Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Immunity ; 44(6): 1299-311, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27234056

ABSTRACT

Mitochondrial respiration is regulated in CD8(+) T cells during the transition from naive to effector and memory cells, but mechanisms controlling this process have not been defined. Here we show that MCJ (methylation-controlled J protein) acted as an endogenous break for mitochondrial respiration in CD8(+) T cells by interfering with the formation of electron transport chain respiratory supercomplexes. Metabolic profiling revealed enhanced mitochondrial metabolism in MCJ-deficient CD8(+) T cells. Increased oxidative phosphorylation and subcellular ATP accumulation caused by MCJ deficiency selectively increased the secretion, but not expression, of interferon-γ. MCJ also adapted effector CD8(+) T cell metabolism during the contraction phase. Consequently, memory CD8(+) T cells lacking MCJ provided superior protection against influenza virus infection. Thus, MCJ offers a mechanism for fine-tuning CD8(+) T cell mitochondrial metabolism as an alternative to modulating mitochondrial mass, an energetically expensive process. MCJ could be a therapeutic target to enhance CD8(+) T cell responses.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Electron Transport Chain Complex Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Molecular Chaperones/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Adenosine Triphosphate/metabolism , Animals , Cell Respiration , Cells, Cultured , Immunologic Memory , Interferon-gamma/metabolism , Lymphocyte Activation , Metabolome , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , Molecular Chaperones/genetics , Oxidative Phosphorylation
2.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34507993

ABSTRACT

Interleukin 6 (IL-6) is known to regulate the CD4 T cell function by inducing gene expression of a number of cytokines through activation of Stat3 transcription factor. Here, we reveal that IL-6 strengthens the mechanics of CD4 T cells. The presence of IL-6 during activation of mouse and human CD4 T cells enhances their motility (random walk and exploratory spread), resulting in an increase in travel distance and higher velocity. This is an intrinsic effect of IL-6 on CD4 T-cell fitness that involves an increase in mitochondrial Ca2+ Although Stat3 transcriptional activity is dispensable for this process, IL-6 uses mitochondrial Stat3 to enhance mitochondrial Ca2+-mediated motility of CD4 T cells. Thus, through a noncanonical pathway, IL-6 can improve competitive fitness of CD4 T cells by facilitating cell motility. These results could lead to alternative therapeutic strategies for inflammatory diseases in which IL-6 plays a pathogenic role.


Subject(s)
Cell Movement/physiology , Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Calcium/metabolism , Cell Differentiation/drug effects , Cell Movement/drug effects , Cytokines/metabolism , Female , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , STAT3 Transcription Factor/physiology , Signal Transduction/drug effects
3.
J Autoimmun ; 82: 47-61, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28551033

ABSTRACT

T lymphocyte homeostatic proliferation, driven by the engagement of T cell antigen receptor with self-peptide/major histocompatibility complexes, and signaling through the common γ-chain-containing cytokine receptors, is critical for the maintenance of the T cell compartment and is regulated by the Fas death receptor (Fas, CD95). In the absence of Fas, Fas-deficient lymphoproliferation spontaneous mutation (lpr) mice accumulate homeostatically expanded T cells. The functional consequences of sequential rounds of homeostatic expansion are not well defined. We thus examined the gene expression profiles of murine wild-type and Fas-deficient lpr CD8+ T cell subsets that have undergone different amounts of homeostatic proliferation as defined by their level of CD44 expression, and the CD4-CD8-TCRαß+ T cell subset that results from extensive homeostatic expansion of CD8+ T cells. Our studies show that recurrent T cell homeostatic proliferation results in global gene expression changes, including the progressive upregulation of both cytolytic proteins such as Fas-Ligand and granzyme B as well as inhibitory proteins such as programmed cell death protein 1 (PD-1) and lymphocyte activating 3 (Lag3). These findings provide an explanation for how augmented T cell homeostatic expansion could lead to the frequently observed clinical paradox of simultaneous autoinflammatory and immunodeficiency syndromes and provide further insight into the regulatory programs that control chronically stimulated T cells.


Subject(s)
Inflammation/genetics , Inflammation/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Biomarkers , Cell Proliferation , Cell Survival/genetics , Computational Biology/methods , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Gene Expression Profiling , Homeostasis , Immunomodulation , Inflammation/metabolism , Mice , Mice, Transgenic , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/metabolism , Transcriptome
4.
J Allergy Clin Immunol ; 137(3): 822-32.e7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26435004

ABSTRACT

BACKGROUND: Evidence for association between asthma and the unfolded protein response is emerging. Endoplasmic reticulum resident protein 57 (ERp57) is an endoplasmic reticulum-localized redox chaperone involved in folding and secretion of glycoproteins. We have previously demonstrated that ERp57 is upregulated in allergen-challenged human and murine lung epithelial cells. However, the role of ERp57 in asthma pathophysiology is unknown. OBJECTIVES: Here we sought to examine the contribution of airway epithelium-specific ERp57 in the pathogenesis of allergic asthma. METHODS: We examined the expression of ERp57 in human asthmatic airway epithelium and used murine models of allergic asthma to evaluate the relevance of epithelium-specific ERp57. RESULTS: Lung biopsy specimens from asthmatic and nonasthmatic patients revealed a predominant increase in ERp57 levels in epithelium of asthmatic patients. Deletion of ERp57 resulted in a significant decrease in inflammatory cell counts and airways resistance in a murine model of allergic asthma. Furthermore, we observed that disulfide bridges in eotaxin, epidermal growth factor, and periostin were also decreased in the lungs of house dust mite-challenged ERp57-deleted mice. Fibrotic markers, such as collagen and α smooth muscle actin, were also significantly decreased in the lungs of ERp57-deleted mice. Furthermore, adaptive immune responses were dispensable for house dust mite-induced endoplasmic reticulum stress and airways fibrosis. CONCLUSIONS: Here we show that ERp57 levels are increased in the airway epithelium of asthmatic patients and in mice with allergic airways disease. The ERp57 level increase is associated with redox modification of proinflammatory, apoptotic, and fibrotic mediators and contributes to airways hyperresponsiveness. The strategies to inhibit ERp57 specifically within the airways epithelium might provide an opportunity to alleviate the allergic asthma phenotype.


Subject(s)
Allergens/immunology , Asthma/immunology , Asthma/metabolism , Protein Disulfide-Isomerases/metabolism , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Animals , Asthma/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biopsy , Caspase 3/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Fibrosis , Gene Expression , Humans , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Transgenic , Protein Disulfide-Isomerases/genetics , Respiratory Hypersensitivity/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , bcl-2 Homologous Antagonist-Killer Protein/chemistry , bcl-2 Homologous Antagonist-Killer Protein/metabolism
5.
J Biol Chem ; 289(2): 1183-91, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24275659

ABSTRACT

Caspase-8 is now appreciated to govern both apoptosis following death receptor ligation and cell survival and growth via inhibition of the Ripoptosome. Cells must therefore carefully regulate the high level of caspase-8 activity during apoptosis versus the modest levels observed during cell growth. The caspase-8 paralogue c-FLIP is a good candidate for a molecular rheostat of caspase-8 activity. c-FLIP can inhibit death receptor-mediated apoptosis by competing with caspase-8 for recruitment to FADD. However, full-length c-FLIPL can also heterodimerize with caspase-8 independent of death receptor ligation and activate caspase-8 via an activation loop in the C terminus of c-FLIPL. This triggers cleavage of c-FLIPL at Asp-376 by caspase-8 to produce p43FLIP. The continued function of p43FLIP has, however, not been determined. We demonstrate that acute deletion of endogenous c-FLIP in murine effector T cells results in loss of caspase-8 activity and cell death. The lethality and caspase-8 activity can both be rescued by the transgenic expression of p43FLIP. Furthermore, p43FLIP associates with Raf1, TRAF2, and RIPK1, which augments ERK and NF-κB activation, IL-2 production, and T cell proliferation. Thus, not only is c-FLIP the initiator of caspase-8 activity during T cell activation, it is also an initial caspase-8 substrate, with cleaved p43FLIP serving to both stabilize caspase-8 activity and promote activation of pathways involved with T cell growth.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , NF-kappa B/metabolism , Peptide Fragments/metabolism , T-Lymphocytes/metabolism , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/chemistry , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 8/genetics , Cell Proliferation , Cell Survival , Cells, Cultured , Humans , Immunoblotting , Interleukin-2/metabolism , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peptide Fragments/genetics , Proto-Oncogene Proteins c-raf , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/cytology , TNF Receptor-Associated Factor 2/metabolism
6.
J Cell Physiol ; 230(8): 1929-43, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25573156

ABSTRACT

Glioblastoma (GBM), the most common primary adult malignant brain tumor, is associated with a poor prognosis due, in part, to tumor recurrence mediated by chemotherapy and radiation resistant glioma stem-like cells (GSCs). The metabolic and epigenetic state of GSCs differs from their non-GSC counterparts, with GSCs exhibiting greater glycolytic metabolism and global hypoacetylation. However, little attention has been focused on the potential use of acetate supplementation as a therapeutic approach. N-acetyl-l-aspartate (NAA), the primary storage form of brain acetate, and aspartoacylase (ASPA), the enzyme responsible for NAA catalysis, are significantly reduced in GBM tumors. We recently demonstrated that NAA supplementation is not an appropriate therapeutic approach since it increases GSC proliferation and pursued an alternative acetate source. The FDA approved food additive Triacetin (glyceryl triacetate, GTA) has been safely used for acetate supplementation therapy in Canavan disease, a leukodystrophy due to ASPA mutation. This study characterized the effects of GTA on the proliferation and differentiation of six primary GBM-derived GSCs relative to established U87 and U251 GBM cell lines, normal human cerebral cortical astrocytes, and murine neural stem cells. GTA reduced proliferation of GSCs greater than established GBM lines. Moreover, GTA reduced growth of the more aggressive mesenchymal GSCs greater than proneural GSCs. Although sodium acetate induced a dose-dependent reduction of GSC growth, it also reduced cell viability. GTA-mediated growth inhibition was not associated with differentiation, but increased protein acetylation. These data suggest that GTA-mediated acetate supplementation is a novel therapeutic strategy to inhibit GSC growth.


Subject(s)
Antineoplastic Agents/pharmacology , Glioblastoma/pathology , Neoplastic Stem Cells/drug effects , Triacetin/pharmacology , Adult , Aged , Animals , Astrocytes/drug effects , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Female , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Neural Stem Cells/drug effects , Reverse Transcriptase Polymerase Chain Reaction
7.
Immunology ; 135(4): 276-86, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22117649

ABSTRACT

Caspase-8 serves two paradoxical roles in T lymphocytes: it initiates apoptosis following death receptor engagement, and is also indispensible for proliferation following T-cell antigen receptor (TCR) signalling. These opposing processes appear to be controlled by both spatial and quantitative differences in caspase-8 activation. Given differences in the turnover of T-cell subsets, we compared caspase activity and susceptibility to cell death following TCR restimulation in murine CD4(+) and CD8(+) αß T cells and γδ T cells. We observed a spectrum of caspase activity in non-dying effector T cells in which CD4(+) T cells manifested the lowest levels of active caspases whereas γδ T cells manifested the highest levels. Further analysis revealed that most of the difference in T-cell subsets was the result of high levels of active caspase-3 in non-dying effector γδ T cells. Despite this, γδ T cells manifested little spontaneous or CD3 restimulation-induced cell death as the result of confinement of active caspases to the cell membrane. By contrast, CD4(+) T cells were highly sensitive to CD3-induced cell death, associated with the appearance of active caspases in the cytoplasm and cleavage of the caspase substrates Bid and ICAD. Hence, the location and amount of active caspases distinguishes effector T-cell subsets and profoundly influences the fate of the T-cell response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Caspase 3/metabolism , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Animals , Apoptosis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Caspase 3/genetics , Caspase 8/metabolism , Cells, Cultured , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction
8.
Int Immunol ; 23(2): 75-88, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21266499

ABSTRACT

Fas-deficient mice (Fas(lpr/lpr)) and humans have profoundly dysregulated T lymphocyte homeostasis, which manifests as an accumulation of CD4(+) and CD8(+) T cells as well as an unusual population of CD4(-)CD8(-)TCRαß(+) T cells. To date, no unifying model has explained both the increased T-cell numbers and the origin of the CD4(-)CD8(-)TCRαß(+) T cells. As Fas(lpr/lpr) mice raised in a germ-free environment still manifest lymphadenopathy, we considered that this process is primarily driven by recurrent low-avidity TCR signaling in response to self-peptide/MHC as occurs during homeostatic proliferation. In these studies, we developed two independent systems to decrease the number of self-peptide/MHC contacts. First, expression of MHC class I was reduced in OT-I TCR transgenic mice. Although OT-I Fas(lpr/lpr) mice did not develop lymphadenopathy characteristic of Fas(lpr/lpr) mice, in the absence of MHC class I, OT-I Fas(lpr/lpr) T cells accumulated as both CD8(+) and CD4(-)CD8(-) T cells. In the second system, re-expression of ß(2)m limited to thymic cortical epithelial cells of Fas(lpr/lpr) ß(2)m-deficient mice yielded a model in which polyclonal CD8(+) thymocytes entered a peripheral environment devoid of MHC class I. These mice accumulated significantly greater numbers of CD4(-)CD8(-)TCRαß(+) T cells than conventional Fas(lpr/lpr) mice. Thus, Fas shapes the peripheral T-cell repertoire by regulating the survival of a subset of T cells proliferating in response to limited self-peptide/MHC contacts.


Subject(s)
Histocompatibility Antigens Class I/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , fas Receptor/immunology , Animals , CD5 Antigens/immunology , Cell Proliferation , Female , Lymphocyte Activation , Lymphopenia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets/immunology
9.
Antioxid Redox Signal ; 36(7-9): 410-422, 2022 03.
Article in English | MEDLINE | ID: mdl-34328790

ABSTRACT

Significance: Numerous abnormalities in T cells have been described in patients with systemic lupus erythematosus (SLE), including lymphopenia, DNA demethylation, expression of endogenous retroviruses (ERVs), increased cell death, enlarged mitochondria, production of reactive oxygen species (ROS), and the appearance of unusual CD4-CD8- T cells. Our studies propose a model in which accelerated homeostatic proliferation of T cells promotes an epigenetic and metabolic program, leading to this cluster of abnormalities. Recent Advances: Growing knowledge of the innate immune disorders in SLE has included increased mitochondrial size and ROS production that induces oligomerization of the mitochondrial antiviral signaling (MAVS) protein and type I interferon production, as well as DNA demethylation, upregulation of inflammatory genes, and expression of certain ERVs in SLE peripheral blood mononuclear cells. All these events are part of the cellular program that occurs during homeostatic proliferation of T cells. Evidence from a murine model of SLE as well as in human SLE reveals that increased T cell homeostatic proliferation may be a driving factor in these processes. Critical Issues: Despite extensive knowledge of the myriad autoantibodies in SLE and other immune abnormalities, a cogent model has been lacking to link the numerous and seemingly disparate immune aberrations. This may partly explain the general lack of new drugs specifically for SLE in over 50 years. A more coherent model of SLE would not only unify the variety of immune abnormalities is SLE but would also suggest new therapies. Future Directions: The model of augmented homeostatic proliferation leading to increased mitochondrial mass, ROS, DNA demethylation, and upregulation of inflammatory genes suggests strategic new targets for SLE, including antioxidants and certain inhibitors of metabolism. Antioxid. Redox Signal. 36, 410-422.


Subject(s)
Leukocytes, Mononuclear , Lupus Erythematosus, Systemic , Animals , CD4-Positive T-Lymphocytes , Cell Proliferation , Epigenesis, Genetic , Humans , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/genetics , Mice , Oxidation-Reduction , T-Lymphocytes/metabolism , Up-Regulation
10.
Infect Immun ; 79(10): 3940-6, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21768278

ABSTRACT

Little is known regarding the function of γδ T cells, although they accumulate at sites of inflammation in infections and autoimmune disorders. We previously observed that γδ T cells in vitro are activated by Borrelia burgdorferi in a TLR2-dependent manner. We now observe that the activated γδ T cells can in turn stimulate dendritic cells in vitro to produce cytokines and chemokines that are important for the adaptive immune response. This suggested that in vivo γδ T cells may assist in activating the adaptive immune response. We examined this possibility in vivo and observed that γδ T cells are activated and expand in number during Borrelia infection, and this was reduced in the absence of TLR2. Furthermore, in the absence of γδ T cells, there was a significantly blunted response of adaptive immunity, as reflected in reduced expansion of T and B cells and reduced serum levels of anti-Borrelia antibodies, cytokines, and chemokines. This paralleled a greater Borrelia burden in γδ-deficient mice as well as more cardiac inflammation. These findings are consistent with a model of γδ T cells functioning to promote the adaptive immune response during infection.


Subject(s)
Borrelia burgdorferi/immunology , Lyme Disease/immunology , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Adaptive Immunity , Animals , Antibodies, Bacterial/blood , Chemokines/blood , Cytokines/blood , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Humans , Lyme Disease/microbiology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/physiology , T-Lymphocyte Subsets/immunology
11.
Eur J Immunol ; 40(11): 3043-53, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21061436

ABSTRACT

The size of the peripheral T-lymphocyte compartment is governed by complex homeostatic mechanisms that balance T-cell proliferation and death. Proliferation and survival signals are mediated in part by recurrent self-peptide/MHC-TCR interactions and signaling by the common γ chain-containing cytokine receptors, including those for IL-7 and IL-15. We have previously shown that the death receptor Fas (CD95/APO-1) regulates apoptosis in response to repeated TCR stimulation, whereas the Bcl-2 homology domain 3-only protein Bim mediates cytokine withdrawal-induced apoptosis. We therefore reasoned that these two molecules might cooperate in the regulation of homeostatic proliferation. In this study, we observe that the combined loss of Fas and Bim synergistically enhances the accumulation of T cells in lymphopenic host mice, and this is particularly pronounced for the unusual CD4(-) CD8(-) TCRαß(+) T cells that are characteristic of Fas-deficient (Fas(lpr/lpr) ) mice. Our findings demonstrate that these CD4(-) CD8(-) TCRαß(+) T cells arise from homeostatic proliferation of CD8(+) T cells. These studies also underscore the profound rate of baseline T-cell proliferation that likely occurs in wild-type mice even in the absence of foreign antigen, and the consequent need for its coordinated regulation by multiple death-signaling pathways.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Homeostasis/immunology , Membrane Proteins/immunology , Proto-Oncogene Proteins/immunology , fas Receptor/immunology , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/agonists , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Histocompatibility Antigens/immunology , Homeostasis/genetics , Interleukin-15/genetics , Interleukin-15/immunology , Interleukin-7/genetics , Interleukin-7/immunology , Membrane Proteins/agonists , Membrane Proteins/genetics , Mice , Mice, Knockout , Peptides/immunology , Proto-Oncogene Proteins/agonists , Proto-Oncogene Proteins/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction/genetics , Signal Transduction/immunology , fas Receptor/agonists , fas Receptor/genetics
12.
Nat Commun ; 12(1): 2804, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990571

ABSTRACT

Chemotherapy remains the standard of care for most cancers worldwide, however development of chemoresistance due to the presence of the drug-effluxing ATP binding cassette (ABC) transporters remains a significant problem. The development of safe and effective means to overcome chemoresistance is critical for achieving durable remissions in many cancer patients. We have investigated the energetic demands of ABC transporters in the context of the metabolic adaptations of chemoresistant cancer cells. Here we show that ABC transporters use mitochondrial-derived ATP as a source of energy to efflux drugs out of cancer cells. We further demonstrate that the loss of methylation-controlled J protein (MCJ) (also named DnaJC15), an endogenous negative regulator of mitochondrial respiration, in chemoresistant cancer cells boosts their ability to produce ATP from mitochondria and fuel ABC transporters. We have developed MCJ mimetics that can attenuate mitochondrial respiration and safely overcome chemoresistance in vitro and in vivo. Administration of MCJ mimetics in combination with standard chemotherapeutic drugs could therefore become an alternative strategy for treatment of multiple cancers.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Drug Resistance, Neoplasm/physiology , Mitochondria/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Line, Tumor , Cell Respiration/drug effects , Cell Respiration/physiology , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Resistance, Multiple/physiology , Female , HSP40 Heat-Shock Proteins/deficiency , HSP40 Heat-Shock Proteins/metabolism , Humans , In Vitro Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Oxygen Consumption/drug effects
13.
Immunology ; 131(3): 426-37, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20553337

ABSTRACT

Evolving models of immune tolerance have challenged the view that the response of the maternal immune system to environmental or fetal antigens must be suppressed or deviated. CD8 T cells play a central role in the immune response to viruses and intracellular pathogens so the maintenance of both the number and function of these cells is critical to protect both the mother and fetus. We show that the numbers of maternal CD8 T cells in both the spleen and the uterine draining lymph nodes are transiently increased at mid-gestation and this correlates with enhanced CD8 T-cell proliferation and an increased relative expression of both pro-survival and pro-apoptotic molecules. In transgenic mice bearing T-cell antigen receptors specific for the male HY or allo-antigens, the transgenic CD8 T cells retain the ability to proliferate and function during pregnancy. Moreover, anti-HY T-cell receptor transgenic mice have normal numbers of male pups despite the presence of CD8 T cells at the maternal-fetal interface. These data suggest that pregnancy is a dynamic state in which CD8 T-cell turnover is increased while the function and ending size of the CD8 T-cell compartment are maintained.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Fas Ligand Protein/metabolism , Homeostasis , Interleukin-7 Receptor alpha Subunit/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis/genetics , Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Count , Cell Proliferation , Cells, Cultured , Fas Ligand Protein/immunology , Female , H-Y Antigen/immunology , Immune Tolerance/genetics , Interleukin-7 Receptor alpha Subunit/immunology , Litter Size/genetics , Litter Size/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy/immunology , Proto-Oncogene Proteins c-bcl-2/immunology , Receptors, Antigen, T-Cell/genetics
14.
J Immunol ; 181(4): 2392-8, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18684928

ABSTRACT

Activation of the innate immune system typically precedes engagement of adaptive immunity. Cells at the interface between these two arms of the immune response are thus critical to provide full engagement of host defense. Among the innate T cells at this interface are gammadelta T cells. gammadelta T cells contribute to the defense from a variety of infectious organisms, yet little is understood regarding how they are activated. We have previously observed that human gammadelta T cells of the Vdelta1 subset accumulate in inflamed joints in Lyme arthritis and proliferate in response to stimulation with the causative spirochete, Borrelia burgdorferi. We now observe that murine gammadelta T cells are also activated by B. burgdorferi and that in both cases the activation is indirect via TLR stimulation on dendritic cells or monocytes. Furthermore, B. burgdorferi stimulation of monocytes via TLR, and secondary activation of gammadelta T cells, are both caspase-dependent.


Subject(s)
Borrelia burgdorferi/immunology , Caspases/physiology , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , Toll-Like Receptors/physiology , Animals , Cell Communication/immunology , Cells, Cultured , Clone Cells , Coculture Techniques , Dendritic Cells/immunology , Humans , Lyme Disease/enzymology , Lyme Disease/immunology , Lyme Disease/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Synovial Membrane/cytology , Synovial Membrane/immunology , Synovial Membrane/metabolism , T-Lymphocyte Subsets/enzymology
15.
Immunohorizons ; 4(10): 679-687, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097564

ABSTRACT

Systemic lupus erythematosus (SLE) is characterized by increased DNA demethylation in T cells, although it is unclear whether this occurs primarily in a subset of SLE T cells. The process driving the DNA demethylation and the consequences on overall gene expression are also poorly understood and whether this represents a secondary consequence of SLE or a primary contributing factor. Lupus-prone lpr mice accumulate large numbers of T cells with age because of a mutation in Fas (CD95). The accumulating T cells include an unusual population of CD4-CD8-TCR-αß+ (DN) T cells that arise from CD8+ precursors and are also found in human SLE. We have previously observed that T cell accumulation in lpr mice is due to dysregulation of T cell homeostatic proliferation, which parallels an increased expression of numerous genes in the DN subset, including several proinflammatory molecules and checkpoint blockers. We thus determined the DNA methylome in lpr DN T cells compared with their CD8+ precursors. Our findings show that DN T cells manifest discrete sites of extensive demethylation throughout the genome, and these sites correspond to the location of a large proportion of the upregulated genes. Thus, dysregulated homeostatic proliferation in lpr mice and consequent epigenetic alterations may be a contributing factor to lupus pathogenesis.


Subject(s)
DNA Demethylation , Lupus Erythematosus, Systemic/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , fas Receptor/immunology , Animals , Cell Proliferation , Gene Expression Regulation , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Lupus Sci Med ; 7(1)2020 04.
Article in English | MEDLINE | ID: mdl-32343673

ABSTRACT

OBJECTIVES: Recent investigations in humans and mouse models with lupus have revealed evidence of mitochondrial dysfunction and production of mitochondrial reactive oxygen species (mROS) in T cells and neutrophils. This can provoke numerous cellular changes including oxidation of nucleic acids, proteins, lipids and even induction of cell death. We have previously observed that in T cells from patients with lupus, the increased mROS is capable of provoking oligomerisation of mitochondrial antiviral stimulator (MAVS) and production of type I interferon (IFN-I). mROS in SLE neutrophils also promotes the formation of neutrophil extracellular traps (NETs), which are increased in lupus and implicated in renal damage. As a result, in addition to traditional immunosuppression, more comprehensive treatments for lupus may also include non-immune therapy, such as antioxidants. METHODS: Lupus-prone MRL-lpr mice were treated from weaning for 11 weeks with the mitochondria-targeted antioxidant, MitoQ (200 µM) in drinking water. Mice were then assessed for ROS production in neutrophils, NET formation, MAVS oligomerisation, serum IFN-I, autoantibody production and renal function. RESULTS: MitoQ-treated mice manifested reduced neutrophil ROS and NET formation, decreased MAVS oligomerisation and serum IFN-I, and reduced immune complex formation in kidneys, despite no change in serum autoantibody . CONCLUSIONS: These findings reveal the potential utility of targeting mROS in addition to traditional immunosuppressive therapy for lupus.


Subject(s)
Extracellular Traps/immunology , Kidney Diseases/metabolism , Lupus Erythematosus, Systemic/immunology , Mitochondria/metabolism , Organophosphorus Compounds/pharmacology , Ubiquinone/analogs & derivatives , Animals , Autoantibodies/metabolism , Disease Models, Animal , Female , Humans , Interferon Type I/immunology , Kidney/metabolism , Kidney/physiopathology , Kidney Diseases/physiopathology , Lupus Erythematosus, Systemic/physiopathology , Male , Mice , Mice, Inbred MRL lpr , Neutrophils/immunology , Oxidation-Reduction/drug effects , Oxidative Stress/immunology , Reactive Oxygen Species/metabolism , T-Lymphocytes/immunology , Ubiquinone/pharmacology
17.
Nat Commun ; 11(1): 3360, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32620763

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is considered the next major health epidemic with an estimated 25% worldwide prevalence. No drugs have yet been approved and NAFLD remains a major unmet need. Here, we identify MCJ (Methylation-Controlled J protein) as a target for non-alcoholic steatohepatitis (NASH), an advanced phase of NAFLD. MCJ is an endogenous negative regulator of the respiratory chain Complex I that acts to restrain mitochondrial respiration. We show that therapeutic targeting of MCJ in the liver with nanoparticle- and GalNAc-formulated siRNA efficiently reduces liver lipid accumulation and fibrosis in multiple NASH mouse models. Decreasing MCJ expression enhances the capacity of hepatocytes to mediate ß-oxidation of fatty acids and minimizes lipid accumulation, which results in reduced hepatocyte damage and fibrosis. Moreover, MCJ levels in the liver of NAFLD patients are elevated relative to healthy subjects. Thus, inhibition of MCJ emerges as an alternative approach to treat NAFLD.


Subject(s)
Fatty Acids/metabolism , HSP40 Heat-Shock Proteins/metabolism , Liver/pathology , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Molecular Chaperones/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Adult , Aged , Animals , Datasets as Topic , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , HSP40 Heat-Shock Proteins/antagonists & inhibitors , HSP40 Heat-Shock Proteins/genetics , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/cytology , Liver/drug effects , Male , Middle Aged , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/genetics , Nanoparticles/administration & dosage , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Oxidation-Reduction/drug effects , Primary Cell Culture , RNA, Small Interfering/administration & dosage , RNA-Seq
18.
Biol Reprod ; 81(3): 457-64, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19369644

ABSTRACT

Pregnancy induces dynamic changes in the maternal environment that include reversible modifications in response to systemic mediators and local signals. The spleen can be used to determine the effects of pregnancy on multiple cellular populations, including those of the erythroid lineage and the immune system. Current evidence suggests that the transient increase in the size of the spleen during pregnancy is due to the expansion of erythroid precursors. However, it is unclear what factors contribute to this increase. Moreover, the additional erythroid cells may compete with neighboring leukocytes for growth factors or space, and this may in turn alter the function of these populations. Therefore, we assessed proliferation and apoptosis throughout gestation using in vivo bromodeoxyuridine incorporation and the TUNEL assay, respectively. Here, we show that erythroid-lineage TER-119(+) cells expanded significantly in midgestation because of enhanced proliferation and diminished apoptosis. This correlated with increased expression of the erythropoietin receptor (Epor) and decreased expression of the death receptor Fas, respectively. Leukocytes demonstrated population-specific responses. Natural killer cells proliferated in early pregnancy. Both lymphocytes and CD11B(+) cells underwent enhanced proliferation during midgestation. In contrast, neutrophils exhibited augmented proliferation throughout pregnancy. These subset-specific alterations in proliferation and death in the spleen suggest that complex regulation of population dynamics exists during pregnancy.


Subject(s)
Apoptosis/physiology , Cell Proliferation , Erythroid Cells , Leukocytes/physiology , Pregnancy, Animal , Animals , Blood Cell Count , Blood Group Antigens/metabolism , Cell Lineage/physiology , Erythroid Cells/cytology , Erythroid Cells/metabolism , Female , Gestational Age , Leukocytes/cytology , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Organ Size , Pregnancy , Pregnancy, Animal/blood , Pregnancy, Animal/immunology , Pregnancy, Animal/physiology , Spleen/anatomy & histology
19.
Front Cell Dev Biol ; 7: 28, 2019.
Article in English | MEDLINE | ID: mdl-30915331

ABSTRACT

An effective adaptive immune response requires rapid T cell proliferation, followed by equally robust cell death. These two processes are coordinately regulated to allow sufficient magnitude of response followed by its rapid resolution, while also providing the maintenance of T cell memory. Both aspects of this T cell response are characterized by profound changes in metabolism; glycolysis drives proliferation whereas oxidative phosphorylation supports the survival of memory T cells. While much is known about the separate aspects of T cell expansion and contraction, considerably less is understood regarding how these processes might be connected. We report a link between the induction of glycolysis in CD8+ T cells and upregulation of the inhibitor of complex I and oxidative phosphorylation, methylation-controlled J protein (MCJ). MCJ acts synergistically with glycolysis to promote caspase-3 activity. Effector CD8+ T cells from MCJ-deficient mice manifest reduced glycolysis and considerably less active caspase-3 compared to wild-type cells. Consistent with these observations, in non-glycolytic CD8+ T cells cultured in the presence of IL-15, MCJ expression is repressed by methylation, which parallels their reduced active caspase-3 and increased survival compared to glycolytic IL-2-cultured T cells. Elevated levels of MCJ are also observed in vivo in the highly proliferative and glycolytic subset of CD4-CD8- T cells in Fas-deficient lpr mice. This subset also manifests elevated levels of activated caspase-3 and rapid cell death. Collectively, these data demonstrate tight linkage of glycolysis, MCJ expression, and active caspase-3 that serves to prevent the accumulation and promote the timely death of highly proliferative CD8+ T cells.

20.
Cell Death Dis ; 9(2): 62, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29352186

ABSTRACT

Resting T cells undergo a rapid metabolic shift to glycolysis upon activation in the presence of interleukin (IL)-2, in contrast to oxidative mitochondrial respiration with IL-15. Paralleling these different metabolic states are striking differences in susceptibility to restimulation-induced cell death (RICD); glycolytic effector T cells are highly sensitive to RICD, whereas non-glycolytic T cells are resistant. It is unclear whether the metabolic state of a T cell is linked to its susceptibility to RICD. Our findings reveal that IL-2-driven glycolysis promotes caspase-3 activity and increases sensitivity to RICD. Neither caspase-7, caspase-8, nor caspase-9 activity is affected by these metabolic differences. Inhibition of glycolysis with 2-deoxyglucose reduces caspase-3 activity as well as sensitivity to RICD. By contrast, IL-15-driven oxidative phosphorylation actively inhibits caspase-3 activity through its glutathionylation. We further observe active caspase-3 in the lipid rafts of glycolytic but not non-glycolytic T cells, suggesting a proximity-induced model of self-activation. Finally, we observe that effector T cells during influenza infection manifest higher levels of active caspase-3 than naive T cells. Collectively, our findings demonstrate that glycolysis drives caspase-3 activity and susceptibility to cell death in effector T cells independently of upstream caspases. Linking metabolism, caspase-3 activity, and cell death provides an intrinsic mechanism for T cells to limit the duration of effector function.


Subject(s)
Caspase 3/metabolism , Glycolysis/genetics , T-Lymphocytes/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL