Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24.769
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 37: 295-324, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30649989

ABSTRACT

Cytokines are secreted or otherwise released polypeptide factors that exert autocrine and/or paracrine actions, with most cytokines acting in the immune and/or hematopoietic system. They are typically pleiotropic, controlling development, cell growth, survival, and/or differentiation. Correspondingly, cytokines are clinically important, and augmenting or attenuating cytokine signals can have deleterious or therapeutic effects. Besides physiological fine-tuning of cytokine signals, altering the nature or potency of the signal can be important in pathophysiological responses and can also provide novel therapeutic approaches. Here, we give an overview of cytokines, their signaling and actions, and the physiological mechanisms and pharmacologic strategies to fine-tune their actions. In particular, the differential utilization of STAT proteins by a single cytokine or by different cytokines and STAT dimerization versus tetramerization are physiological mechanisms of fine-tuning, whereas anticytokine and anticytokine receptor antibodies and cytokines with altered activities, including cytokine superagonists, partial agonists, and antagonists, represent new ways of fine-tuning cytokine signals.


Subject(s)
Cytokines/metabolism , Immunotherapy/trends , Animals , Cytokines/genetics , Humans , Immunity, Humoral , Immunomodulation , Protein Multimerization , STAT Transcription Factors/metabolism , Signal Transduction/immunology
2.
Annu Rev Immunol ; 33: 139-67, 2015.
Article in English | MEDLINE | ID: mdl-25493332

ABSTRACT

Cytokines exert a vast array of immunoregulatory actions critical to human biology and disease. However, the desired immunotherapeutic effects of native cytokines are often mitigated by toxicity or lack of efficacy, either of which results from cytokine receptor pleiotropy and/or undesired activation of off-target cells. As our understanding of the structural principles of cytokine-receptor interactions has advanced, mechanism-based manipulation of cytokine signaling through protein engineering has become an increasingly feasible and powerful approach. Modified cytokines, both agonists and antagonists, have been engineered with narrowed target cell specificities, and they have also yielded important mechanistic insights into cytokine biology and signaling. Here we review the theory and practice of cytokine engineering and rationalize the mechanisms of several engineered cytokines in the context of structure. We discuss specific examples of how structure-based cytokine engineering has opened new opportunities for cytokines as drugs, with a focus on the immunotherapeutic cytokines interferon, interleukin-2, and interleukin-4.


Subject(s)
Cytokines/genetics , Cytokines/metabolism , Genetic Engineering , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Animals , Cytokines/chemistry , Extracellular Space/metabolism , Humans , Intracellular Space/metabolism , Protein Binding , Protein Transport , Receptors, Cytokine/chemistry , Signal Transduction
3.
Cell ; 182(3): 641-654.e20, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32615085

ABSTRACT

Targeting glycolysis has been considered therapeutically intractable owing to its essential housekeeping role. However, the context-dependent requirement for individual glycolytic steps has not been fully explored. We show that CRISPR-mediated targeting of glycolysis in T cells in mice results in global loss of Th17 cells, whereas deficiency of the glycolytic enzyme glucose phosphate isomerase (Gpi1) selectively eliminates inflammatory encephalitogenic and colitogenic Th17 cells, without substantially affecting homeostatic microbiota-specific Th17 cells. In homeostatic Th17 cells, partial blockade of glycolysis upon Gpi1 inactivation was compensated by pentose phosphate pathway flux and increased mitochondrial respiration. In contrast, inflammatory Th17 cells experience a hypoxic microenvironment known to limit mitochondrial respiration, which is incompatible with loss of Gpi1. Our study suggests that inhibiting glycolysis by targeting Gpi1 could be an effective therapeutic strategy with minimum toxicity for Th17-mediated autoimmune diseases, and, more generally, that metabolic redundancies can be exploited for selective targeting of disease processes.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Glucose-6-Phosphate Isomerase/metabolism , Glycolysis/genetics , Oxidative Phosphorylation , Pentose Phosphate Pathway/physiology , Th17 Cells/metabolism , Animals , Cell Hypoxia/genetics , Cell Hypoxia/immunology , Chimera/genetics , Chromatography, Gas , Chromatography, Liquid , Clostridium Infections/immunology , Cytokines/deficiency , Cytokines/genetics , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Glucose-6-Phosphate Isomerase/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Glycolysis/immunology , Homeostasis/genetics , Homeostasis/immunology , Inflammation/genetics , Inflammation/immunology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/microbiology , Pentose Phosphate Pathway/genetics , Pentose Phosphate Pathway/immunology , RNA-Seq , Single-Cell Analysis , Th17 Cells/immunology , Th17 Cells/pathology
4.
Nat Immunol ; 23(1): 75-85, 2022 01.
Article in English | MEDLINE | ID: mdl-34937930

ABSTRACT

We report a pleiotropic disease due to loss-of-function mutations in RHBDF2, the gene encoding iRHOM2, in two kindreds with recurrent infections in different organs. One patient had recurrent pneumonia but no colon involvement, another had recurrent infectious hemorrhagic colitis but no lung involvement and the other two experienced recurrent respiratory infections. Loss of iRHOM2, a rhomboid superfamily member that regulates the ADAM17 metalloproteinase, caused defective ADAM17-dependent cleavage and release of cytokines, including tumor-necrosis factor and amphiregulin. To understand the diverse clinical phenotypes, we challenged Rhbdf2-/- mice with Pseudomonas aeruginosa by nasal gavage and observed more severe pneumonia, whereas infection with Citrobacter rodentium caused worse inflammatory colitis than in wild-type mice. The fecal microbiota in the colitis patient had characteristic oral species that can predispose to colitis. Thus, a human immunodeficiency arising from iRHOM2 deficiency causes divergent disease phenotypes that can involve the local microbial environment.


Subject(s)
ADAM17 Protein/genetics , Carrier Proteins/genetics , Primary Immunodeficiency Diseases/genetics , A549 Cells , Animals , Child , Child, Preschool , Citrobacter rodentium/pathogenicity , Colitis/genetics , Cytokines/genetics , Enterobacteriaceae Infections/genetics , Female , HEK293 Cells , Humans , Infant, Newborn , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation/genetics , Pseudomonas Infections/genetics , Pseudomonas aeruginosa/pathogenicity , Signal Transduction/genetics
5.
Cell ; 178(5): 1041-1056, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442399

ABSTRACT

The current understanding of inflammatory bowel disease (IBD) pathogenesis implicates a complex interaction between host genetics, host immunity, microbiome, and environmental exposures. Mechanisms gleaned from genetics and molecular pathogenesis offer clues to the critical triggers of mucosal inflammation and guide the development of therapeutic interventions. A complex network of interactions between host genetic factors, microbes, and microbial metabolites governs intestinal homeostasis, making classification and mechanistic dissection of involved pathways challenging. In this Review, we discuss these challenges, areas of active translation, and opportunities for development of next-generation therapies.


Subject(s)
Inflammatory Bowel Diseases/pathology , Microbiota , Adaptive Immunity , Animals , Bacteria/genetics , Bacteria/metabolism , Biological Products/pharmacology , Cytokines/genetics , Cytokines/metabolism , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Intestines/immunology , Intestines/microbiology , Microbiota/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
Cell ; 178(5): 1189-1204.e23, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442407

ABSTRACT

CD8 T cells play essential roles in anti-tumor immune responses. Here, we performed genome-scale CRISPR screens in CD8 T cells directly under cancer immunotherapy settings and identified regulators of tumor infiltration and degranulation. The in vivo screen robustly re-identified canonical immunotherapy targets such as PD-1 and Tim-3, along with genes that have not been characterized in T cells. The infiltration and degranulation screens converged on an RNA helicase Dhx37. Dhx37 knockout enhanced the efficacy of antigen-specific CD8 T cells against triple-negative breast cancer in vivo. Immunological characterization in mouse and human CD8 T cells revealed that DHX37 suppresses effector functions, cytokine production, and T cell activation. Transcriptomic profiling and biochemical interrogation revealed a role for DHX37 in modulating NF-κB. These data demonstrate high-throughput in vivo genetic screens for immunotherapy target discovery and establishes DHX37 as a functional regulator of CD8 T cells.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , RNA Helicases/genetics , Animals , Breast Neoplasms/pathology , Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Cytokines/genetics , Cytokines/metabolism , Female , Humans , Immunologic Memory , Immunotherapy , Male , Mice , Mice, Knockout , NF-kappa B/metabolism , Programmed Cell Death 1 Receptor/metabolism , RNA Helicases/deficiency , RNA, Guide, Kinetoplastida/metabolism , Transcriptome
7.
Nat Immunol ; 22(3): 287-300, 2021 03.
Article in English | MEDLINE | ID: mdl-33574617

ABSTRACT

Sub-Saharan Africa currently experiences an unprecedented wave of urbanization, which has important consequences for health and disease patterns. This study aimed to investigate and integrate the immune and metabolic consequences of rural or urban lifestyles and the role of nutritional changes associated with urban living. In a cohort of 323 healthy Tanzanians, urban as compared to rural living was associated with a pro-inflammatory immune phenotype, both at the transcript and protein levels. We identified different food-derived and endogenous circulating metabolites accounting for these differences. Serum from urban dwellers induced reprogramming of innate immune cells with higher tumor necrosis factor production upon microbial re-stimulation in an in vitro model of trained immunity. These data demonstrate important shifts toward an inflammatory phenotype associated with an urban lifestyle and provide new insights into the underlying dietary and metabolic factors, which may affect disease epidemiology in sub-Sahara African countries.


Subject(s)
Cytokines/blood , Diet, Healthy , Energy Metabolism , Immunity, Innate , Inflammation Mediators/blood , Rural Health , Urban Health , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Cytokines/genetics , Energy Metabolism/genetics , Female , Humans , Immunity, Innate/genetics , Male , Metabolome , Middle Aged , Nutritional Status , Nutritive Value , Risk Reduction Behavior , Seasons , Tanzania , Transcriptome , Tumor Necrosis Factor-alpha/blood , Urbanization , Young Adult
8.
Nat Immunol ; 22(11): 1416-1427, 2021 11.
Article in English | MEDLINE | ID: mdl-34663977

ABSTRACT

Ubiquitin-like protein ISG15 (interferon-stimulated gene 15) (ISG15) is a ubiquitin-like modifier induced during infections and involved in host defense mechanisms. Not surprisingly, many viruses encode deISGylating activities to antagonize its effect. Here we show that infection by Zika, SARS-CoV-2 and influenza viruses induce ISG15-modifying enzymes. While influenza and Zika viruses induce ISGylation, SARS-CoV-2 triggers deISGylation instead to generate free ISG15. The ratio of free versus conjugated ISG15 driven by the papain-like protease (PLpro) enzyme of SARS-CoV-2 correlates with macrophage polarization toward a pro-inflammatory phenotype and attenuated antigen presentation. In vitro characterization of purified wild-type and mutant PLpro revealed its strong deISGylating over deubiquitylating activity. Quantitative proteomic analyses of PLpro substrates and secretome from SARS-CoV-2-infected macrophages revealed several glycolytic enzymes previously implicated in the expression of inflammatory genes and pro-inflammatory cytokines, respectively. Collectively, our results indicate that altered free versus conjugated ISG15 dysregulates macrophage responses and probably contributes to the cytokine storms triggered by SARS-CoV-2.


Subject(s)
COVID-19/immunology , Cytokines/metabolism , Inflammation/immunology , Macrophages/immunology , SARS-CoV-2/physiology , Ubiquitins/metabolism , Cell Differentiation , Coronavirus Papain-Like Proteases/metabolism , Cytokines/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , Immune Evasion , Immunity, Innate , Influenza A virus/physiology , Influenza, Human/immunology , Pluripotent Stem Cells/cytology , Ubiquitination , Ubiquitins/genetics , Zika Virus/physiology , Zika Virus Infection/immunology
9.
Nat Immunol ; 22(4): 471-484, 2021 04.
Article in English | MEDLINE | ID: mdl-33664518

ABSTRACT

The diversity of regulatory T (Treg) cells in health and in disease remains unclear. Individuals with colorectal cancer harbor a subpopulation of RORγt+ Treg cells with elevated expression of ß-catenin and pro-inflammatory properties. Here we show progressive expansion of RORγt+ Treg cells in individuals with inflammatory bowel disease during inflammation and early dysplasia. Activating Wnt-ß-catenin signaling in human and murine Treg cells was sufficient to recapitulate the disease-associated increase in the frequency of RORγt+ Treg cells coexpressing multiple pro-inflammatory cytokines. Binding of the ß-catenin interacting partner, TCF-1, to DNA overlapped with Foxp3 binding at enhancer sites of pro-inflammatory pathway genes. Sustained Wnt-ß-catenin activation induced newly accessible chromatin sites in these genes and upregulated their expression. These findings indicate that TCF-1 and Foxp3 together limit the expression of pro-inflammatory genes in Treg cells. Activation of ß-catenin signaling interferes with this function and promotes the disease-associated RORγt+ Treg phenotype.


Subject(s)
Cell Proliferation , Cellular Reprogramming , Colitis, Ulcerative/metabolism , Colitis-Associated Neoplasms/metabolism , Crohn Disease/metabolism , Epigenesis, Genetic , Lymphocyte Activation , T-Lymphocytes, Regulatory/metabolism , Wnt Signaling Pathway , Animals , Case-Control Studies , Cells, Cultured , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Colitis-Associated Neoplasms/genetics , Colitis-Associated Neoplasms/immunology , Crohn Disease/genetics , Crohn Disease/immunology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype , T Cell Transcription Factor 1 , T-Lymphocytes, Regulatory/immunology
10.
Nat Immunol ; 22(1): 32-40, 2021 01.
Article in English | MEDLINE | ID: mdl-33277638

ABSTRACT

A central paradigm of immunity is that interferon (IFN)-mediated antiviral responses precede pro-inflammatory ones, optimizing host protection and minimizing collateral damage1,2. Here, we report that for coronavirus disease 2019 (COVID-19) this paradigm does not apply. By investigating temporal IFN and inflammatory cytokine patterns in 32 moderate-to-severe patients with COVID-19 hospitalized for pneumonia and longitudinally followed for the development of respiratory failure and death, we reveal that IFN-λ and type I IFN production were both diminished and delayed, induced only in a fraction of patients as they became critically ill. On the contrary, pro-inflammatory cytokines such as tumor necrosis factor (TNF), interleukin (IL)-6 and IL-8 were produced before IFNs in all patients and persisted for a prolonged time. This condition was reflected in blood transcriptomes wherein prominent IFN signatures were only seen in critically ill patients who also exhibited augmented inflammation. By comparison, in 16 patients with influenza (flu) hospitalized for pneumonia with similar clinicopathological characteristics to those of COVID-19 and 24 nonhospitalized patients with flu with milder symptoms, IFN-λ and type I IFN were robustly induced earlier, at higher levels and independently of disease severity, whereas pro-inflammatory cytokines were only acutely produced. Notably, higher IFN-λ concentrations in patients with COVID-19 correlated with lower viral load in bronchial aspirates and faster viral clearance and a higher IFN-λ to type I IFN ratio correlated with improved outcome for critically ill patients. Moreover, altered cytokine patterns in patients with COVID-19 correlated with longer hospitalization and higher incidence of critical disease and mortality compared to flu. These data point to an untuned antiviral response in COVID-19, contributing to persistent viral presence, hyperinflammation and respiratory failure.


Subject(s)
COVID-19/immunology , Immunity/immunology , Influenza, Human/immunology , Interferon Type I/immunology , Interferons/immunology , SARS-CoV-2/immunology , Antiviral Agents/immunology , Antiviral Agents/metabolism , COVID-19/genetics , COVID-19/virology , Cytokines/genetics , Cytokines/immunology , Disease Progression , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Profiling/methods , Humans , Immunity/genetics , Inflammation/genetics , Inflammation/immunology , Influenza, Human/genetics , Interferon Type I/genetics , Interferons/genetics , Length of Stay , Prognosis , SARS-CoV-2/physiology , Viral Load/genetics , Viral Load/immunology , Interferon Lambda
11.
Nat Immunol ; 22(2): 216-228, 2021 02.
Article in English | MEDLINE | ID: mdl-33462454

ABSTRACT

CD4+ effector lymphocytes (Teff) are traditionally classified by the cytokines they produce. To determine the states that Teff cells actually adopt in frontline tissues in vivo, we applied single-cell transcriptome and chromatin analyses to colonic Teff cells in germ-free or conventional mice or in mice after challenge with a range of phenotypically biasing microbes. Unexpected subsets were marked by the expression of the interferon (IFN) signature or myeloid-specific transcripts, but transcriptome or chromatin structure could not resolve discrete clusters fitting classic helper T cell (TH) subsets. At baseline or at different times of infection, transcripts encoding cytokines or proteins commonly used as TH markers were distributed in a polarized continuum, which was functionally validated. Clones derived from single progenitors gave rise to both IFN-γ- and interleukin (IL)-17-producing cells. Most of the transcriptional variance was tied to the infecting agent, independent of the cytokines produced, and chromatin variance primarily reflected activities of activator protein (AP)-1 and IFN-regulatory factor (IRF) transcription factor (TF) families, not the canonical subset master regulators T-bet, GATA3 or RORγ.


Subject(s)
Bacteria/pathogenicity , Bacterial Infections/microbiology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/parasitology , Colon/microbiology , Colon/parasitology , Gastrointestinal Microbiome , Heligmosomatoidea/pathogenicity , Intestinal Diseases, Parasitic/parasitology , Animals , Bacteria/immunology , Bacterial Infections/genetics , Bacterial Infections/immunology , Bacterial Infections/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chromatin/genetics , Chromatin/metabolism , Citrobacter rodentium/immunology , Citrobacter rodentium/pathogenicity , Colon/immunology , Colon/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Heligmosomatoidea/immunology , Host-Pathogen Interactions , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Intestinal Diseases, Parasitic/genetics , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Nematospiroides dubius/immunology , Nematospiroides dubius/pathogenicity , Nippostrongylus/immunology , Nippostrongylus/pathogenicity , Phenotype , Salmonella enterica/immunology , Salmonella enterica/pathogenicity , Single-Cell Analysis , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcriptome
12.
Nat Immunol ; 21(12): 1528-1539, 2020 12.
Article in English | MEDLINE | ID: mdl-33020661

ABSTRACT

Mutations that impact immune cell migration and result in immune deficiency illustrate the importance of cell movement in host defense. In humans, loss-of-function mutations in DOCK8, a guanine exchange factor involved in hematopoietic cell migration, lead to immunodeficiency and, paradoxically, allergic disease. Here, we demonstrate that, like humans, Dock8-/- mice have a profound type 2 CD4+ helper T (TH2) cell bias upon pulmonary infection with Cryptococcus neoformans and other non-TH2 stimuli. We found that recruited Dock8-/-CX3CR1+ mononuclear phagocytes are exquisitely sensitive to migration-induced cell shattering, releasing interleukin (IL)-1ß that drives granulocyte-macrophage colony-stimulating factor (GM-CSF) production by CD4+ T cells. Blocking IL-1ß, GM-CSF or caspase activation eliminated the type-2 skew in mice lacking Dock8. Notably, treatment of infected wild-type mice with apoptotic cells significantly increased GM-CSF production and TH2 cell differentiation. This reveals an important role for cell death in driving type 2 signals during infection, which may have implications for understanding the etiology of type 2 CD4+ T cell responses in allergic disease.


Subject(s)
Guanine Nucleotide Exchange Factors/deficiency , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Biomarkers , Caspases/metabolism , Cell Movement/genetics , Cell Movement/immunology , Cytokines/genetics , Cytokines/metabolism , Disease Susceptibility , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunophenotyping , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Signal Transduction
13.
Annu Rev Immunol ; 28: 445-89, 2010.
Article in English | MEDLINE | ID: mdl-20192806

ABSTRACT

CD4 T cells play critical roles in mediating adaptive immunity to a variety of pathogens. They are also involved in autoimmunity, asthma, and allergic responses as well as in tumor immunity. During TCR activation in a particular cytokine milieu, naive CD4 T cells may differentiate into one of several lineages of T helper (Th) cells, including Th1, Th2, Th17, and iTreg, as defined by their pattern of cytokine production and function. In this review, we summarize the discovery, functions, and relationships among Th cells; the cytokine and signaling requirements for their development; the networks of transcription factors involved in their differentiation; the epigenetic regulation of their key cytokines and transcription factors; and human diseases involving defective CD4 T cell differentiation.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Lineage , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Humans , Signal Transduction , Transcription Factors/immunology , Transcription Factors/metabolism
14.
Nat Immunol ; 20(6): 724-735, 2019 06.
Article in English | MEDLINE | ID: mdl-30936494

ABSTRACT

Regulatory T cells (Treg cells) maintain host self-tolerance but are a major barrier to effective cancer immunotherapy. Treg cells subvert beneficial anti-tumor immunity by modulating inhibitory receptor expression on tumor-infiltrating lymphocytes (TILs); however, the underlying mediators and mechanisms have remained elusive. Here, we found that the cytokines IL-10 and IL-35 (Ebi3-IL-12α heterodimer) were divergently expressed by Treg cell subpopulations in the tumor microenvironment (TME) and cooperatively promoted intratumoral T cell exhaustion by modulating several inhibitory receptor expression and exhaustion-associated transcriptomic signature of CD8+ TILs. While expression of BLIMP1 (encoded by Prdm1) was a common target, IL-10 and IL-35 differentially affected effector T cell versus memory T cell fates, respectively, highlighting their differential, partially overlapping but non-redundant regulation of anti-tumor immunity. Our results reveal previously unappreciated cooperative roles for Treg cell-derived IL-10 and IL-35 in promoting BLIMP1-dependent exhaustion of CD8+ TILs that limits effective anti-tumor immunity.


Subject(s)
Immunity, Cellular , Interleukin-10/metabolism , Interleukins/metabolism , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Adoptive Transfer , Animals , Cytokines/genetics , Cytokines/metabolism , Gene Expression Profiling , Humans , Melanoma, Experimental , Mice , Neoplasms/pathology , Signal Transduction , Transcriptome
15.
Nat Immunol ; 20(3): 288-300, 2019 03.
Article in English | MEDLINE | ID: mdl-30692620

ABSTRACT

Although tissue-resident memory T cells (TRM cells) have been shown to regulate host protection in infectious disorders, their function in inflammatory bowel disease (IBD) remains to be investigated. Here we characterized TRM cells in human IBD and in experimental models of intestinal inflammation. Pro-inflammatory TRM cells accumulated in the mucosa of patients with IBD, and the presence of CD4+CD69+CD103+ TRM cells was predictive of the development of flares. In vivo, functional impairment of TRM cells in mice with double knockout of the TRM-cell-associated transcription factors Hobit and Blimp-1 attenuated disease in several models of colitis, due to impaired cross-talk between the adaptive and innate immune system. Finally, depletion of TRM cells led to a suppression of colitis activity. Together, our data demonstrate a central role for TRM cells in the pathogenesis of chronic intestinal inflammation and suggest that these cells could be targets for future therapeutic approaches in IBD.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colitis/immunology , Immunologic Memory/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , Transcription Factors/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chronic Disease , Colitis/genetics , Colitis/metabolism , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Humans , Immunologic Memory/genetics , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1/deficiency , Positive Regulatory Domain I-Binding Factor 1/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
16.
Nat Immunol ; 20(5): 581-592, 2019 05.
Article in English | MEDLINE | ID: mdl-30962591

ABSTRACT

Succinate is a signaling metabolite sensed extracellularly by succinate receptor 1 (SUNCR1). The accumulation of succinate in macrophages is known to activate a pro-inflammatory program; however, the contribution of SUCNR1 to macrophage phenotype and function has remained unclear. Here we found that activation of SUCNR1 had a critical role in the anti-inflammatory responses in macrophages. Myeloid-specific deficiency in SUCNR1 promoted a local pro-inflammatory phenotype, disrupted glucose homeostasis in mice fed a normal chow diet, exacerbated the metabolic consequences of diet-induced obesity and impaired adipose-tissue browning in response to cold exposure. Activation of SUCNR1 promoted an anti-inflammatory phenotype in macrophages and boosted the response of these cells to type 2 cytokines, including interleukin-4. Succinate decreased the expression of inflammatory markers in adipose tissue from lean human subjects but not that from obese subjects, who had lower expression of SUCNR1 in adipose-tissue-resident macrophages. Our findings highlight the importance of succinate-SUCNR1 signaling in determining macrophage polarization and assign a role to succinate in limiting inflammation.


Subject(s)
Inflammation/immunology , Macrophages/immunology , Obesity/immunology , Receptors, G-Protein-Coupled/immunology , Adipose Tissue/drug effects , Adipose Tissue/immunology , Adipose Tissue/metabolism , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Gene Expression Profiling/methods , Humans , Inflammation/genetics , Inflammation/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Obesity/genetics , Obesity/metabolism , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Succinic Acid/immunology , Succinic Acid/metabolism , Succinic Acid/pharmacology , THP-1 Cells
17.
Nat Immunol ; 20(5): 559-570, 2019 05.
Article in English | MEDLINE | ID: mdl-30996332

ABSTRACT

The C-type lectin receptor-Syk (spleen tyrosine kinase) adaptor CARD9 facilitates protective antifungal immunity within the central nervous system (CNS), as human deficiency in CARD9 causes susceptibility to fungus-specific, CNS-targeted infection. CARD9 promotes the recruitment of neutrophils to the fungus-infected CNS, which mediates fungal clearance. In the present study we investigated host and pathogen factors that promote protective neutrophil recruitment during invasion of the CNS by Candida albicans. The cytokine IL-1ß served an essential function in CNS antifungal immunity by driving production of the chemokine CXCL1, which recruited neutrophils expressing the chemokine receptor CXCR2. Neutrophil-recruiting production of IL-1ß and CXCL1 was induced in microglia by the fungus-secreted toxin Candidalysin, in a manner dependent on the kinase p38 and the transcription factor c-Fos. Notably, microglia relied on CARD9 for production of IL-1ß, via both transcriptional regulation of Il1b and inflammasome activation, and of CXCL1 in the fungus-infected CNS. Microglia-specific Card9 deletion impaired the production of IL-1ß and CXCL1 and neutrophil recruitment, and increased fungal proliferation in the CNS. Thus, an intricate network of host-pathogen interactions promotes antifungal immunity in the CNS; this is impaired in human deficiency in CARD9, which leads to fungal disease of the CNS.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , Candidiasis/immunology , Chemokine CXCL1/immunology , Interleukin-1beta/immunology , Microglia/immunology , Neutrophils/immunology , Animals , Brain/immunology , Brain/metabolism , Brain/microbiology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Candida albicans/immunology , Candida albicans/physiology , Candidiasis/genetics , Candidiasis/microbiology , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Host-Pathogen Interactions/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice, Knockout , Mice, Transgenic , Microglia/metabolism , Microglia/microbiology , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/metabolism , Neutrophils/microbiology
18.
Nat Immunol ; 20(5): 593-601, 2019 05.
Article in English | MEDLINE | ID: mdl-30886417

ABSTRACT

Interferon-λ (IFN-λ) acts on mucosal epithelial cells and thereby confers direct antiviral protection. In contrast, the role of IFN-λ in adaptive immunity is far less clear. Here, we report that mice deficient in IFN-λ signaling exhibited impaired CD8+ T cell and antibody responses after infection with a live-attenuated influenza virus. Virus-induced release of IFN-λ triggered the synthesis of thymic stromal lymphopoietin (TSLP) by M cells in the upper airways that, in turn, stimulated migratory dendritic cells and boosted antigen-dependent germinal center reactions in draining lymph nodes. The IFN-λ-TSLP axis also boosted production of the immunoglobulins IgG1 and IgA after intranasal immunization with influenza virus subunit vaccines and improved survival of mice after challenge with virulent influenza viruses. IFN-λ did not influence the efficacy of vaccines applied by subcutaneous or intraperitoneal routes, indicating that IFN-λ plays a vital role in potentiating adaptive immune responses that initiate at mucosal surfaces.


Subject(s)
Adaptive Immunity/immunology , Cytokines/immunology , Immunity, Mucosal/immunology , Interleukins/immunology , Adaptive Immunity/drug effects , Adaptive Immunity/genetics , Animals , Antibody Formation/drug effects , Antibody Formation/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/virology , Immunity, Mucosal/drug effects , Immunity, Mucosal/genetics , Immunization/methods , Influenza A virus/drug effects , Influenza A virus/immunology , Influenza A virus/physiology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Interleukins/administration & dosage , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Receptors, Interferon/metabolism , Thymic Stromal Lymphopoietin
19.
Cell ; 167(4): 1099-1110.e14, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27814507

ABSTRACT

As part of the Human Functional Genomics Project, which aims to understand the factors that determine the variability of immune responses, we investigated genetic variants affecting cytokine production in response to ex vivo stimulation in two independent cohorts of 500 and 200 healthy individuals. We demonstrate a strong impact of genetic heritability on cytokine production capacity after challenge with bacterial, fungal, viral, and non-microbial stimuli. In addition to 17 novel genome-wide significant cytokine QTLs (cQTLs), our study provides a comprehensive picture of the genetic variants that influence six different cytokines in whole blood, blood mononuclear cells, and macrophages. Important biological pathways that contain cytokine QTLs map to pattern recognition receptors (TLR1-6-10 cluster), cytokine and complement inhibitors, and the kallikrein system. The cytokine QTLs show enrichment for monocyte-specific enhancers, are more often located in regions under positive selection, and are significantly enriched among SNPs associated with infections and immune-mediated diseases. PAPERCLIP.


Subject(s)
Cytokines/genetics , Cytokines/immunology , Infections/immunology , Adolescent , Adult , Aged , Blood/immunology , Female , Genome-Wide Association Study , Human Genome Project , Humans , Infections/microbiology , Infections/virology , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Male , Middle Aged , Polymorphism, Single Nucleotide , Quantitative Trait Loci
20.
Cell ; 165(4): 896-909, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27153497

ABSTRACT

The circadian clock controls metabolic and physiological processes through finely tuned molecular mechanisms. The clock is remarkably plastic and adapts to exogenous "zeitgebers," such as light and nutrition. How a pathological condition in a given tissue influences systemic circadian homeostasis in other tissues remains an unanswered question of conceptual and biomedical importance. Here, we show that lung adenocarcinoma operates as an endogenous reorganizer of circadian metabolism. High-throughput transcriptomics and metabolomics revealed unique signatures of transcripts and metabolites cycling exclusively in livers of tumor-bearing mice. Remarkably, lung cancer has no effect on the core clock but rather reprograms hepatic metabolism through altered pro-inflammatory response via the STAT3-Socs3 pathway. This results in disruption of AKT, AMPK, and SREBP signaling, leading to altered insulin, glucose, and lipid metabolism. Thus, lung adenocarcinoma functions as a potent endogenous circadian organizer (ECO), which rewires the pathophysiological dimension of a distal tissue such as the liver. PAPERCLIP.


Subject(s)
Adenocarcinoma/physiopathology , Circadian Clocks , Liver/physiopathology , Lung Neoplasms/physiopathology , Adenocarcinoma of Lung , Animals , Cytokines/genetics , Glucose/metabolism , Homeostasis , Insulin/metabolism , Mice , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL