Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Immunol ; 24(3): 516-530, 2023 03.
Article in English | MEDLINE | ID: mdl-36732424

ABSTRACT

How lipidome changes support CD8+ effector T (Teff) cell differentiation is not well understood. Here we show that, although naive T cells are rich in polyunsaturated phosphoinositides (PIPn with 3-4 double bonds), Teff cells have unique PIPn marked by saturated fatty acyl chains (0-2 double bonds). PIPn are precursors for second messengers. Polyunsaturated phosphatidylinositol bisphosphate (PIP2) exclusively supported signaling immediately upon T cell antigen receptor activation. In late Teff cells, activity of phospholipase C-γ1, the enzyme that cleaves PIP2 into downstream mediators, waned, and saturated PIPn became essential for sustained signaling. Saturated PIP was more rapidly converted to PIP2 with subsequent recruitment of phospholipase C-γ1, and loss of saturated PIPn impaired Teff cell fitness and function, even in cells with abundant polyunsaturated PIPn. Glucose was the substrate for de novo PIPn synthesis, and was rapidly utilized for saturated PIP2 generation. Thus, separate PIPn pools with distinct acyl chain compositions and metabolic dependencies drive important signaling events to initiate and then sustain effector function during CD8+ T cell differentiation.


Subject(s)
Phosphatidylinositol Phosphates , Phosphatidylinositols , Phosphatidylinositols/metabolism , Signal Transduction , Type C Phospholipases/metabolism , CD8-Positive T-Lymphocytes/metabolism
2.
Cell ; 171(2): 385-397.e11, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28919076

ABSTRACT

T cell receptor (TCR) signaling without CD28 can elicit primary effector T cells, but memory T cells generated during this process are anergic, failing to respond to secondary antigen exposure. We show that, upon T cell activation, CD28 transiently promotes expression of carnitine palmitoyltransferase 1a (Cpt1a), an enzyme that facilitates mitochondrial fatty acid oxidation (FAO), before the first cell division, coinciding with mitochondrial elongation and enhanced spare respiratory capacity (SRC). microRNA-33 (miR33), a target of thioredoxin-interacting protein (TXNIP), attenuates Cpt1a expression in the absence of CD28, resulting in cells that thereafter are metabolically compromised during reactivation or periods of increased bioenergetic demand. Early CD28-dependent mitochondrial engagement is needed for T cells to remodel cristae, develop SRC, and rapidly produce cytokines upon restimulation-cardinal features of protective memory T cells. Our data show that initial CD28 signals during T cell activation prime mitochondria with latent metabolic capacity that is essential for future T cell responses.


Subject(s)
CD28 Antigens/metabolism , Lymphocyte Activation , Mitochondria/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Carnitine O-Palmitoyltransferase , Enzyme Inhibitors/pharmacology , Epoxy Compounds/pharmacology , Humans , Interleukin-15/immunology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism , Stress, Physiological , T-Lymphocytes/metabolism
3.
Cell ; 166(1): 63-76, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27293185

ABSTRACT

Activated effector T (TE) cells augment anabolic pathways of metabolism, such as aerobic glycolysis, while memory T (TM) cells engage catabolic pathways, like fatty acid oxidation (FAO). However, signals that drive these differences remain unclear. Mitochondria are metabolic organelles that actively transform their ultrastructure. Therefore, we questioned whether mitochondrial dynamics controls T cell metabolism. We show that TE cells have punctate mitochondria, while TM cells maintain fused networks. The fusion protein Opa1 is required for TM, but not TE cells after infection, and enforcing fusion in TE cells imposes TM cell characteristics and enhances antitumor function. Our data suggest that, by altering cristae morphology, fusion in TM cells configures electron transport chain (ETC) complex associations favoring oxidative phosphorylation (OXPHOS) and FAO, while fission in TE cells leads to cristae expansion, reducing ETC efficiency and promoting aerobic glycolysis. Thus, mitochondrial remodeling is a signaling mechanism that instructs T cell metabolic programming.


Subject(s)
Mitochondrial Dynamics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Cell Differentiation , Electron Transport , Fatty Acids/metabolism , GTP Phosphohydrolases/metabolism , Glycolysis , Humans , Immunologic Memory , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Signal Transduction , T-Lymphocytes/immunology
4.
Cell ; 162(6): 1229-41, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26321679

ABSTRACT

Failure of T cells to protect against cancer is thought to result from lack of antigen recognition, chronic activation, and/or suppression by other cells. Using a mouse sarcoma model, we show that glucose consumption by tumors metabolically restricts T cells, leading to their dampened mTOR activity, glycolytic capacity, and IFN-γ production, thereby allowing tumor progression. We show that enhancing glycolysis in an antigenic "regressor" tumor is sufficient to override the protective ability of T cells to control tumor growth. We also show that checkpoint blockade antibodies against CTLA-4, PD-1, and PD-L1, which are used clinically, restore glucose in tumor microenvironment, permitting T cell glycolysis and IFN-γ production. Furthermore, we found that blocking PD-L1 directly on tumors dampens glycolysis by inhibiting mTOR activity and decreasing expression of glycolysis enzymes, reflecting a role for PD-L1 in tumor glucose utilization. Our results establish that tumor-imposed metabolic restrictions can mediate T cell hyporesponsiveness during cancer.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Glycolysis , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/metabolism , Tumor Microenvironment , Animals , Antibodies, Monoclonal/administration & dosage , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Interferon-gamma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology
5.
Cell ; 153(6): 1239-51, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23746840

ABSTRACT

A "switch" from oxidative phosphorylation (OXPHOS) to aerobic glycolysis is a hallmark of T cell activation and is thought to be required to meet the metabolic demands of proliferation. However, why proliferating cells adopt this less efficient metabolism, especially in an oxygen-replete environment, remains incompletely understood. We show here that aerobic glycolysis is specifically required for effector function in T cells but that this pathway is not necessary for proliferation or survival. When activated T cells are provided with costimulation and growth factors but are blocked from engaging glycolysis, their ability to produce IFN-γ is markedly compromised. This defect is translational and is regulated by the binding of the glycolysis enzyme GAPDH to AU-rich elements within the 3' UTR of IFN-γ mRNA. GAPDH, by engaging/disengaging glycolysis and through fluctuations in its expression, controls effector cytokine production. Thus, aerobic glycolysis is a metabolically regulated signaling mechanism needed to control cellular function.


Subject(s)
Glycolysis , Lymphocyte Activation , Oxidative Phosphorylation , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , 3' Untranslated Regions , Animals , Cell Proliferation , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Interferon-gamma/genetics , Listeria monocytogenes , Listeriosis/immunology , Mice , Mice, Inbred C57BL , Protein Biosynthesis , T-Lymphocytes/immunology
6.
Immunity ; 49(6): 1021-1033.e6, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30566880

ABSTRACT

Metabolic engagement is intrinsic to immune cell function. Prostaglandin E2 (PGE2) has been shown to modulate macrophage activation, yet how PGE2 might affect metabolism is unclear. Here, we show that PGE2 caused mitochondrial membrane potential (Δψm) to dissipate in interleukin-4-activated (M(IL-4)) macrophages. Effects on Δψm were a consequence of PGE2-initiated transcriptional regulation of genes, particularly Got1, in the malate-aspartate shuttle (MAS). Reduced Δψm caused alterations in the expression of 126 voltage-regulated genes (VRGs), including those encoding resistin-like molecule α (RELMα), a key marker of M(IL-4) cells, and genes that regulate the cell cycle. The transcription factor ETS variant 1 (ETV1) played a role in the regulation of 38% of the VRGs. These results reveal ETV1 as a Δψm-sensitive transcription factor and Δψm as a mediator of mitochondrial-directed nuclear gene expression.


Subject(s)
Cell Nucleus/drug effects , Dinoprostone/pharmacology , Gene Expression Regulation/drug effects , Macrophages/drug effects , Membrane Potential, Mitochondrial/physiology , Animals , Cell Nucleus/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Interleukin-4/pharmacology , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophages/metabolism , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Nature ; 591(7850): 471-476, 2021 03.
Article in English | MEDLINE | ID: mdl-33627869

ABSTRACT

The behaviour of Dictyostelium discoideum depends on nutrients1. When sufficient food is present these amoebae exist in a unicellular state, but upon starvation they aggregate into a multicellular organism2,3. This biology makes D. discoideum an ideal model for investigating how fundamental metabolism commands cell differentiation and function. Here we show that reactive oxygen species-generated as a consequence of nutrient limitation-lead to the sequestration of cysteine in the antioxidant glutathione. This sequestration limits the use of the sulfur atom of cysteine in processes that contribute to mitochondrial metabolism and cellular proliferation, such as protein translation and the activity of enzymes that contain an iron-sulfur cluster. The regulated sequestration of sulfur maintains D. discoideum in a nonproliferating state that paves the way for multicellular development. This mechanism of signalling through reactive oxygen species highlights oxygen and sulfur as simple signalling molecules that dictate cell fate in an early eukaryote, with implications for responses to nutrient fluctuations in multicellular eukaryotes.


Subject(s)
Dictyostelium/cytology , Dictyostelium/metabolism , Food Deprivation/physiology , Nutrients/metabolism , Sulfur/metabolism , Amino Acids, Essential/metabolism , Amino Acids, Essential/pharmacology , Antioxidants/metabolism , Cell Aggregation/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Respiration/drug effects , Cysteine/chemistry , Cysteine/metabolism , Cysteine/pharmacology , Dictyostelium/drug effects , Glutathione/chemistry , Glutathione/metabolism , Glutathione/pharmacology , Iron-Sulfur Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
8.
Nat Immunol ; 15(9): 884-93, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25029552

ABSTRACT

Although the transcription factor c-Myc is essential for the establishment of a metabolically active and proliferative state in T cells after priming, its expression is transient. It remains unknown how T cell activation is maintained after c-Myc expression is downregulated. Here we identified AP4 as the transcription factor that was induced by c-Myc and sustained activation of antigen-specific CD8+ T cells. Despite normal priming, AP4-deficient CD8+ T cells failed to continue transcription of a broad range of c-Myc-dependent targets. Mice lacking AP4 specifically in CD8+ T cells showed enhanced susceptibility to infection with West Nile virus. Genome-wide analysis suggested that many activation-induced genes encoding molecules involved in metabolism were shared targets of c-Myc and AP4. Thus, AP4 maintains c-Myc-initiated cellular activation programs in CD8+ T cells to control microbial infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Proto-Oncogene Proteins c-myc/immunology , Transcription Factors/immunology , Animals , Mice , West Nile Fever/immunology
9.
Immunity ; 45(1): 60-73, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27396958

ABSTRACT

Durable antibody production after vaccination or infection is mediated by long-lived plasma cells (LLPCs). Pathways that specifically allow LLPCs to persist remain unknown. Through bioenergetic profiling, we found that human and mouse LLPCs could robustly engage pyruvate-dependent respiration, whereas their short-lived counterparts could not. LLPCs took up more glucose than did short-lived plasma cells (SLPCs) in vivo, and this glucose was essential for the generation of pyruvate. Glucose was primarily used to glycosylate antibodies, but glycolysis could be promoted by stimuli such as low ATP levels and the resultant pyruvate used for respiration by LLPCs. Deletion of Mpc2, which encodes an essential component of the mitochondrial pyruvate carrier, led to a progressive loss of LLPCs and of vaccine-specific antibodies in vivo. Thus, glucose uptake and mitochondrial pyruvate import prevent bioenergetic crises and allow LLPCs to persist. Immunizations that maximize these plasma cell metabolic properties might thus provide enduring antibody-mediated immunity.


Subject(s)
Antibody-Producing Cells/immunology , Glucose/metabolism , Mitochondria/metabolism , Plasma Cells/immunology , Pyruvic Acid/metabolism , Animals , Biological Transport, Active , Cell Respiration , Cells, Cultured , Glycosylation , Humans , Immunoglobulins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Proprotein Convertase 2/genetics , Proprotein Convertase 2/metabolism , Stress, Physiological/immunology
10.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Article in English | MEDLINE | ID: mdl-34161266

ABSTRACT

Fever can provide a survival advantage during infection. Metabolic processes are sensitive to environmental conditions, but the effect of fever on T cell metabolism is not well characterized. We show that in activated CD8+ T cells, exposure to febrile temperature (39 °C) augmented metabolic activity and T cell effector functions, despite having a limited effect on proliferation or activation marker expression. Transcriptional profiling revealed an up-regulation of mitochondrial pathways, which was consistent with increased mass and metabolism observed in T cells exposed to 39 °C. Through in vitro and in vivo models, we determined that mitochondrial translation is integral to the enhanced metabolic activity and function of CD8+ T cells exposed to febrile temperature. Transiently exposing donor lymphocytes to 39 °C prior to infusion in a myeloid leukemia mouse model conferred enhanced therapeutic efficacy, raising the possibility that exposure of T cells to febrile temperatures could have clinical potential.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Fever/immunology , Mitochondria/metabolism , Protein Biosynthesis , Animals , Antineoplastic Agents/metabolism , CD8-Positive T-Lymphocytes/ultrastructure , Cytokines/biosynthesis , Glucose/metabolism , Leukemia, Myeloid/immunology , Leukemia, Myeloid/pathology , Leukemia, Myeloid/prevention & control , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitochondria/ultrastructure , Models, Biological , Temperature
11.
Immunity ; 41(1): 75-88, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25001241

ABSTRACT

Generation of CD8(+) memory T cells requires metabolic reprogramming that is characterized by enhanced mitochondrial fatty-acid oxidation (FAO). However, where the fatty acids (FA) that fuel this process come from remains unclear. While CD8(+) memory T cells engage FAO to a greater extent, we found that they acquired substantially fewer long-chain FA from their external environment than CD8(+) effector T (Teff) cells. Rather than using extracellular FA directly, memory T cells used extracellular glucose to support FAO and oxidative phosphorylation (OXPHOS), suggesting that lipids must be synthesized to generate the substrates needed for FAO. We have demonstrated that memory T cells rely on cell intrinsic expression of the lysosomal hydrolase LAL (lysosomal acid lipase) to mobilize FA for FAO and memory T cell development. Our observations link LAL to metabolic reprogramming in lymphocytes and show that cell intrinsic lipolysis is deterministic for memory T cell fate.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Fatty Acids/metabolism , Immunologic Memory/immunology , Lipolysis/immunology , Sterol Esterase/metabolism , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/pharmacology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/genetics , Fatty Acids/biosynthesis , Glucose/metabolism , Interleukin-15/immunology , Interleukin-2/immunology , Lipolysis/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Oxidation-Reduction , Oxidative Phosphorylation , Oxygen/metabolism , Protein Kinases/genetics , RNA Interference , RNA, Small Interfering , Sterol Esterase/biosynthesis
13.
Immunity ; 36(1): 68-78, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22206904

ABSTRACT

CD8(+) T cells undergo major metabolic changes upon activation, but how metabolism influences the establishment of long-lived memory T cells after infection remains a key question. We have shown here that CD8(+) memory T cells, but not CD8(+) T effector (Teff) cells, possessed substantial mitochondrial spare respiratory capacity (SRC). SRC is the extra capacity available in cells to produce energy in response to increased stress or work and as such is associated with cellular survival. We found that interleukin-15 (IL-15), a cytokine critical for CD8(+) memory T cells, regulated SRC and oxidative metabolism by promoting mitochondrial biogenesis and expression of carnitine palmitoyl transferase (CPT1a), a metabolic enzyme that controls the rate-limiting step to mitochondrial fatty acid oxidation (FAO). These results show how cytokines control the bioenergetic stability of memory T cells after infection by regulating mitochondrial metabolism.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Mitochondria/metabolism , Animals , CD8-Positive T-Lymphocytes/enzymology , Carnitine O-Palmitoyltransferase/metabolism , Cell Respiration/physiology , Fatty Acids/metabolism , Flow Cytometry , Interleukin-15/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/enzymology , Models, Biological , Oxidation-Reduction , Real-Time Polymerase Chain Reaction
14.
Proc Natl Acad Sci U S A ; 110(35): 14336-41, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23940348

ABSTRACT

A characteristic of memory T (TM) cells is their ability to mount faster and stronger responses to reinfection than naïve T (TN) cells do in response to an initial infection. However, the mechanisms that allow this rapid recall are not completely understood. We found that CD8 TM cells have more mitochondrial mass than CD8 TN cells and, that upon activation, the resulting secondary effector T (TE) cells proliferate more quickly, produce more cytokines, and maintain greater ATP levels than primary effector T cells. We also found that after activation, TM cells increase oxidative phosphorylation and aerobic glycolysis and sustain this increase to a greater extent than TN cells, suggesting that greater mitochondrial mass in TM cells not only promotes oxidative capacity, but also glycolytic capacity. We show that mitochondrial ATP is essential for the rapid induction of glycolysis in response to activation and the initiation of proliferation of both TN and TM cells. We also found that fatty acid oxidation is needed for TM cells to rapidly respond upon restimulation. Finally, we show that dissociation of the glycolysis enzyme hexokinase from mitochondria impairs proliferation and blocks the rapid induction of glycolysis upon T-cell receptor stimulation in TM cells. Our results demonstrate that greater mitochondrial mass endows TM cells with a bioenergetic advantage that underlies their ability to rapidly recall in response to reinfection.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Energy Metabolism , Lymphocyte Activation , Mice , Mitochondrial Size
15.
J Immunol ; 189(5): 2151-8, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22826320

ABSTRACT

Dendritic cells (DCs) are potent inducers of T cell immunity, and autologous DC vaccination holds promise for the treatment of cancers and chronic infectious diseases. In practice, however, therapeutic vaccines of this type have had mixed success. In this article, we show that brief exposure to inhibitors of mechanistic target of rapamycin (mTOR) in DCs during the period that they are responding to TLR agonists makes them particularly potent activators of naive CD8+ T cells and able to enhance control of B16 melanoma in a therapeutic autologous vaccination model in the mouse. The improved performance of DCs in which mTOR has been inhibited is correlated with an extended life span after activation and prolonged, increased expression of costimulatory molecules. Therapeutic autologous vaccination with DCs treated with TLR agonists plus the mTOR inhibitor rapamycin results in improved generation of Ag-specific CD8+ T cells in vivo and improved antitumor immunity compared with that observed with DCs treated with TLR agonists alone. These findings define mTOR as a molecular target for augmenting DC survival and activation, and document a novel pharmacologic approach for enhancing the efficacy of therapeutic autologous DC vaccination.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/transplantation , Immunotherapy, Adoptive/methods , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/physiology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/therapeutic use , Animals , Cells, Cultured , Coculture Techniques , Dendritic Cells/metabolism , Humans , Lipopolysaccharides/physiology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Transgenic , Transplantation, Autologous , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
16.
Sci Immunol ; 7(70): eabl7482, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35427180

ABSTRACT

Macrophages populate every organ during homeostasis and disease, displaying features of tissue imprinting and heterogeneous activation. The disconnected picture of macrophage biology that has emerged from these observations is a barrier for integration across models or with in vitro macrophage activation paradigms. We set out to contextualize macrophage heterogeneity across mouse tissues and inflammatory conditions, specifically aiming to define a common framework of macrophage activation. We built a predictive model with which we mapped the activation of macrophages across 12 tissues and 25 biological conditions, finding a notable commonality and finite number of transcriptional profiles, in particular among infiltrating macrophages, which we modeled as defined stages along four conserved activation paths. These activation paths include a "phagocytic" regulatory path, an "inflammatory" cytokine-producing path, an "oxidative stress" antimicrobial path, or a "remodeling" extracellular matrix deposition path. We verified this model with adoptive cell transfer experiments and identified transient RELMɑ expression as a feature of monocyte-derived macrophage tissue engraftment. We propose that this integrative approach of macrophage classification allows the establishment of a common predictive framework of monocyte-derived macrophage activation in inflammation and homeostasis.


Subject(s)
Macrophage Activation , Macrophages , Animals , Cytokines/metabolism , Homeostasis , Inflammation/metabolism , Mice
17.
Cell Metab ; 34(5): 747-760.e6, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35508110

ABSTRACT

Adipose tissue (AT) plays a central role in systemic metabolic homeostasis, but its function during bacterial infection remains unclear. Following subcutaneous bacterial infection, adipocytes surrounding draining lymph nodes initiated a transcriptional response indicative of stimulation with IFN-γ and a shift away from lipid metabolism toward an immunologic function. Natural killer (NK) and invariant NK T (iNKT) cells were identified as sources of infection-induced IFN-γ in perinodal AT (PAT). IFN-γ induced Nos2 expression in adipocytes through a process dependent on nuclear-binding oligomerization domain 1 (NOD1) sensing of live intracellular bacteria. iNOS expression was coupled to metabolic rewiring, inducing increased diversion of extracellular L-arginine through the arginosuccinate shunt and urea cycle to produce nitric oxide (NO), directly mediating bacterial clearance. In vivo, control of infection in adipocytes was dependent on adipocyte-intrinsic sensing of IFN-γ and expression of iNOS. Thus, adipocytes are licensed by innate lymphocytes to acquire anti-bacterial functions during infection.


Subject(s)
Cues , Killer Cells, Natural , Adipocytes/metabolism , Immunity , Interferon-gamma/metabolism
18.
Cell Stem Cell ; 29(1): 131-148.e10, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34706256

ABSTRACT

Hematopoietic stem cells (HSCs) rely on complex regulatory networks to preserve stemness. Due to the scarcity of HSCs, technical challenges have limited our insights into the interplay between metabolites, transcription, and the epigenome. In this study, we generated low-input metabolomics, transcriptomics, chromatin accessibility, and chromatin immunoprecipitation data, revealing distinct metabolic hubs that are enriched in HSCs and their downstream multipotent progenitors. Mechanistically, we uncover a non-classical retinoic acid (RA) signaling axis that regulates HSC function. We show that HSCs rely on Cyp26b1, an enzyme conventionally considered to limit RA effects in the cell. In contrast to the traditional view, we demonstrate that Cyp26b1 is indispensable for production of the active metabolite 4-oxo-RA. Further, RA receptor beta (Rarb) is required for complete transmission of 4-oxo-RA-mediated signaling to maintain stem cells. Our findings emphasize that a single metabolite controls stem cell fate by instructing epigenetic and transcriptional attributes.


Subject(s)
Hematopoietic Stem Cells , Tretinoin , Cell Differentiation , Retinoic Acid 4-Hydroxylase/genetics , Signal Transduction , Tretinoin/pharmacology
19.
Sci Immunol ; 7(76): eadd3263, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36240286

ABSTRACT

Type 2 immunity is associated with adipose tissue (AT) homeostasis and infection with parasitic helminths, but whether AT participates in immunity to these parasites is unknown. We found that the fat content of mesenteric AT (mAT) declined in mice during infection with a gut-restricted helminth. This was associated with the accumulation of metabolically activated, interleukin-33 (IL-33), thymic stromal lymphopoietin (TSLP), and extracellular matrix (ECM)-producing stromal cells. These cells shared transcriptional features, including the expression of Dpp4 and Pi16, with multipotent progenitor cells (MPC) that have been identified in numerous tissues and are reported to be capable of differentiating into fibroblasts and adipocytes. Concomitantly, mAT became infiltrated with resident T helper 2 (TH2) cells that responded to TSLP and IL-33 by producing stromal cell-stimulating cytokines, including transforming growth factor ß1 (TGFß1) and amphiregulin. These TH2 cells expressed genes previously associated with type 2 innate lymphoid cells (ILC2), including Nmur1, Calca, Klrg1, and Arg1, and persisted in mAT for at least 11 months after anthelmintic drug-mediated clearance of infection. We found that MPC and TH2 cells localized to ECM-rich interstitial spaces that appeared shared between mesenteric lymph node, mAT, and intestine. Stromal cell expression of epidermal growth factor receptor (EGFR), the receptor for amphiregulin, was required for immunity to infection. Our findings point to the importance of MPC and TH2 cell interactions within the interstitium in orchestrating AT remodeling and immunity to an intestinal infection.


Subject(s)
Immunity, Innate , Interleukin-33 , Adipose Tissue/metabolism , Amphiregulin , Animals , Cytokines/metabolism , Dipeptidyl Peptidase 4 , ErbB Receptors , Lymphocytes , Mice , Th2 Cells , Transforming Growth Factor beta1
20.
J Immunol ; 182(12): 7353-63, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494257

ABSTRACT

We examined the expression and influence of IL-10 during influenza infection. We found that IL-10 does not impact sublethal infection, heterosubtypic immunity, or the maintenance of long-lived influenza Ag depots. However, IL-10-deficient mice display dramatically increased survival compared with wild-type mice when challenged with lethal doses of virus, correlating with increased expression of several Th17-associated cytokines in the lungs of IL-10-deficient mice during the peak of infection, but not with unchecked inflammation or with increased cellular responses. Foxp3(-) CD4 T cell effectors at the site of infection represent the most abundant source of IL-10 in wild-type mice during high-dose influenza infection, and the majority of these cells coproduce IFN-gamma. Finally, compared with predominant Th1 responses in wild-type mice, virus-specific T cell responses in the absence of IL-10 display a strong Th17 component in addition to a strong Th1 response and we show that Th17-polarized CD4 T cell effectors can protect naive mice against an otherwise lethal influenza challenge and utilize unique mechanisms to do so. Our results show that IL-10 expression inhibits development of Th17 responses during influenza infection and that this is correlated with compromised protection during high-dose primary, but not secondary, challenge.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Interleukin-10/deficiency , Interleukin-10/immunology , Interleukin-17/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigens, Viral/immunology , Forkhead Transcription Factors/immunology , Interleukin-10/genetics , Interleukin-10/metabolism , Mice , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/metabolism , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL