Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4350, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782954

ABSTRACT

mRNA lipid nanoparticle (LNP) vaccines would be useful during an influenza virus pandemic since they can be produced rapidly and do not require the generation of egg-adapted vaccine seed stocks. Highly pathogenic avian influenza viruses from H5 clade 2.3.4.4b are circulating at unprecedently high levels in wild and domestic birds and have the potential to adapt to humans. Here, we generate an mRNA lipid nanoparticle (LNP) vaccine encoding the hemagglutinin (HA) glycoprotein from a clade 2.3.4.4b H5 isolate. The H5 mRNA-LNP vaccine elicits strong T cell and antibody responses in female mice, including neutralizing antibodies and broadly-reactive anti-HA stalk antibodies. The H5 mRNA-LNP vaccine elicits antibodies at similar levels compared to whole inactivated vaccines in female mice with and without prior H1N1 exposures. Finally, we find that the H5 mRNA-LNP vaccine is immunogenic in male ferrets and prevents morbidity and mortality of animals following 2.3.4.4b H5N1 challenge. Together, our data demonstrate that a monovalent mRNA-LNP vaccine expressing 2.3.4.4b H5 is immunogenic and protective in pre-clinical animal models.


Subject(s)
Antibodies, Viral , Ferrets , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H5N1 Subtype , Influenza Vaccines , Nanoparticles , Orthomyxoviridae Infections , mRNA Vaccines , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Female , Mice , Nanoparticles/chemistry , Male , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/genetics , Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , mRNA Vaccines/immunology , Antibodies, Neutralizing/immunology , Mice, Inbred BALB C , Influenza in Birds/prevention & control , Influenza in Birds/immunology , Influenza in Birds/virology , Humans , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/genetics , Birds/virology , Lipids/chemistry , Liposomes
2.
mBio ; 15(3): e0008324, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38376210

ABSTRACT

Mice that lack the genes for IL-27, or the IL-27 receptor, and infected with Toxoplasma gondii develop T cell-mediated pathology. Here, studies were performed to determine the impact of endogenous IL-27 on the immune response to T. gondii in wild-type (WT) mice. Analysis of infected mice revealed the early production of IL-27p28 by a subset of Ly6Chi, inflammatory monocytes, and sustained IL-27p28 production at sites of acute and chronic infection. Administration of anti-IL-27p28 prior to infection resulted in an early (day 5) increase in levels of macrophage and granulocyte activation, as well as enhanced effector T cell responses, as measured by both cellularity, cytokine production, and transcriptional profiling. This enhanced acute response led to immune pathology, while blockade during the chronic phase of infection resulted in enhanced T cell responses but no systemic pathology. In the absence of IL-27, the enhanced monocyte responses observed at day 10 were a secondary consequence of activated CD4+ T cells. Thus, in WT mice, IL-27 has distinct suppressive effects that impact innate and adaptive immunity during different phases of this infection. IMPORTANCE: The molecule IL-27 is critical in limiting the immune response to the parasite Toxoplasma gondii. In the absence of IL-27, a lethal, overactive immune response develops during infection. However, when exactly in the course of infection this molecule is needed was unclear. By selectively inhibiting IL-27 during this parasitic infection, we discovered that IL-27 was only needed during, but not prior to, infection. Additionally, IL-27 is only needed in the active areas in which the parasite is replicating. Finally, our work found that a previously unstudied cell type, monocytes, was regulated by IL-27, which contributes further to our understanding of the regulatory networks established by this molecule.


Subject(s)
Interleukin-27 , Toxoplasma , Toxoplasmosis , Animals , Mice , Interleukin-27/metabolism , Mice, Inbred C57BL , Monocytes , T-Lymphocytes , Toxoplasmosis/parasitology
3.
bioRxiv ; 2023 Apr 30.
Article in English | MEDLINE | ID: mdl-37162920

ABSTRACT

Highly pathogenic avian influenza viruses from H5 clade 2.3.4.4b are circulating at unprecedently high levels in wild and domestic birds and have the potential to adapt to humans. We generated an mRNA lipid nanoparticle (LNP) vaccine encoding the hemagglutinin (HA) glycoprotein from a clade 2.3.4.4b H5 isolate. We show that the vaccine is immunogenic in mice and ferrets and prevents morbidity and mortality of ferrets following 2.3.4.4b H5N1 challenge.

4.
J Exp Med ; 220(2)2023 02 06.
Article in English | MEDLINE | ID: mdl-36445307

ABSTRACT

The T-box transcription factor T-bet is regarded as a "master regulator" of CD4+ Th1 differentiation and IFN-γ production. However, in multiple models of infection, T-bet appears less critical for CD8+ T cell expansion and effector function. Here, we show that following vaccination with a replication-deficient strain of Toxoplasma gondii, CD8+ T cell expression of T-bet is required for optimal expansion of parasite-specific effector CD8+ T cells. Analysis of the early events associated with T cell activation reveals that the α chain of LFA1, CD11a, is a target of T-bet, and T-bet is necessary for CD8+ T cell upregulation of this integrin, which influences the initial priming of CD8+ effector T cells. We propose that the early expression of T-bet represents a T cell-intrinsic factor that optimizes T-DC interactions necessary to generate effector responses.


Subject(s)
Lymphocyte Activation , Memory T Cells , Up-Regulation , Transcriptional Activation , CD8-Positive T-Lymphocytes
5.
Sci Immunol ; 7(75): eabq7432, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36179012

ABSTRACT

In the peritoneal cavity, the omentum contains fat-associated lymphoid clusters (FALCs) whose role in response to infection is poorly understood. After intraperitoneal immunization with Toxoplasma gondii, conventional type 1 dendritic cells (cDC1s) were critical to induce innate sources of IFN-γ and cellular changes in the FALCs. Unexpectedly, infected peritoneal macrophages that migrated into the FALCs primed CD8+ T cells. Although T cell priming was cDC1 independent, these DCs were required for maximal CD8+ T cell expansion. An agent-based computational model and experimental data highlighted that cDC1s affected the magnitude of the proliferative burst and promoted CD8+ T cell expression of nutrient uptake receptors and cell survival. Thus, although FALCs lack the organization of secondary lymphoid organs, cDC1s resident in this tissue coordinate innate responses to microbial challenge and provide secondary signals required for T cell expansion and memory formation.


Subject(s)
CD8-Positive T-Lymphocytes , Omentum , Dendritic Cells
6.
PLoS Pathog ; 18(6): e1010296, 2022 06.
Article in English | MEDLINE | ID: mdl-35727849

ABSTRACT

Initial TCR engagement (priming) of naive CD8+ T cells results in T cell expansion, and these early events influence the generation of diverse effector and memory populations. During infection, activated T cells can re-encounter cognate antigen, but how these events influence local effector responses or formation of memory populations is unclear. To address this issue, OT-I T cells which express the Nur77-GFP reporter of TCR activation were paired with the parasite Toxoplasma gondii that expresses OVA to assess how secondary encounter with antigen influences CD8+ T cell responses. During acute infection, TCR stimulation in affected tissues correlated with parasite burden and was associated with markers of effector cells while Nur77-GFP- OT-I showed signs of effector memory potential. However, both Nur77-GFP- and Nur77-GFP+ OT-I from acutely infected mice formed similar memory populations when transferred into naive mice. During the chronic stage of infection in the CNS, TCR activation was associated with large scale transcriptional changes and the acquisition of an effector T cell phenotype as well as the generation of a population of CD103+ CD69+ Trm like cells. While inhibition of parasite replication resulted in reduced effector responses it did not alter the Trm population. These data sets highlight that recent TCR activation contributes to the phenotypic heterogeneity of the CD8+ T cell response but suggest that this process has a limited impact on memory populations at acute and chronic stages of infection.


Subject(s)
Toxoplasma , Toxoplasmosis , Animals , CD8-Positive T-Lymphocytes , Immunologic Memory , Mice , Receptors, Antigen, T-Cell
7.
Immunohorizons ; 5(6): 384-394, 2021 06 04.
Article in English | MEDLINE | ID: mdl-34088852

ABSTRACT

Resistance to the parasite Toxoplasma gondii is mediated by NK and T cell production of IFN-γ, but the failure to contract this response can lead to severe T cell-dependent immunopathology. Although the cytokines IL-10 and IL-27 prevent immune hyperactivity during toxoplasmosis, inhibitory receptors, expressed by NK and T cells, are also implicated in this process. The inhibitory receptor TIGIT is expressed on NK and T cells and competes with the costimulatory receptor CD226 for binding of the ligand CD155. During toxoplasmosis, the activation of NK and T cells is associated with increased expression of CD226 and TIGIT, whereas DCs express increased levels of CD155. To determine if the loss of TIGIT impacts NK and T cell activities, wild-type and TIGIT knockout mice were infected with T. gondii During the acute stage of infection, wild-type and TIGIT knockout mice had comparable parasite burdens and similar NK and T cell responses. Likewise, during the chronic phase of this infection, the loss of TIGIT did not affect the magnitude or phenotype of the T cell response nor the ability to control pathogen load. These data suggest that during toxoplasmosis, despite upregulation of relevant ligands, TIGIT signaling does not limit NK and T cell activities. Thus, TIGIT-independent mechanisms dominate the restraint of the immune response during toxoplasmosis.


Subject(s)
Killer Cells, Natural/immunology , Receptors, Immunologic/metabolism , T-Lymphocytes/immunology , Toxoplasmosis/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Killer Cells, Natural/metabolism , Male , Mice , Mice, Knockout , Parasite Load , Receptors, Immunologic/genetics , Receptors, Virus/metabolism , T-Lymphocytes/metabolism , Toxoplasma/immunology , Toxoplasma/isolation & purification , Toxoplasmosis/blood , Toxoplasmosis/parasitology
8.
Immunity ; 54(6): 1200-1218.e9, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33951416

ABSTRACT

Tissue macrophages self-renew during homeostasis and produce inflammatory mediators upon microbial infection. We examined the relationship between proliferative and inflammatory properties of tissue macrophages by defining the impact of the Wnt/ß-catenin pathway, a central regulator of self-renewal, in alveolar macrophages (AMs). Activation of ß-catenin by Wnt ligand inhibited AM proliferation and stemness, but promoted inflammatory activity. In a murine influenza viral pneumonia model, ß-catenin-mediated AM inflammatory activity promoted acute host morbidity; in contrast, AM proliferation enabled repopulation of reparative AMs and tissue recovery following viral clearance. Mechanistically, Wnt treatment promoted ß-catenin-HIF-1α interaction and glycolysis-dependent inflammation while suppressing mitochondrial metabolism and thereby, AM proliferation. Differential HIF-1α activities distinguished proliferative and inflammatory AMs in vivo. This ß-catenin-HIF-1α axis was conserved in human AMs and enhanced HIF-1α expression associated with macrophage inflammation in COVID-19 patients. Thus, inflammatory and reparative activities of lung macrophages are regulated by ß-catenin-HIF-1α signaling, with implications for the treatment of severe respiratory diseases.


Subject(s)
COVID-19/immunology , COVID-19/virology , Cell Self Renewal/immunology , Host-Pathogen Interactions/immunology , Macrophages/immunology , SARS-CoV-2/immunology , Biomarkers , COVID-19/metabolism , Cytokines/metabolism , Disease Susceptibility/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation Mediators/metabolism , Macrophages/cytology , Macrophages/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Signal Transduction
9.
J Clin Invest ; 131(7)2021 04 01.
Article in English | MEDLINE | ID: mdl-33792560

ABSTRACT

Adoptive T cell therapies (ACTs) hold great promise in cancer treatment, but low overall response rates in patients with solid tumors underscore remaining challenges in realizing the potential of this cellular immunotherapy approach. Promoting CD8+ T cell adaptation to tissue residency represents an underutilized but promising strategy to improve tumor-infiltrating lymphocyte (TIL) function. Here, we report that deletion of the HIF negative regulator von Hippel-Lindau (VHL) in CD8+ T cells induced HIF-1α/HIF-2α-dependent differentiation of tissue-resident memory-like (Trm-like) TILs in mouse models of malignancy. VHL-deficient TILs accumulated in tumors and exhibited a core Trm signature despite an exhaustion-associated phenotype, which led to retained polyfunctionality and response to αPD-1 immunotherapy, resulting in tumor eradication and protective tissue-resident memory. VHL deficiency similarly facilitated enhanced accumulation of chimeric antigen receptor (CAR) T cells with a Trm-like phenotype in tumors. Thus, HIF activity in CD8+ TILs promotes accumulation and antitumor activity, providing a new strategy to enhance the efficacy of ACTs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , CD8-Positive T-Lymphocytes/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Immunity, Cellular , Immunologic Memory , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Experimental/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/immunology
10.
J Exp Med ; 217(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-31961916

ABSTRACT

The ability of Toxoplasma gondii to inject the rhoptry kinase ROP16 into host cells results in the activation of the transcription factors STAT3 and STAT6, but it is unclear how these events impact infection. Here, parasites that inject Cre-recombinase with rhoptry proteins were used to distinguish infected macrophages from those only injected with parasite proteins. Transcriptional profiling revealed that injection of rhoptry proteins alone was sufficient to induce an M2 phenotype that is dependent on STAT3 and STAT6, but only infected cells displayed reduced expression of genes associated with antimicrobial activity and protective immunity. In vivo, the absence of STAT3 or STAT6 improved parasite control, while the loss of ROP16 resulted in a marked reduction in parasite numbers and heightened parasite-specific T cell responses. Thus, ROP16 is a virulence factor that can act in cis and trans to promote M2 programs and which limits the magnitude of parasite-specific T cell responses.


Subject(s)
Protein-Tyrosine Kinases/immunology , Protozoan Proteins/immunology , T-Lymphocytes/immunology , Toxoplasma/immunology , Virulence Factors/immunology , Animals , Macrophages/immunology , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/immunology , STAT6 Transcription Factor/immunology
11.
Immunohorizons ; 3(1): 13-25, 2019 01 15.
Article in English | MEDLINE | ID: mdl-31356173

ABSTRACT

Inhibitory receptors (IR) are a diverse group of cell surface molecules that modulate T cell activation, but there are gaps in our knowledge of the cell-extrinsic factors that regulate their expression. The present study found that in vivo overexpression of IL-27 in mice led to increased T cell expression of PD-L1, LAG-3, TIGIT, and TIM-3. In vitro, TCR stimulation alone promoted expression of multiple IRs, whereas IL-27 alone induced expression of PD-L1. However, the combination of intermediate TCR stimulation and IL-27 resulted in synergistic induction of LAG-3, CTLA-4, and TIGIT. In vivo, infection with Toxoplasma gondii resulted in parasite-specific effector T cells that expressed high levels of IR, and at local sites of infection where IL-27 production was highest, IL-27 was required for maximal effector cell expression of PD-L1, LAG-3, CTLA-4, and TIGIT. Together, these results affirm the critical role of TCR signals in the induction of IR expression but find that during infection, IL-27 promotes T cell expression of IR.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Costimulatory and Inhibitory T-Cell Receptors/metabolism , Interleukins/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/parasitology , CTLA-4 Antigen/metabolism , Costimulatory and Inhibitory T-Cell Receptors/genetics , Female , Interleukins/genetics , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/genetics , Receptors, Immunologic/metabolism , Spleen/pathology , Toxoplasma , Toxoplasmosis/immunology , Transcriptome , Transfection
13.
Nature ; 552(7684): 253-257, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29211713

ABSTRACT

Tissue-resident memory CD8+ T (TRM) cells are found at common sites of pathogen exposure, where they elicit rapid and robust protective immune responses. However, the molecular signals that control TRM cell differentiation and homeostasis are not fully understood. Here we show that mouse TRM precursor cells represent a unique CD8+ T cell subset that is distinct from the precursors of circulating memory cell populations at the levels of gene expression and chromatin accessibility. Using computational and pooled in vivo RNA interference screens, we identify the transcription factor Runx3 as a key regulator of TRM cell differentiation and homeostasis. Runx3 was required to establish TRM cell populations in diverse tissue environments, and supported the expression of crucial tissue-residency genes while suppressing genes associated with tissue egress and recirculation. Furthermore, we show that human and mouse tumour-infiltrating lymphocytes share a core tissue-residency gene-expression signature with TRM cells that is associated with Runx3 activity. In a mouse model of adoptive T cell therapy for melanoma, Runx3-deficient CD8+ tumour-infiltrating lymphocytes failed to accumulate in tumours, resulting in greater rates of tumour growth and mortality. Conversely, overexpression of Runx3 enhanced tumour-specific CD8+ T cell abundance, delayed tumour growth, and prolonged survival. In addition to establishing Runx3 as a central regulator of TRM cell differentiation, these results provide insight into the signals that promote T cell residency in non-lymphoid sites, which could be used to enhance vaccine efficacy or adoptive cell therapy treatments that target cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Core Binding Factor Alpha 3 Subunit/metabolism , Immunologic Memory , Melanoma/immunology , Organ Specificity/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Proliferation , Chromatin/genetics , Chromatin/metabolism , Core Binding Factor Alpha 3 Subunit/deficiency , Core Binding Factor Alpha 3 Subunit/genetics , Disease Models, Animal , Female , Gene Expression Regulation , Homeostasis , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy , Mice , Organ Specificity/genetics , Survival Analysis , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
14.
Cancer Cell ; 32(5): 669-683.e5, 2017 11 13.
Article in English | MEDLINE | ID: mdl-29136509

ABSTRACT

Cytotoxic T cells infiltrating tumors are thought to utilize HIF transcription factors during adaptation to the hypoxic tumor microenvironment. Deletion analyses of the two key HIF isoforms found that HIF-1α, but not HIF-2α, was essential for the effector state in CD8+ T cells. Furthermore, loss of HIF-1α in CD8+ T cells reduced tumor infiltration and tumor cell killing, and altered tumor vascularization. Deletion of VEGF-A, an HIF target gene, in CD8+ T cells accelerated tumorigenesis while also altering vascularization. Analyses of human breast cancer showed inverse correlations between VEGF-A expression and CD8+ T cell infiltration, and a link between T cell infiltration and vascularization. These data demonstrate that the HIF-1α/VEGF-A axis is an essential aspect of tumor immunity.


Subject(s)
Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neoplasms, Experimental/genetics , T-Lymphocytes, Cytotoxic/metabolism , Vascular Endothelial Growth Factor A/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cells, Cultured , Disease Progression , Female , Gene Expression Profiling/methods , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism
15.
J Clin Invest ; 127(10): 3609-3623, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28846070

ABSTRACT

During an immune response, CD8+ T lymphocytes can undergo asymmetric division, giving rise to daughter cells that exhibit distinct tendencies to adopt terminal effector and memory cell fates. Here we show that "pre-effector" and "pre-memory" cells resulting from the first CD8+ T cell division in vivo exhibited low and high rates of endogenous proteasome activity, respectively. Pharmacologic reduction of proteasome activity in CD8+ T cells early during differentiation resulted in acquisition of terminal effector cell characteristics, whereas enhancement of proteasome activity conferred attributes of memory lymphocytes. Transcriptomic and proteomic analyses revealed that modulating proteasome activity in CD8+ T cells affected cellular metabolism. These metabolic changes were mediated, in part, through differential expression of Myc, a transcription factor that controls glycolysis and metabolic reprogramming. Taken together, these results demonstrate that proteasome activity is an important regulator of CD8+ T cell fate and raise the possibility that increasing proteasome activity may be a useful therapeutic strategy to enhance the generation of memory lymphocytes.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Division/immunology , Glycolysis/immunology , Immunologic Memory , Proteasome Endopeptidase Complex/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Mice , Mice, Mutant Strains , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-myc/immunology , Proto-Oncogene Proteins c-myc/metabolism
16.
Immunity ; 46(5): 714-729, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514673

ABSTRACT

Recognition of pathogens by innate and adaptive immune cells instructs rapid alterations of cellular processes to promote effective resolution of infection. To accommodate increased bioenergetic and biosynthetic demands, metabolic pathways are harnessed to maximize proliferation and effector molecule production. In parallel, activation initiates context-specific gene-expression programs that drive effector functions and cell fates that correlate with changes in epigenetic landscapes. Many chromatin- and DNA-modifying enzymes make use of substrates and cofactors that are intermediates of metabolic pathways, providing potential cross talk between metabolism and epigenetic regulation of gene expression. In this review, we discuss recent studies of T cells and macrophages supporting a role for metabolic activity in integrating environmental signals with activation-induced gene-expression programs through modulation of the epigenome and speculate as to how this may influence context-specific macrophage and T cell responses to infection.


Subject(s)
Energy Metabolism , Epigenesis, Genetic , Gene Expression Regulation , Immunity , Macrophages/immunology , Macrophages/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cellular Microenvironment/genetics , Cellular Microenvironment/immunology , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/cytology , Metabolic Networks and Pathways , T-Lymphocytes/cytology
17.
Nature ; 540(7632): 236-241, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27798602

ABSTRACT

R-2-hydroxyglutarate accumulates to millimolar levels in cancer cells with gain-of-function isocitrate dehydrogenase 1/2 mutations. These levels of R-2-hydroxyglutarate affect 2-oxoglutarate-dependent dioxygenases. Both metabolite enantiomers, R- and S-2-hydroxyglutarate, are detectible in healthy individuals, yet their physiological function remains elusive. Here we show that 2-hydroxyglutarate accumulates in mouse CD8+ T cells in response to T-cell receptor triggering, and accumulates to millimolar levels in physiological oxygen conditions through a hypoxia-inducible factor 1-alpha (HIF-1α)-dependent mechanism. S-2-hydroxyglutarate predominates over R-2-hydroxyglutarate in activated T cells, and we demonstrate alterations in markers of CD8+ T-cell differentiation in response to this metabolite. Modulation of histone and DNA demethylation, as well as HIF-1α stability, mediate these effects. S-2-hydroxyglutarate treatment greatly enhances the in vivo proliferation, persistence and anti-tumour capacity of adoptively transferred CD8+ T cells. Thus, S-2-hydroxyglutarate acts as an immunometabolite that links environmental context, through a metabolic-epigenetic axis, to immune fate and function.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Cell Differentiation/drug effects , Glutarates/pharmacology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , DNA/chemistry , DNA/metabolism , DNA Methylation/drug effects , Dioxygenases/metabolism , Glutarates/immunology , Glutarates/metabolism , Histones/metabolism , Homeostasis/drug effects , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ketoglutaric Acids/metabolism , Lymphocyte Activation , Lysine/metabolism , Mice , Oxygen/metabolism , Protein Stability , Receptors, Antigen, T-Cell/immunology , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
18.
Immunity ; 45(5): 1024-1037, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27836431

ABSTRACT

Extensive metabolic changes accompany T cell activation, including a switch to glycolytic energy production and increased biosynthesis. Recent studies suggest that subsequent return to reliance on oxidative phosphorylation and increasing spare respiratory capacity are essential for the differentiation of memory CD8+ T cells. In contrast, we found that constitutive glycolytic metabolism and suppression of oxidative phosphorylation in CD8+ T cells, achieved by conditional deletion of hypoxia-inducible factor regulator Vhl, accelerated CD8+ memory cell differentiation during viral infection. Despite sustained glycolysis, CD8+ memory cells emerged that upregulated key memory-associated cytokine receptors and transcription factors and showed a heightened response to secondary challenge. In addition, increased glycolysis not only permitted memory formation, but it also favored the formation of long-lived effector-memory CD8+ T cells. These data redefine the role of cellular metabolism in memory cell differentiation, showing that reliance on glycolytic metabolism does not hinder formation of a protective memory population.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Glycolysis/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Animals , Arenaviridae Infections/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cell Separation , Disease Models, Animal , Flow Cytometry , Lymphocytic choriomeningitis virus , Mice , Mice, Transgenic , Oxidative Phosphorylation
19.
Cell ; 166(5): 1117-1131.e14, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27565342

ABSTRACT

Cancer cells must evade immune responses at distant sites to establish metastases. The lung is a frequent site for metastasis. We hypothesized that lung-specific immunoregulatory mechanisms create an immunologically permissive environment for tumor colonization. We found that T-cell-intrinsic expression of the oxygen-sensing prolyl-hydroxylase (PHD) proteins is required to maintain local tolerance against innocuous antigens in the lung but powerfully licenses colonization by circulating tumor cells. PHD proteins limit pulmonary type helper (Th)-1 responses, promote CD4(+)-regulatory T (Treg) cell induction, and restrain CD8(+) T cell effector function. Tumor colonization is accompanied by PHD-protein-dependent induction of pulmonary Treg cells and suppression of IFN-γ-dependent tumor clearance. T-cell-intrinsic deletion or pharmacological inhibition of PHD proteins limits tumor colonization of the lung and improves the efficacy of adoptive cell transfer immunotherapy. Collectively, PHD proteins function in T cells to coordinate distinct immunoregulatory programs within the lung that are permissive to cancer metastasis. PAPERCLIP.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung/immunology , Oxygen/metabolism , Prolyl Hydroxylases/metabolism , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/enzymology , Glycolysis/immunology , Interferon-gamma/immunology , Lung/pathology , Lung Neoplasms/therapy , Lymphocyte Activation , Mice , Mice, Knockout , Neoplasm Metastasis , Neuropilin-1/metabolism , Prolyl Hydroxylases/genetics , T-Lymphocytes, Regulatory/enzymology , Th1 Cells/enzymology , Th1 Cells/immunology
20.
Mol Immunol ; 68(2 Pt C): 527-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26298577

ABSTRACT

Resolution of infection requires the coordinated response of heterogeneous cell types to a range of physiological and pathological signals to regulate their proliferation, migration, differentiation, and effector functions. One mechanism by which immune cells integrate these signals is through modulating metabolic activity. A well-studied regulator of cellular metabolism is the hypoxia-inducible factor (HIF) family, the highly conserved central regulators of adaptation to limiting oxygen tension. HIF's regulation of cellular metabolism and a variety of effector, signaling, and trafficking molecules has made these transcription factors a recent topic of interest in T cell biology. Low oxygen availability, or hypoxia, increases expression and stabilization of HIF in immune cells, activating molecular programs both unique and common among cell types, including glycolytic metabolism. Notably, numerous oxygen-independent signals, many of which are active in T cells, also result in enhanced HIF activity. Here, we discuss both oxygen-dependent and -independent regulation of HIF activity in T cells and the resulting impacts on metabolism, differentiation, function, and immunity.


Subject(s)
Cell Hypoxia/immunology , Hypoxia-Inducible Factor 1/immunology , T-Lymphocytes/metabolism , Animals , Cell Differentiation/immunology , Humans , Hypoxia-Inducible Factor 1/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...