Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 300(1): 105488, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38000657

ABSTRACT

Cellular therapies are currently employed to treat a variety of disease processes. For T cell-based therapies, success often relies on the metabolic fitness of the T cell product, where cells with enhanced metabolic capacity demonstrate improved in vivo efficacy. AMP-activated protein kinase (AMPK) is a cellular energy sensor which combines environmental signals with cellular energy status to enforce efficient and flexible metabolic programming. We hypothesized that increasing AMPK activity in human T cells would augment their oxidative capacity, creating an ideal product for adoptive cellular therapies. Lentiviral transduction of the regulatory AMPKγ2 subunit stably enhanced intrinsic AMPK signaling and promoted mitochondrial respiration with increased basal oxygen consumption rates, higher maximal oxygen consumption rate, and augmented spare respiratory capacity. These changes were accompanied by increased proliferation and inflammatory cytokine production, particularly within restricted glucose environments. Introduction of AMPKγ2 into bulk CD4 T cells decreased RNA expression of canonical Th2 genes, including the cytokines interleukin (IL)-4 and IL-5, while introduction of AMPKγ2 into individual Th subsets universally favored proinflammatory cytokine production and a downregulation of IL-4 production in Th2 cells. When AMPKγ2 was overexpressed in regulatory T cells, both in vitro proliferation and suppressive capacity increased. Together, these data suggest that augmenting intrinsic AMPK signaling via overexpression of AMPKγ2 can improve the expansion and functional potential of human T cells for use in a variety of adoptive cellular therapies.


Subject(s)
AMP-Activated Protein Kinases , Gene Expression , Signal Transduction , T-Lymphocytes , Humans , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Cytokines/metabolism , Mitochondria/metabolism , Th2 Cells/metabolism , Gene Expression/genetics , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Memory T Cells/enzymology , Glucose/metabolism , CD4-Positive T-Lymphocytes/enzymology , Cells, Cultured
2.
Immun Ageing ; 20(1): 40, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37528458

ABSTRACT

BACKGROUND: Lower respiratory infections are a leading cause of severe morbidity and mortality among older adults. Despite ubiquitous exposure to common respiratory pathogens throughout life and near universal seropositivity, antibodies fail to effectively protect the elderly. Therefore, we hypothesized that severe respiratory illness in the elderly is due to deficient CD8+ T cell responses. RESULTS: Here, we establish an aged mouse model of human metapneumovirus infection (HMPV) wherein aged C57BL/6 mice exhibit worsened weight loss, clinical disease, lung pathology and delayed viral clearance compared to young adult mice. Aged mice generate fewer lung-infiltrating HMPV epitope-specific CD8+ T cells. Those that do expand demonstrate higher expression of PD-1 and other inhibitory receptors and are functionally impaired. Transplant of aged T cells into young mice and vice versa, as well as adoptive transfer of young versus aged CD8+ T cells into Rag1-/- recipients, recapitulates the HMPV aged phenotype, suggesting a cell-intrinsic age-associated defect. HMPV-specific aged CD8+ T cells exhibit a terminally exhausted TCF1/7- TOX+ EOMES+ phenotype. We confirmed similar terminal exhaustion of aged CD8+ T cells during influenza viral infection. CONCLUSIONS: This study identifies terminal CD8+ T cell exhaustion as a mechanism of severe disease from respiratory viral infections in the elderly.

3.
bioRxiv ; 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37398326

ABSTRACT

Allogeneic T cells reprogram their metabolism during acute graft-versus-host disease (GVHD) in a process reliant on the cellular energy sensor AMP-activated protein kinase (AMPK). Deletion of AMPK in donor T cells limits GVHD but still preserves homeostatic reconstitution and graft-versus-leukemia (GVL) effects. In the current studies, murine T cells lacking AMPK decreased oxidative metabolism at early timepoints post-transplant and were also unable to mediate a compensatory increase in glycolysis following inhibition of the electron transport chain. Human T cells lacking AMPK gave similar results, with glycolytic compensation impaired both in vitro and following expansion in vivo in a modified model of GVHD. Immunoprecipitation of proteins from day 7 allogeneic T cells, using an antibody specific to phosphorylated AMPK targets, recovered lower levels of multiple glycolysis-related proteins including the glycolytic enzymes aldolase, enolase, pyruvate kinase M (PKM), and glyceraldehyde 3-phosphate dehydrogenase (GAPDH). Functionally, murine T cells lacking AMPK exhibited impaired aldolase activity following anti-CD3/CD28 stimulation and a decrease in GAPDH activity on day 7 post-transplant. Importantly, these changes in glycolysis correlated with an impaired ability of AMPK KO T cells to produce significant amounts of interferon gamma (IFNγ) upon antigenic re-stimulation. Together these data highlight a significant role for AMPK in controlling oxidative and glycolytic metabolism in both murine and human T cells during GVHD and endorse further study of AMPK inhibition as a potential target for future clinical therapies. KEY POINTS: AMPK plays a key role in driving both and oxidative and glycolytic metabolism in T cells during graft-versus-host disease (GVHD)Absence of AMPK simultaneously impairs both glycolytic enzyme activity, most notably by aldolase, and interferon gamma (IFNγ) production.

4.
Immunometabolism (Cobham) ; 4(4): e00009, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36275779

ABSTRACT

Allogeneic stem cell transplantation is a curative therapy for multiple hematologic disorders. However, this life-saving procedure is often complicated by acute graft-versus-host disease (GVHD), where donor T cells attack tissues in the recipient's skin, liver, and gastrointestinal tract. Previous research has demonstrated that GVHD-causing T cells undergo significant metabolic reprogramming during disease pathogenesis, with an increased reliance on oxidative metabolism. This dependence makes metabolic modulation a potential approach to treat and/or prevent GVHD. Here, we provide an overview on the metabolic changes adopted by allogeneic T cells during disease initiation, highlighting the role played by AMP-activated protein kinase (AMPK) and identifying ways in which these insights might be leveraged to therapeutic advantage clinically.

6.
JCI Insight ; 6(14)2021 07 22.
Article in English | MEDLINE | ID: mdl-34291733

ABSTRACT

Allogeneic hematopoietic stem cell transplantation is a viable treatment for multiple hematologic diseases, but its application is often limited by graft-versus-host disease (GVHD), where donor T cells attack host tissues in the skin, liver, and gastrointestinal tract. Here, we examined the role of the cellular energy sensor AMP kinase (AMPK) in alloreactive T cells during GVHD development. Early posttransplant, AMPK activity increased more than 15-fold in allogeneic T cells, and transplantation of T cells deficient in both AMPKα1 and AMPKα2 decreased GVHD severity in multiple disease models. Importantly, a lack of AMPK lessened GVHD without compromising antileukemia responses or impairing lymphopenia-driven immune reconstitution. Mechanistically, absence of AMPK decreased both CD4+ and CD8+ effector T cell numbers as early as day 3 posttransplant, while simultaneously increasing regulatory T cell (Treg) percentages. Improvements in GVHD resulted from cell-intrinsic perturbations in conventional effector T cells as depletion of donor Tregs had minimal impact on AMPK-related improvements. Together, these results highlight a specific role for AMPK in allogeneic effector T cells early posttransplant and suggest that AMPK inhibition may be an innovative approach to mitigate GVHD while preserving graft-versus-leukemia responses and maintaining robust immune reconstitution.


Subject(s)
AMP-Activated Protein Kinases/deficiency , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , T-Lymphocytes, Regulatory/immunology , AMP-Activated Protein Kinases/genetics , Animals , Bone Marrow Transplantation/adverse effects , Disease Models, Animal , Female , Graft vs Host Disease/blood , Graft vs Host Disease/pathology , Humans , Male , Mice , Mice, Knockout , Severity of Illness Index , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous/adverse effects
7.
Curr Opin Hematol ; 27(6): 353-359, 2020 11.
Article in English | MEDLINE | ID: mdl-33003083

ABSTRACT

PURPOSE OF REVIEW: Controlling T cell activity through metabolic manipulation has become a prominent feature in immunology and practitioners of both adoptive cellular therapy (ACT) and haematopoietic stem cell transplantation (HSCT) have utilized metabolic interventions to control T cell function. This review will survey recent metabolic research efforts in HSCT and ACT to paint a broad picture of immunometabolism and highlight advances in each area. RECENT FINDINGS: In HSCT, recent publications have focused on modifying reactive oxygen species, sirtuin signalling or the NAD salvage pathway within alloreactive T cells and regulatory T cells. In ACT, metabolic interventions that bolster memory T cell development, increase mitochondrial density and function, or block regulatory signals in the tumour microenvironment (TME) have recently been published. SUMMARY: Metabolic interventions control immune responses. In ACT, efforts seek to improve the in-vivo metabolic fitness of T cells, while in HSCT energies have focused on blocking alloreactive T cell expansion or promoting regulatory T cells. Methods to identify new, metabolically targetable pathways, as well as the ability of metabolic biomarkers to predict disease onset and therapeutic response, will continue to advance the field towards clinically applicable interventions.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Immunotherapy, Adoptive/methods , T-Lymphocytes/immunology , Animals , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Humans , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , T-Lymphocytes/metabolism
8.
Front Immunol ; 11: 1517, 2020.
Article in English | MEDLINE | ID: mdl-32793207

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (aHSCT) is a curative therapy for a range of hematologic illnesses including aplastic anemia, sickle cell disease, immunodeficiency, and high-risk leukemia, but the efficacy of aHSCT is often undermined by graft-versus-host disease (GVHD), where T cells from the donor attack and destroy recipient tissues. Given the strong interconnection between T cell metabolism and cellular function, determining the metabolic pathways utilized by alloreactive T cells is fundamental to deepening our understanding of GVHD biology, including its initiation, propagation, and potential mitigation. This review summarizes the metabolic pathways available to alloreactive T cells and highlights key metabolic proteins and pathways linking T cell metabolism to effector function. Our current knowledge of alloreactive T cell metabolism is then explored, showing support for glycolysis, fat oxidation, and glutamine metabolism but also offering a potential explanation for how these presumably contradictory metabolic findings might be reconciled. Examples of additional ways in which metabolism impacts aHSCT are addressed, including the influence of butyrate metabolism on GVHD resolution. Finally, the caveats and challenges of assigning causality using our current metabolic toolbox is discussed, as well as likely future directions in immunometabolism, both to highlight the strengths of the current evidence as well as recognize some of its limitations.


Subject(s)
Isoantigens/immunology , Metabolic Networks and Pathways , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Energy Metabolism , Graft vs Host Disease/etiology , Graft vs Tumor Effect/immunology , Humans , Immunity, Cellular , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
9.
Gastroenterology ; 159(3): 1036-1050.e8, 2020 09.
Article in English | MEDLINE | ID: mdl-32445858

ABSTRACT

BACKGROUND & AIMS: Calcineurin is a ubiquitously expressed central Ca2+-responsive signaling molecule that mediates acute pancreatitis, but little is known about its effects. We compared the effects of calcineurin expression by hematopoietic cells vs pancreas in mouse models of pancreatitis and pancreatitis-associated lung inflammation. METHODS: We performed studies with mice with hematopoietic-specific or pancreas-specific deletion of protein phosphatase 3, regulatory subunit B, alpha isoform (PPP3R1, also called CNB1), in mice with deletion of CNB1 (Cnb1UBC△/△) and in the corresponding controls for each deletion of CNB1. Acute pancreatitis was induced in mice by administration of caerulein or high-pressure infusion of radiocontrast into biliopancreatic ducts; some mice were also given intraductal infusions of an adeno-associated virus vector that expressed nuclear factor of activated T -cells (NFAT)-luciferase into pancreas. Pancreas, bone marrow, liver, kidney, heart, and lung were collected and analyzed by histopathology, immunohistochemistry, and immunoblots; levels of cytokines were measured in serum. Mouse and human primary pancreatic acinar cells were transfected with a vector that expressed NFAT-luciferase and incubated with an agent that blocks interaction of NFAT with calcineurin; cells were analyzed by immunofluorescence. Calcineurin-mediated neutrophil chemotaxis and reactive oxygen species production were measured in neutrophils from mice. RESULTS: Mice with hematopoietic-specific deletion of CNB1 developed the same level of local pancreatic inflammation as control mice after administration of caerulein or infusion of radiocontrast into biliopancreatic ducts. Cnb1UBC△/△ mice or mice with pancreas-specific deletion of CNB1 developed less severe pancreatitis and reduced pancreatic inflammation after administration of caerulein or infusion of radiocontrast into biliopancreatic ducts compared with control mice. NFAT was activated in pancreas of Swiss Webster mice given caerulein or infusions of radiocontrast into biliopancreatic ducts. Blocking the interaction between calcineurin and NFAT did not reduce pancreatic acinar cell necrosis in response to caerulein or infusions of radiocontrast. Mice with hematopoietic-specific deletion of CNB1 (but not mice with pancreas-specific deletion of CNB1) had reduced infiltration of lung tissues by neutrophils. Neutrophil chemotaxis and production of reactive oxygen species were decreased after incubation with a calcineurin inhibitor. CONCLUSIONS: Hematopoietic and neutrophil expression of calcineurin promotes pancreatitis-associated lung inflammation, whereas pancreatic calcineurin promotes local pancreatic inflammation. The findings indicate that the protective effects of blocking or deleting calcineurin on pancreatitis are mediated by the source of its expression. This information should be used in the development of strategies to inhibit calcineurin for the prevention of pancreatitis and pancreatitis-associated lung inflammation.


Subject(s)
Acute Lung Injury/immunology , Calcineurin Inhibitors/therapeutic use , Calcineurin/metabolism , Calcium-Binding Proteins/metabolism , Muscle Proteins/metabolism , Pancreatitis/immunology , Acinar Cells/metabolism , Acute Lung Injury/blood , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Calcineurin/genetics , Calcineurin/immunology , Calcium-Binding Proteins/genetics , Cells, Cultured , Ceruletide/administration & dosage , Ceruletide/toxicity , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Muscle Proteins/genetics , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Pancreas/cytology , Pancreas/immunology , Pancreas/metabolism , Pancreatitis/chemically induced , Pancreatitis/complications , Pancreatitis/drug therapy , Primary Cell Culture
10.
Cancer Lett ; 414: 127-135, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29126914

ABSTRACT

The immunology community has made significant strides in recent years in using the immune system to target and eliminate cancer. Therapies such as hematopoietic stem cell transplantation (HSCT) are the standard of care treatment for several malignancies, while therapies incorporating chimeric antigen receptor (CAR) T cells or checkpoint molecule blockade have been revolutionary. However, these approaches are not optimal for all cancers and in some cases, have failed outright. The greatest obstacle to making these therapies more effective may be rooted in one of the most basic concepts of cell biology, metabolism. Research over the last decade has revealed that T cell proliferation and differentiation is intimately linked to robust changes in metabolic activity, delineation of which may provide ways to manipulate the immuno-oncologic responses to our advantage. Here, we provide a basic overview of T cell metabolism, discuss what is known about metabolic regulation of T cells during allogeneic HSCT, point to evidence on the importance of T cell metabolism during CAR T cell and solid tumor therapies, and speculate about the role for compounds that might have dual-action on both immune cells and tumor cells simultaneously.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Proliferation , Humans , Neoplasms/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Transplantation, Homologous , Tumor Microenvironment/immunology
11.
J Biol Chem ; 292(24): 10068-10086, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28432125

ABSTRACT

Hepatocellular carcinoma (HCC) is a common cancer that frequently overexpresses the c-Myc (Myc) oncoprotein. Using a mouse model of Myc-induced HCC, we studied the metabolic, biochemical, and molecular changes accompanying HCC progression, regression, and recurrence. These involved altered rates of pyruvate and fatty acid ß-oxidation and the likely re-directing of glutamine into biosynthetic rather than energy-generating pathways. Initial tumors also showed reduced mitochondrial mass and differential contributions of electron transport chain complexes I and II to respiration. The uncoupling of complex II's electron transport function from its succinate dehydrogenase activity also suggested a mechanism by which Myc generates reactive oxygen species. RNA sequence studies revealed an orderly progression of transcriptional changes involving pathways pertinent to DNA damage repair, cell cycle progression, insulin-like growth factor signaling, innate immunity, and further metabolic re-programming. Only a subset of functions deregulated in initial tumors was similarly deregulated in recurrent tumors thereby indicating that the latter can "normalize" some behaviors to suit their needs. An interactive and freely available software tool was developed to allow continued analyses of these and other transcriptional profiles. Collectively, these studies define the metabolic, biochemical, and molecular events accompanyingHCCevolution, regression, and recurrence in the absence of any potentially confounding therapies.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Liver/metabolism , Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Up-Regulation , Animals , Carcinogenesis , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , DNA Repair , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex II/genetics , Electron Transport Complex II/metabolism , Female , Gene Expression Profiling , Gene Silencing , Humans , Liver/pathology , Male , Mice, Transgenic , Mitochondrial Turnover , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/physiopathology , Neoplasm Recurrence, Local/prevention & control , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Proto-Oncogene Proteins c-myc/genetics , Reactive Oxygen Species/metabolism , Tumor Burden
12.
Mol Immunol ; 68(2 Pt C): 564-74, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26359186

ABSTRACT

The classic paradigm of T cell metabolism posits that activated Teff cells utilize glycolysis to keep pace with increased energetic demands, while resting and Tmem cells rely on the oxidation of fat. In contrast, Teff cells during graft-versus-host disease (GVHD) increase their reliance on oxidative metabolism and, in particular, on fatty acid oxidation (FAO). To explore the potential mechanisms driving adoption of this alternative metabolism, we first review key pathways regulating FAO across a variety of disparate tissue types, including liver, heart, and skeletal muscle. Based upon these comparative studies, we then outline a consensus network of transcriptional and signaling pathways that predict a model for regulating FAO in Teff cells during GVHD. This model raises important implications about the dynamic nature of metabolic reprogramming in T cells and suggests exciting future directions for further study of in vivo T cell metabolism.


Subject(s)
Fatty Acids/metabolism , Lymphocyte Activation/immunology , Models, Immunological , Oxidation-Reduction , T-Lymphocytes/metabolism , Animals , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Humans , Lipid Metabolism/immunology , T-Lymphocytes/immunology
13.
J Immunol ; 194(12): 5789-800, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25972478

ABSTRACT

The coinhibitory receptor programmed death-1 (PD-1) maintains immune homeostasis by negatively regulating T cell function and survival. Blockade of PD-1 increases the severity of graft-versus-host disease (GVHD), but the interplay between PD-1 inhibition and T cell metabolism is not well studied. We found that both murine and human alloreactive T cells concomitantly upregulated PD-1 expression and increased levels of reactive oxygen species (ROS) following allogeneic bone marrow transplantation. This PD-1(Hi)ROS(Hi) phenotype was specific to alloreactive T cells and was not observed in syngeneic T cells during homeostatic proliferation. Blockade of PD-1 signaling decreased both mitochondrial H2O2 and total cellular ROS levels, and PD-1-driven increases in ROS were dependent upon the oxidation of fatty acids, because treatment with etomoxir nullified changes in ROS levels following PD-1 blockade. Downstream of PD-1, elevated ROS levels impaired T cell survival in a process reversed by antioxidants. Furthermore, PD-1-driven changes in ROS were fundamental to establishing a cell's susceptibility to subsequent metabolic inhibition, because blockade of PD-1 decreased the efficacy of later F1F0-ATP synthase modulation. These data indicate that PD-1 facilitates apoptosis in alloreactive T cells by increasing ROS in a process dependent upon the oxidation of fat. In addition, blockade of PD-1 undermines the potential for subsequent metabolic inhibition, an important consideration given the increasing use of anti-PD-1 therapies in the clinic.


Subject(s)
Cell Survival , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigens/immunology , Apoptosis/genetics , Apoptosis/immunology , Bone Marrow Transplantation/adverse effects , Cell Survival/genetics , Fatty Acids/metabolism , Female , Gene Expression , Graft vs Host Disease/etiology , Heterografts , Humans , Mice , Mice, Transgenic , Oxidation-Reduction , Phenotype , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Reactive Oxygen Species/metabolism
14.
J Pharmacol Exp Ther ; 351(2): 298-307, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25125579

ABSTRACT

T-cell activation requires increased ATP and biosynthesis to support proliferation and effector function. Most models of T-cell activation are based on in vitro culture systems and posit that aerobic glycolysis is employed to meet increased energetic and biosynthetic demands. By contrast, T cells activated in vivo by alloantigens in graft-versus-host disease (GVHD) increase mitochondrial oxygen consumption, fatty acid uptake, and oxidation, with small increases of glucose uptake and aerobic glycolysis. Here we show that these differences are not a consequence of alloactivation, because T cells activated in vitro either in a mixed lymphocyte reaction to the same alloantigens used in vivo or with agonistic anti-CD3/anti-CD28 antibodies increased aerobic glycolysis. Using targeted metabolic (13)C tracer fate associations, we elucidated the metabolic pathway(s) employed by alloreactive T cells in vivo that support this phenotype. We find that glutamine (Gln)-dependent tricarboxylic acid cycle anaplerosis is increased in alloreactive T cells and that Gln carbon contributes to ribose biosynthesis. Pharmacological modulation of oxidative phosphorylation rapidly reduces anaplerosis in alloreactive T cells and improves GVHD. On the basis of these data, we propose a model of T-cell metabolism that is relevant to activated lymphocytes in vivo, with implications for the discovery of new drugs for immune disorders.


Subject(s)
Graft vs Host Disease/immunology , Isoantigens/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , CD28 Antigens/immunology , CD3 Complex/immunology , Citric Acid Cycle/immunology , Female , Glutamine/metabolism , Glycolysis/immunology , Graft vs Host Disease/metabolism , Mice , Oxidative Phosphorylation , Ribose/biosynthesis
15.
Blood ; 122(18): 3230-7, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-24046012

ABSTRACT

Activated T cells require increased energy to proliferate and mediate effector functions, but the metabolic changes that occur in T cells following stimulation in vivo are poorly understood, particularly in the context of inflammation. We have previously shown that T cells activated during graft-versus-host disease (GVHD) primarily rely on oxidative phosphorylation to synthesize adenosine 5'-triphosphate. Here, we demonstrate that alloreactive effector T cells (Teff) use fatty acids (FAs) as a fuel source to support their in vivo activation. Alloreactive T cells increased FA transport, elevated levels of FA oxidation enzymes, up-regulated transcriptional coactivators to drive oxidative metabolism, and increased their rates of FA oxidation. Importantly, increases in FA transport and up-regulation of FA oxidation machinery occurred specifically in T cells during GVHD and were not seen in Teff following acute activation. Pharmacological blockade of FA oxidation decreased the survival of alloreactive T cells but did not influence the survival of T cells during normal immune reconstitution. These studies suggest that pathways controlling FA metabolism might serve as therapeutic targets to treat GVHD and other T-cell-mediated immune diseases.


Subject(s)
Fatty Acids/immunology , Graft vs Host Disease/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Animals , Blotting, Western , Bone Marrow Transplantation/methods , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/immunology , Carnitine O-Palmitoyltransferase/metabolism , Enzyme Inhibitors/pharmacology , Epoxy Compounds/pharmacology , Fatty Acids/metabolism , Female , Flow Cytometry , Graft vs Host Disease/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism , Transplantation, Homologous
16.
Immunol Rev ; 249(1): 104-15, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22889218

ABSTRACT

For several decades, it has been known that T-cell activation in vitro leads to increased glycolytic metabolism that fuels proliferation and effector function. Recently, this simple model has been complicated by the observation that different T-cell subsets differentially regulate fundamental metabolic pathways under the control of distinct molecular regulators. Although the majority of these data have been generated in vitro, several recent studies have documented the metabolism of T cells activated in vivo. Here, we review the recent data surrounding the differential regulation of metabolism by distinct T-cell subsets in vitro and in vivo and discuss how differential metabolic regulation might facilitate T-cell function vis-à-vis proliferation, survival, and energy production. We further discuss the important therapeutic implications of differential metabolism across T-cell subsets and review recent successes in exploiting lymphocyte metabolism to treat immune-mediated diseases.


Subject(s)
Immunomodulation , Lymphocyte Activation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Cell Differentiation , Cell Proliferation , Energy Metabolism , Glycolysis , Humans , Mitochondria/metabolism , Oxidative Phosphorylation , Reactive Oxygen Species/metabolism
17.
J Immunol ; 183(12): 7909-18, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19933868

ABSTRACT

Using hen egg-white lysozyme, the effect of blood proteins on CD4 thymic cells was examined. A small fraction of i.v. injected hen egg-white lysozyme rapidly entered the thymus into the medulla. There it was captured and presented by dendritic cells (DCs) to thymocytes from two TCR transgenic mice, one directed to a dominant peptide and a second to a poorly displayed peptide, both presented by MHC class II molecules I-A(k). Presentation by DC led to negative selection and induction of regulatory T cells, independent of epithelial cells. Presentation took place at very low levels, less than 100 peptide-MHC complexes per DC. Such low levels could induce negative selection, but even lower levels could induce regulatory T cells. The anatomy of the thymus-blood barrier, the highly efficient presentation by DC, together with the high sensitivity of thymic T cells to peptide-MHC complexes, results in blood protein Ags having a profound effect on thymic T cells.


Subject(s)
Cell Differentiation/immunology , Down-Regulation/immunology , Growth Inhibitors/metabolism , Muramidase/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Cell Differentiation/genetics , Chickens , Clone Cells , Dendritic Cells/enzymology , Dendritic Cells/immunology , Down-Regulation/genetics , Female , Growth Inhibitors/antagonists & inhibitors , Growth Inhibitors/blood , Hybridomas , Immune Tolerance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muramidase/antagonists & inhibitors , Muramidase/blood , Ovalbumin/antagonists & inhibitors , Ovalbumin/blood , Ovalbumin/immunology , Receptors, Antigen, T-Cell/blood , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Spleen/cytology , Spleen/enzymology , Spleen/immunology , T-Lymphocytes, Regulatory/enzymology , Thymus Gland/enzymology , Up-Regulation/genetics , Up-Regulation/immunology
18.
J Immunol ; 175(7): 4347-54, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16177075

ABSTRACT

Two novel transgenic (Tg) strains were created expressing hen egg-white lysozyme (HEL) in a pancreas-specific fashion. RmHP.111 mice had levels of HEL per cell similar to that of the established ILK-3 strain, while RmHP.117 mice had 10-fold lower levels (50,000 molecules per cell). When bred to 3A9 TCR Tg mice, negative selection occurred equally in all three double-Tg combinations, yet only ILK-3 x 3A9 and RmHP.111 x 3A9 mice became diabetic. Additionally, activated 3A9 cells readily transferred diabetes into ILK-3 or RmHP.111 mice, but only marginally into the RmHP.117 strain. In the peripancreatic lymph node, division of naive 3A9 cells was similar between RmHP.111 and RmHP.117 strains, but pancreatic APCs from RmHP.111 x 3A9 mice stimulated HEL-reactive cells to a much greater degree than those from RmHP.117 x 3A9 mice. In this model, diabetes was dependent upon both initial priming in the peripancreatic lymph node and subsequent presentation in the pancreas, with disease incidence predicted by the beta cell level of autoantigen.


Subject(s)
Autoantigens/metabolism , Diabetes Mellitus, Type 1/diagnosis , Islets of Langerhans/immunology , Animals , Antibodies/metabolism , Cell Line , Clonal Deletion/physiology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Enzyme-Linked Immunosorbent Assay , Insulin/genetics , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muramidase/genetics , Muramidase/metabolism , Predictive Value of Tests , Promoter Regions, Genetic , Rats , Receptors, Antigen, T-Cell/genetics
19.
J Immunol ; 173(11): 6627-34, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15557153

ABSTRACT

Immunization with the hen egg-white lysozyme (HEL) protein induces T cells to various of its peptide determinants. The distribution of such T cells, however, does not correlate with the peptide level of each epitope on class II molecules. For this reason, we sought information on the cells responsible for Ag presentation following immunization, hoping to understand the lack of immunodominance in this system. By tracking HEL, and the ensuing peptide/MHC complexes, we find the following: 1) that HEL in the draining lymph node gets concentrated in a limited number of APC, particularly in dendritic cells and macrophages, 2) that these APC are functionally capable of presenting both major and minor determinants of HEL over a 100-fold range of Ag dose, and 3) that B cells present Ag gained at early times after immunization, but only following higher dose immunization. These data indicate that the breadth of a response is maintained over a wide dosage range by concentration of Ag in a limited number of cells presenting high levels and a great diversity of epitopes.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens/administration & dosage , Antigens/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Muramidase/administration & dosage , Muramidase/immunology , Animals , Antigen Presentation , Antigen-Presenting Cells/pathology , Antigens/metabolism , B-Lymphocyte Subsets/pathology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Histocompatibility Antigens Class II/metabolism , Immunization , Immunodominant Epitopes/immunology , Immunodominant Epitopes/metabolism , Injections, Subcutaneous , Iodine Radioisotopes/metabolism , Lymphocyte Count , Lymphopenia/genetics , Lymphopenia/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Muramidase/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...