contrast, monovalent cations such as sodium and potassium mainly regulate the membrane potential, which indirectly controls the influx of calcium and immune cell signaling. Studies investigating human patients with mutations in ion channels and transporters, analysis of gene-targeted mice, or pharmacological experiments with ion channel inhibitors have revealed important roles of ionic signals in lymphocyte development and in innate and adaptive immune responses. We here review the mechanisms underlying the function of ion channels and transporters in lymphocytes and innate immune cells and discuss their roles in lymphocyte development, adaptive and innate immune responses, and autoimmunity, as well as recent efforts to develop pharmacological inhibitors of ion channels for immunomodulatory therapy.


Subject(s)
Adaptive Immunity/physiology , Immunity, Innate/physiology , Ion Channels/metabolism , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Hypersensitivity/metabolism , Immunologic Deficiency Syndromes/drug therapy , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Immunotherapy/methods , Ion Channels/genetics , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Molecular Targeted Therapy , Mutation , Signal Transduction
12.
Cell ; 185(5): 916-938.e58, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35216673

ABSTRACT

Treatment of severe COVID-19 is currently limited by clinical heterogeneity and incomplete description of specific immune biomarkers. We present here a comprehensive multi-omic blood atlas for patients with varying COVID-19 severity in an integrated comparison with influenza and sepsis patients versus healthy volunteers. We identify immune signatures and correlates of host response. Hallmarks of disease severity involved cells, their inflammatory mediators and networks, including progenitor cells and specific myeloid and lymphocyte subsets, features of the immune repertoire, acute phase response, metabolism, and coagulation. Persisting immune activation involving AP-1/p38MAPK was a specific feature of COVID-19. The plasma proteome enabled sub-phenotyping into patient clusters, predictive of severity and outcome. Systems-based integrative analyses including tensor and matrix decomposition of all modalities revealed feature groupings linked with severity and specificity compared to influenza and sepsis. Our approach and blood atlas will support future drug development, clinical trial design, and personalized medicine approaches for COVID-19.


Subject(s)
Biomarkers/blood , COVID-19/pathology , Proteome/analysis , Adult , Blood Proteins/metabolism , COVID-19/blood , COVID-19/virology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Female , Humans , Influenza, Human/blood , Influenza, Human/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Machine Learning , Male , Middle Aged , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Monocytes/immunology , Monocytes/metabolism , Principal Component Analysis , SARS-CoV-2/isolation & purification , Sepsis/blood , Sepsis/pathology , Severity of Illness Index , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
13.
Nat Immunol ; 25(8): 1460-1473, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38956380

ABSTRACT

Group 1 innate lymphoid cells (ILC1s) are cytotoxic and interferon gamma-producing lymphocytes lacking antigen-specific receptors, which include ILC1s and natural killer (NK) cells. In mice, ILC1s differ from NK cells, as they develop independently of the NK-specifying transcription factor EOMES, while requiring the repressor ZFP683 (ZNF683 in humans) for tissue residency. Here we identify highly variable ILC1 subtypes across tissues through investigation of human ILC1 diversity by single-cell RNA sequencing and flow cytometry. The intestinal epithelium contained abundant mature EOMES- ILC1s expressing PRDM1 rather than ZNF683, alongside a few immature TCF7+PRDM1- ILC1s. Other tissues harbored NK cells expressing ZNF683 and EOMES transcripts; however, EOMES protein content was variable. These ZNF683+ NK cells are tissue-imprinted NK cells phenotypically resembling ILC1s. The tissue ILC1-NK spectrum also encompassed conventional NK cells and NK cells distinguished by PTGDS expression. These findings establish a foundation for evaluating phenotypic and functional changes within the NK-ILC1 spectrum in diseases.


Subject(s)
Immunity, Innate , Killer Cells, Natural , Lymphocytes , Positive Regulatory Domain I-Binding Factor 1 , T-Box Domain Proteins , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Lymphocytes/immunology , Lymphocytes/metabolism , Single-Cell Analysis , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Animals , Mice , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics
14.
Nat Immunol ; 25(9): 1565-1579, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39103576

ABSTRACT

Nucleophosmin 1 (NPM1) is commonly mutated in myelodysplastic syndrome (MDS) and acute myeloid leukemia. Concurrent inflammatory bowel diseases (IBD) and MDS are common, indicating a close relationship between IBD and MDS. Here we examined the function of NPM1 in IBD and colitis-associated colorectal cancer (CAC). NPM1 expression was reduced in patients with IBD. Npm1+/- mice were more susceptible to acute colitis and experimentally induced CAC than littermate controls. Npm1 deficiency impaired the function of interleukin-22 (IL-22)-producing group three innate lymphoid cells (ILC3s). Mice lacking Npm1 in ILC3s exhibited decreased IL-22 production and accelerated development of colitis. NPM1 was important for mitochondrial biogenesis and metabolism by oxidative phosphorylation in ILC3s. Further experiments revealed that NPM1 cooperates with p65 to promote mitochondrial transcription factor A (TFAM) transcription in ILC3s. Overexpression of Npm1 in mice enhanced ILC3 function and reduced the severity of dextran sulfate sodium-induced colitis. Thus, our findings indicate that NPM1 in ILC3s protects against IBD by regulating mitochondrial metabolism through a p65-TFAM axis.


Subject(s)
Colitis , Immunity, Mucosal , Mice, Knockout , Mitochondria , Nuclear Proteins , Nucleophosmin , Oxidative Phosphorylation , Animals , Mitochondria/metabolism , Mice , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Humans , Colitis/immunology , Colitis/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred C57BL , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Interleukin-22 , Immunity, Innate , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Dextran Sulfate , Male , Interleukins/metabolism , Interleukins/genetics , Interleukins/immunology , Female
15.
Cell ; 184(13): 3573-3587.e29, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34062119

ABSTRACT

The simultaneous measurement of multiple modalities represents an exciting frontier for single-cell genomics and necessitates computational methods that can define cellular states based on multimodal data. Here, we introduce "weighted-nearest neighbor" analysis, an unsupervised framework to learn the relative utility of each data type in each cell, enabling an integrative analysis of multiple modalities. We apply our procedure to a CITE-seq dataset of 211,000 human peripheral blood mononuclear cells (PBMCs) with panels extending to 228 antibodies to construct a multimodal reference atlas of the circulating immune system. Multimodal analysis substantially improves our ability to resolve cell states, allowing us to identify and validate previously unreported lymphoid subpopulations. Moreover, we demonstrate how to leverage this reference to rapidly map new datasets and to interpret immune responses to vaccination and coronavirus disease 2019 (COVID-19). Our approach represents a broadly applicable strategy to analyze single-cell multimodal datasets and to look beyond the transcriptome toward a unified and multimodal definition of cellular identity.


Subject(s)
SARS-CoV-2/immunology , Single-Cell Analysis/methods , 3T3 Cells , Animals , COVID-19/immunology , Cell Line , Gene Expression Profiling/methods , Humans , Immunity/immunology , Leukocytes, Mononuclear/immunology , Lymphocytes/immunology , Mice , Sequence Analysis, RNA/methods , Transcriptome/immunology , Vaccination
16.
Cell ; 184(19): 5015-5030.e16, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34407392

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) regulate immunity and inflammation, yet their role in cancer remains elusive. Here, we identify that colorectal cancer (CRC) manifests with altered ILC3s that are characterized by reduced frequencies, increased plasticity, and an imbalance with T cells. We evaluated the consequences of these changes in mice and determined that a dialog between ILC3s and T cells via major histocompatibility complex class II (MHCII) is necessary to support colonization with microbiota that subsequently induce type-1 immunity in the intestine and tumor microenvironment. As a result, mice lacking ILC3-specific MHCII develop invasive CRC and resistance to anti-PD-1 immunotherapy. Finally, humans with dysregulated intestinal ILC3s harbor microbiota that fail to induce type-1 immunity and immunotherapy responsiveness when transferred to mice. Collectively, these data define a protective role for ILC3s in cancer and indicate that their inherent disruption in CRC drives dysfunctional adaptive immunity, tumor progression, and immunotherapy resistance.


Subject(s)
Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Disease Progression , Immunity, Innate , Immunotherapy , Lymphocytes/immunology , Animals , Cell Communication/drug effects , Cell Plasticity/drug effects , Colonic Neoplasms/microbiology , Feces/microbiology , Histocompatibility Antigens Class II/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunity, Innate/drug effects , Inflammation/immunology , Inflammation/pathology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Intestines/pathology , Lymphocytes/drug effects , Mice, Inbred C57BL , Microbiota/drug effects , Neoplasm Invasiveness , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tissue Donors
17.
Nat Immunol ; 24(8): 1256-1264, 2023 08.
Article in English | MEDLINE | ID: mdl-37400674

ABSTRACT

Innate lymphoid cells (ILCs) can quickly switch from a quiescent state to an active state and rapidly produce effector molecules that provide critical early immune protection. How the post-transcriptional machinery processes different stimuli and initiates robust gene expression in ILCs is poorly understood. Here, we show that deletion of the N6-methyladenosine (m6A) writer protein METTL3 has little impact on ILC homeostasis or cytokine-induced ILC1 or ILC3 responses but significantly diminishes ILC2 proliferation, migration and effector cytokine production and results in impaired antihelminth immunity. m6A RNA modification supports an increase in cell size and transcriptional activity in activated ILC2s but not in ILC1s or ILC3s. Among other transcripts, the gene encoding the transcription factor GATA3 is highly m6A methylated in ILC2s. Targeted m6A demethylation destabilizes nascent Gata3 mRNA and abolishes the upregulation of GATA3 and ILC2 activation. Our study suggests a lineage-specific requirement of m6A for ILC2 responses.


Subject(s)
Immunity, Innate , Lymphocytes , Cytokines/metabolism , Homeostasis , Immunity, Innate/genetics , Immunity, Innate/immunology , Lymphocytes/immunology , RNA/metabolism , Animals , Mice
18.
Annu Rev Immunol ; 31: 345-85, 2013.
Article in English | MEDLINE | ID: mdl-23516983

ABSTRACT

Autoimmune diseases currently affect 5-7% of the world's population; in most diseases there are circulating autoantibodies. Brain-reactive antibodies are present in approximately 2-3% of the general population but do not usually contribute to brain pathology. These antibodies penetrate brain tissue only early in development or under pathologic conditions. This restriction on their pathogenicity and the lack of correlation between serum titers and brain pathology have, no doubt, contributed to a delayed appreciation of the contribution of autoantibodies in diseases of the central nervous system. Nonetheless, it is increasingly clear that antibodies can cause damage in the brain and likely initiate or aggravate multiple neurologic conditions; brain-reactive antibodies contribute to symptomatology in autoimmune disease, infectious disease, and malignancy.


Subject(s)
Autoantibodies/metabolism , Brain/immunology , Brain/pathology , Hypoxia-Ischemia, Brain/immunology , Hypoxia-Ischemia, Brain/pathology , Animals , Antigen-Antibody Reactions/immunology , Brain/metabolism , Cells, Cultured , Disease Models, Animal , Humans , Hypoxia-Ischemia, Brain/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology
19.
Nat Immunol ; 23(2): 251-261, 2022 02.
Article in English | MEDLINE | ID: mdl-35102343

ABSTRACT

Tumor necrosis factor (TNF) drives chronic inflammation and cell death in the intestine, and blocking TNF is a therapeutic approach in inflammatory bowel disease (IBD). Despite this knowledge, the pathways that protect the intestine from TNF are incompletely understood. Here we demonstrate that group 3 innate lymphoid cells (ILC3s) protect the intestinal epithelium from TNF-induced cell death. This occurs independent of interleukin-22 (IL-22), and we identify that ILC3s are a dominant source of heparin-binding epidermal growth factor-like growth factor (HB-EGF). ILC3s produce HB-EGF in response to prostaglandin E2 (PGE2) and engagement of the EP2 receptor. Mice lacking ILC3-derived HB-EGF exhibit increased susceptibility to TNF-mediated epithelial cell death and experimental intestinal inflammation. Finally, human ILC3s produce HB-EGF and are reduced from the inflamed intestine. These results define an essential role for ILC3-derived HB-EGF in protecting the intestine from TNF and indicate that disruption of this pathway contributes to IBD.


Subject(s)
Heparin-binding EGF-like Growth Factor/immunology , Immunity, Innate/immunology , Inflammation/immunology , Intestines/immunology , Lymphocytes/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Epithelial Cells/immunology , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Signal Transduction/immunology
20.
Nat Immunol ; 23(2): 262-274, 2022 02.
Article in English | MEDLINE | ID: mdl-35102345

ABSTRACT

Tumors poorly infiltrated by T cells are more resistant to immunogenic chemotherapies and checkpoint inhibition than highly infiltrated tumors. Using murine models, we found that CCR6+ type 3 innate lymphoid cells (ILC3s) can trigger an increase in the number of T cells infiltrating a tumor. Shortly after administration of cisplatin chemotherapy, production of the chemokine CCL20 and proinflammatory cytokine IL-1ß at the tumor site led to the recruitment and activation of ILC3s. Within the tumor, ILC3 production of the chemokine CXCL10 was responsible for the recruitment of CD4+ and CD8+ T lymphocytes to the tumor. ILC3-dependent infiltration of T cells was essential for antitumor immune responses and increased the efficacy of checkpoint inhibition. Thus, we reveal an essential role of CCL20 and IL-1ß, which promote ILC3-dependent antitumor immunity and enhance tumor sensitivity to immunotherapy.


Subject(s)
Immunity, Innate/immunology , Lymphocytes/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cytokines/immunology , Female , Humans , Immune Checkpoint Inhibitors/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL