Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Immunity ; 51(5): 856-870.e5, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31747582

ABSTRACT

Naive CD8+ T cells differentiating into effector T cells increase glucose uptake and shift from quiescent to anabolic metabolism. Although much is known about the metabolism of cultured T cells, how T cells use nutrients during immune responses in vivo is less well defined. Here, we combined bioenergetic profiling and 13C-glucose infusion techniques to investigate the metabolism of CD8+ T cells responding to Listeria infection. In contrast to in vitro-activated T cells, which display hallmarks of Warburg metabolism, physiologically activated CD8+ T cells displayed greater rates of oxidative metabolism, higher bioenergetic capacity, differential use of pyruvate, and prominent flow of 13C-glucose carbon to anabolic pathways, including nucleotide and serine biosynthesis. Glucose-dependent serine biosynthesis mediated by the enzyme Phgdh was essential for CD8+ T cell expansion in vivo. Our data highlight fundamental differences in glucose use by pathogen-specific T cells in vivo, illustrating the impact of environment on T cell metabolic phenotypes.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Energy Metabolism , Glucose/metabolism , Lymphocyte Activation/immunology , Metabolome , Metabolomics , Animals , Cell Proliferation , Gas Chromatography-Mass Spectrometry , Glycolysis , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Lymphocyte Activation/genetics , Metabolomics/methods , Mice , Oxidative Stress , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/metabolism , Virus Diseases/virology
2.
Nature ; 590(7846): 480-485, 2021 02.
Article in English | MEDLINE | ID: mdl-33597756

ABSTRACT

Obesity increases the risk of mortality because of metabolic sequelae such as type 2 diabetes and cardiovascular disease1. Thermogenesis by adipocytes can counteract obesity and metabolic diseases2,3. In thermogenic fat, creatine liberates a molar excess of mitochondrial ADP-purportedly via a phosphorylation cycle4-to drive thermogenic respiration. However, the proteins that control this futile creatine cycle are unknown. Here we show that creatine kinase B (CKB) is indispensable for thermogenesis resulting from the futile creatine cycle, during which it traffics to mitochondria using an internal mitochondrial targeting sequence. CKB is powerfully induced by thermogenic stimuli in both mouse and human adipocytes. Adipocyte-selective inactivation of Ckb in mice diminishes thermogenic capacity, increases predisposition to obesity, and disrupts glucose homeostasis. CKB is therefore a key effector of the futile creatine cycle.


Subject(s)
Adipose Tissue/metabolism , Creatine Kinase, BB Form/metabolism , Creatine/metabolism , Thermogenesis , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/enzymology , Animals , Creatine Kinase, BB Form/deficiency , Creatine Kinase, BB Form/genetics , Cyclic AMP/metabolism , Energy Metabolism/genetics , Female , Glucose/metabolism , Homeostasis , Humans , Male , Mice , Mitochondria/metabolism , Obesity/enzymology , Obesity/genetics , Obesity/metabolism , Signal Transduction
3.
Mol Cell ; 60(2): 195-207, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26474064

ABSTRACT

Cancer cells adapt metabolically to proliferate under nutrient limitation. Here we used combined transcriptional-metabolomic network analysis to identify metabolic pathways that support glucose-independent tumor cell proliferation. We found that glucose deprivation stimulated re-wiring of the tricarboxylic acid (TCA) cycle and early steps of gluconeogenesis to promote glucose-independent cell proliferation. Glucose limitation promoted the production of phosphoenolpyruvate (PEP) from glutamine via the activity of mitochondrial PEP-carboxykinase (PCK2). Under these conditions, glutamine-derived PEP was used to fuel biosynthetic pathways normally sustained by glucose, including serine and purine biosynthesis. PCK2 expression was required to maintain tumor cell proliferation under limited-glucose conditions in vitro and tumor growth in vivo. Elevated PCK2 expression is observed in several human tumor types and enriched in tumor tissue from non-small-cell lung cancer (NSCLC) patients. Our results define a role for PCK2 in cancer cell metabolic reprogramming that promotes glucose-independent cell growth and metabolic stress resistance in human tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Gene Expression Regulation, Neoplastic , Gluconeogenesis/genetics , Lung Neoplasms/metabolism , Neoplasms/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Adaptation, Physiological/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Citric Acid Cycle/genetics , Glucose/deficiency , Glutamine/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Metabolomics , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/pathology , Phosphoenolpyruvate/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Purines/biosynthesis , Pyruvic Acid/metabolism , Serine/biosynthesis
4.
Proc Natl Acad Sci U S A ; 111(7): 2554-9, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24550282

ABSTRACT

One of the major metabolic changes associated with cellular transformation is enhanced nutrient utilization, which supports tumor progression by fueling both energy production and providing biosynthetic intermediates for growth. The liver kinase B1 (LKB1) is a serine/threonine kinase and tumor suppressor that couples bioenergetics to cell-growth control through regulation of mammalian target of rapamycin (mTOR) activity; however, the influence of LKB1 on tumor metabolism is not well defined. Here, we show that loss of LKB1 induces a progrowth metabolic program in proliferating cells. Cells lacking LKB1 display increased glucose and glutamine uptake and utilization, which support both cellular ATP levels and increased macromolecular biosynthesis. This LKB1-dependent reprogramming of cell metabolism is dependent on the hypoxia-inducible factor-1α (HIF-1α), which accumulates under normoxia in LKB1-deficient cells and is antagonized by inhibition of mTOR complex I signaling. Silencing HIF-1α reverses the metabolic advantages conferred by reduced LKB1 signaling and impairs the growth and survival of LKB1-deficient tumor cells under low-nutrient conditions. Together, our data implicate the tumor suppressor LKB1 as a central regulator of tumor metabolism and growth control through the regulation of HIF-1α-dependent metabolic reprogramming.


Subject(s)
Energy Metabolism/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Metabolic Networks and Pathways/genetics , Protein Serine-Threonine Kinases/deficiency , AMP-Activated Protein Kinase Kinases , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Fibroblasts , Gas Chromatography-Mass Spectrometry , Glucose/metabolism , Glutamine/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Metabolic Networks and Pathways/physiology , Mice , Multiprotein Complexes/metabolism , Oxygen Consumption/physiology , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism
5.
J Am Soc Nephrol ; 27(8): 2422-35, 2016 08.
Article in English | MEDLINE | ID: mdl-26802179

ABSTRACT

Podocytes are specialized epithelial cells of the kidney blood filtration barrier that contribute to permselectivity via a series of interdigitating actin-rich foot processes. Positioned between adjacent projections is a unique cell junction known as the slit diaphragm, which is physically connected to the actin cytoskeleton via the transmembrane protein nephrin. Evidence indicates that tyrosine phosphorylation of the intracellular tail of nephrin initiates signaling events, including recruitment of cytoplasmic adaptor proteins Nck1 and Nck2 that regulate actin cytoskeletal dynamics. Nephrin tyrosine phosphorylation is altered in human and experimental renal diseases characterized by pathologic foot process remodeling, prompting the hypothesis that phosphonephrin signaling directly influences podocyte morphology. To explore this possibility, we generated and analyzed knockin mice with mutations that disrupt nephrin tyrosine phosphorylation and Nck1/2 binding (nephrin(Y3F/Y3F) mice). Homozygous nephrin(Y3F/Y3F) mice developed progressive proteinuria accompanied by structural changes in the filtration barrier, including podocyte foot process effacement, irregular thickening of the glomerular basement membrane, and dilated capillary loops, with a similar but later onset phenotype in heterozygous animals. Furthermore, compared with wild-type mice, nephrin(Y3F/Y3F) mice displayed delayed recovery in podocyte injury models. Profiling of nephrin tyrosine phosphorylation dynamics in wild-type mice subjected to podocyte injury indicated site-specific differences in phosphorylation at baseline, injury, and recovery, which correlated with loss of nephrin-Nck1/2 association during foot process effacement. Our results define an essential requirement for nephrin tyrosine phosphorylation in stabilizing podocyte morphology and suggest a model in which dynamic changes in phosphotyrosine-based signaling confer plasticity to the podocyte actin cytoskeleton.


Subject(s)
Podocytes/physiology , Podocytes/ultrastructure , Tyrosine/metabolism , Animals , Female , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Phosphorylation , Signal Transduction
6.
Photosynth Res ; 121(2-3): 125-33, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24504539

ABSTRACT

CcmL is a small, pentameric protein that is argued to fill the vertices of ß-carboxysomal shell. Here we report the structures of two CcmL orthologs, those from Nostoc sp. PCC 7120 and Thermosynechococcus elongatus BP-1. These structures broadly resemble those previously reported for other strains. However, the Nostoc CcmL structure shows an interesting pattern of behavior where two loops that map to the base of the pentamer adopt either an out or in conformation, with a consistent (over six pentamers) out-in-out-in-in pattern of protomers. The pentamers in this structure are also consistently organized into a back-to-back decamer, though evidence suggests that this is likely not present in solution. Förster resonance energy transfer experiments were able to show a weak interaction between CcmL and CcmK2 when CcmK2 was present at >100 µM. Since CcmK2 forms defined bodies with approximately 200 nm diameter at this concentration, this would support the idea that CcmL can only interact with CcmK2 at rare defect points in the growing shell.


Subject(s)
Cyanobacteria/metabolism , Fluorescence Resonance Energy Transfer , Models, Molecular , Nostoc/metabolism
7.
Cell Metab ; 36(3): 526-540.e7, 2024 03 05.
Article in English | MEDLINE | ID: mdl-38272036

ABSTRACT

That uncoupling protein 1 (UCP1) is the sole mediator of adipocyte thermogenesis is a conventional viewpoint that has primarily been inferred from the attenuation of the thermogenic output of mice genetically lacking Ucp1 from birth (germline Ucp1-/-). However, germline Ucp1-/- mice harbor secondary changes within brown adipose tissue. To mitigate these potentially confounding ancillary changes, we constructed mice with inducible adipocyte-selective Ucp1 disruption. We find that, although germline Ucp1-/- mice succumb to cold-induced hypothermia with complete penetrance, most mice with the inducible deletion of Ucp1 maintain homeothermy in the cold. However, inducible adipocyte-selective co-deletion of Ucp1 and creatine kinase b (Ckb, an effector of UCP1-independent thermogenesis) exacerbates cold intolerance. Following UCP1 deletion or UCP1/CKB co-deletion from mature adipocytes, moderate cold exposure triggers the regeneration of mature brown adipocytes that coordinately restore UCP1 and CKB expression. Our findings suggest that thermogenic adipocytes utilize non-paralogous protein redundancy-through UCP1 and CKB-to promote cold-induced energy dissipation.


Subject(s)
Adipocytes, Brown , Adipose Tissue, Brown , Animals , Mice , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Thermogenesis , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Creatine Kinase, BB Form/metabolism
8.
Sci Adv ; 10(22): eadj1431, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38809979

ABSTRACT

Infusion of 13C-labeled metabolites provides a gold standard for understanding the metabolic processes used by T cells during immune responses in vivo. Through infusion of 13C-labeled metabolites (glucose, glutamine, and acetate) in Listeria monocytogenes-infected mice, we demonstrate that CD8 T effector (Teff) cells use metabolites for specific pathways during specific phases of activation. Highly proliferative early Teff cells in vivo shunt glucose primarily toward nucleotide synthesis and leverage glutamine anaplerosis in the tricarboxylic acid (TCA) cycle to support adenosine triphosphate and de novo pyrimidine synthesis. In addition, early Teff cells rely on glutamic-oxaloacetic transaminase 1 (Got1)-which regulates de novo aspartate synthesis-for effector cell expansion in vivo. CD8 Teff cells change fuel preference over the course of infection, switching from glutamine- to acetate-dependent TCA cycle metabolism late in infection. This study provides insights into the dynamics of Teff metabolism, illuminating distinct pathways of fuel consumption associated with CD8 Teff cell function in vivo.


Subject(s)
Acetates , CD8-Positive T-Lymphocytes , Carbon Isotopes , Glutamine , Glutamine/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Acetates/metabolism , Mice , Listeriosis/metabolism , Listeriosis/immunology , Listeriosis/microbiology , Listeria monocytogenes , Citric Acid Cycle , Glucose/metabolism , Mice, Inbred C57BL
9.
J Neurosci Res ; 91(3): 349-62, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23184356

ABSTRACT

The classic myelin basic protein (MBP) family of central nervous system (CNS) myelin arises from transcription start site 3 of the Golli (gene of oligodendrocyte lineage) complex and comprises splice isoforms ranging in nominal molecular mass from 14 kDa to (full-length) 21.5 kDa. We have determined here a number of distinct functional differences between the major 18.5-kDa and minor 21.5-kDa isoforms of classic MBP with respect to oligodendrocyte (OLG) proliferation. We have found that, in contrast to 18.5-kDa MBP, 21.5-kDa MBP increases proliferation of early developmental immortalized N19-OLGs by elevating the levels of phosphorylated ERK1/2 and Akt1 kinases and of ribosomal protein S6. Coculture of N2a neuronal cells with N19-OLGs transfected with the 21.5-kDa isoform (or conditioned medium from), but not the 18.5-kDa isoform, caused the N2a cells to have increased neurite outgrowth and process branching complexity. These roles were dependent on subcellular localization of 21.5-kDa MBP to the nucleus and on the exon II-encoded segment, suggesting that the nuclear localization of early minor isoforms of MBP may play a crucial role in regulating and/or initiating myelin and neuronal development in the mammalian CNS.


Subject(s)
Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Myelin Basic Protein/physiology , Neurites/physiology , Oligodendroglia/metabolism , Animals , Cell Line, Transformed , Cell Membrane/chemistry , Cell Nucleus/chemistry , Cell Nucleus/physiology , Coculture Techniques , Mice , Molecular Weight , Myelin Basic Protein/chemistry , Myelin Basic Protein/metabolism , Neurites/chemistry , Oligodendroglia/chemistry , Protein Isoforms/physiology
10.
bioRxiv ; 2023 Jun 11.
Article in English | MEDLINE | ID: mdl-37333111

ABSTRACT

Infusion of 13C-labeled metabolites provides a gold-standard for understanding the metabolic processes used by T cells during immune responses in vivo. Through infusion of 13C-labeled metabolites (glucose, glutamine, acetate) in Listeria monocytogenes (Lm)-infected mice, we demonstrate that CD8+ T effector (Teff) cells utilize metabolites for specific pathways during specific phases of activation. Highly proliferative early Teff cells in vivo shunt glucose primarily towards nucleotide synthesis and leverage glutamine anaplerosis in the tricarboxylic acid (TCA) cycle to support ATP and de novo pyrimidine synthesis. Additionally, early Teff cells rely on glutamic-oxaloacetic transaminase 1 (Got1)-which regulates de novo aspartate synthesis-for effector cell expansion in vivo. Importantly, Teff cells change fuel preference over the course of infection, switching from glutamine- to acetate-dependent TCA cycle metabolism late in infection. This study provides insights into the dynamics of Teff metabolism, illuminating distinct pathways of fuel consumption associated with Teff cell function in vivo.

11.
Cell Rep ; 38(9): 110446, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35235777

ABSTRACT

The factors that promote T cell expansion are not fully known. Creatine is an abundant circulating metabolite that has recently been implicated in T cell function; however, its cell-autonomous role in immune-cell function is unknown. Here, we show that creatine supports cell-intrinsic CD8+ T cell homeostasis. We further identify creatine kinase B (CKB) as the creatine kinase isoenzyme that supports these T cell properties. Loss of the creatine transporter (Slc6a8) or Ckb results in compromised CD8+ T cell expansion in response to infection without influencing adenylate energy charge. Rather, loss of Slc6a8 or Ckb disrupts naive T cell homeostasis and weakens TCR-mediated activation of mechanistic target of rapamycin complex 1 (mTORC1) signaling required for CD8+ T cell expansion. These data demonstrate a cell-intrinsic role for creatine transport and creatine transphosphorylation, independent of their effects on global cellular energy charge, in supporting CD8+ T cell homeostasis and effector function.


Subject(s)
CD8-Positive T-Lymphocytes , Creatine , Creatine/metabolism , Creatine Kinase/metabolism , Phosphorylation , Signal Transduction
12.
Nat Metab ; 4(11): 1459-1473, 2022 11.
Article in English | MEDLINE | ID: mdl-36344764

ABSTRACT

Noradrenaline (NA) regulates cold-stimulated adipocyte thermogenesis1. Aside from cAMP signalling downstream of ß-adrenergic receptor activation, how NA promotes thermogenic output is still not fully understood. Here, we show that coordinated α1-adrenergic receptor (AR) and ß3-AR signalling induces the expression of thermogenic genes of the futile creatine cycle2,3, and that early B cell factors, oestrogen-related receptors and PGC1α are required for this response in vivo. NA triggers physical and functional coupling between the α1-AR subtype (ADRA1A) and Gαq to promote adipocyte thermogenesis in a manner that is dependent on the effector proteins of the futile creatine cycle, creatine kinase B and tissue-non-specific alkaline phosphatase. Combined Gαq and Gαs signalling selectively in adipocytes promotes a continual rise in whole-body energy expenditure, and creatine kinase B is required for this effect. Thus, the ADRA1A-Gαq-futile creatine cycle axis is a key regulator of facultative and adaptive thermogenesis.


Subject(s)
Creatine , Thermogenesis , Creatine/metabolism , Thermogenesis/genetics , Adipocytes/metabolism , Energy Metabolism/genetics , Creatine Kinase/metabolism
13.
Nat Cancer ; 2(5): 545-562, 2021 05.
Article in English | MEDLINE | ID: mdl-35122017

ABSTRACT

Metastasis is the leading cause of cancer-related deaths, and obesity is associated with increased breast cancer (BC) metastasis. Preclinical studies have shown that obese adipose tissue induces lung neutrophilia associated with enhanced BC metastasis to this site. Here we show that obesity leads to neutrophil-dependent impairment of vascular integrity through loss of endothelial adhesions, enabling cancer cell extravasation into the lung. Mechanistically, neutrophil-produced reactive oxygen species in obese mice increase neutrophil extracellular DNA traps (NETs) and weaken endothelial junctions, facilitating the influx of tumor cells from the peripheral circulation. In vivo treatment with catalase, NET inhibitors or genetic deletion of Nos2 reversed this effect in preclinical models of obesity. Imaging mass cytometry of lung metastasis samples from patients with cancer revealed an enrichment in neutrophils with low catalase levels correlating with elevated body mass index. Our data provide insights into potentially targetable mechanisms that underlie the progression of BC in the obese population.


Subject(s)
Breast Neoplasms , Lung Neoplasms , Animals , Breast Neoplasms/metabolism , Catalase/metabolism , Female , Humans , Lung Neoplasms/metabolism , Mice , Neutrophils/metabolism , Obesity/complications , Oxidative Stress
14.
Cell Metab ; 33(3): 499-512.e6, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33596409

ABSTRACT

Obesity is a major risk factor for adverse outcomes in breast cancer; however, the underlying molecular mechanisms have not been elucidated. To investigate the role of crosstalk between mammary adipocytes and neoplastic cells in the tumor microenvironment (TME), we performed transcriptomic analysis of cancer cells and adjacent adipose tissue in a murine model of obesity-accelerated breast cancer and identified glycine amidinotransferase (Gatm) in adipocytes and Acsbg1 in cancer cells as required for obesity-driven tumor progression. Gatm is the rate-limiting enzyme in creatine biosynthesis, and deletion in adipocytes attenuated obesity-driven tumor growth. Similarly, genetic inhibition of creatine import into cancer cells reduced tumor growth in obesity. In parallel, breast cancer cells in obese animals upregulated the fatty acyl-CoA synthetase Acsbg1 to promote creatine-dependent tumor progression. These findings reveal key nodes in the crosstalk between adipocytes and cancer cells in the TME necessary for obesity-driven breast cancer progression.


Subject(s)
Breast Neoplasms/pathology , Cell Communication/physiology , Creatine/metabolism , Obesity/pathology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Amidinotransferases/deficiency , Amidinotransferases/genetics , Amidinotransferases/metabolism , Animals , Cell Line, Tumor , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Diet, High-Fat , Female , Humans , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Interference , RNA, Small Interfering/metabolism , Tumor Microenvironment
15.
Cell Metab ; 31(2): 250-266.e9, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32023446

ABSTRACT

Epigenetic modifications on DNA and histones regulate gene expression by modulating chromatin accessibility to transcription machinery. Here we identify methionine as a key nutrient affecting epigenetic reprogramming in CD4+ T helper (Th) cells. Using metabolomics, we showed that methionine is rapidly taken up by activated T cells and serves as the major substrate for biosynthesis of the universal methyl donor S-adenosyl-L-methionine (SAM). Methionine was required to maintain intracellular SAM pools in T cells. Methionine restriction reduced histone H3K4 methylation (H3K4me3) at the promoter regions of key genes involved in Th17 cell proliferation and cytokine production. Applied to the mouse model of multiple sclerosis (experimental autoimmune encephalomyelitis), dietary methionine restriction reduced the expansion of pathogenic Th17 cells in vivo, leading to reduced T cell-mediated neuroinflammation and disease onset. Our data identify methionine as a key nutritional factor shaping Th cell proliferation and function in part through regulation of histone methylation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Epigenesis, Genetic/drug effects , Histones/metabolism , Methionine , Multiple Sclerosis , Th17 Cells/metabolism , Animals , Cell Proliferation , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , HEK293 Cells , Humans , Methionine/metabolism , Methionine/pharmacology , Methylation , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Th17 Cells/cytology
16.
Cell Metab ; 28(3): 504-515.e7, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30043753

ABSTRACT

T cell subsets including effector (Teff), regulatory (Treg), and memory (Tmem) cells are characterized by distinct metabolic profiles that influence their differentiation and function. Previous research suggests that engagement of long-chain fatty acid oxidation (LC-FAO) supports Foxp3+ Treg cell and Tmem cell survival. However, evidence for this is mostly based on inhibition of Cpt1a, the rate-limiting enzyme for LC-FAO, with the drug etomoxir. Using genetic models to target Cpt1a specifically in T cells, we dissected the role of LC-FAO in primary, memory, and regulatory T cell responses. Here we show that the ACC2/Cpt1a axis is largely dispensable for Teff, Tmem, or Treg cell formation, and that the effects of etomoxir on T cell differentiation and function are independent of Cpt1a expression. Together our data argue that metabolic pathways other than LC-FAO fuel Tmem or Treg differentiation and suggest alternative mechanisms for the effects of etomoxir that involve mitochondrial respiration.


Subject(s)
Acetyl-CoA Carboxylase/physiology , CD8-Positive T-Lymphocytes/metabolism , Carnitine O-Palmitoyltransferase/physiology , Epoxy Compounds/pharmacology , Fatty Acids/metabolism , Immunologic Memory/drug effects , Mitochondria/metabolism , T-Lymphocytes, Regulatory/drug effects , Acetyl-CoA Carboxylase/genetics , Animals , Carnitine O-Palmitoyltransferase/genetics , Cell Differentiation/drug effects , Cells, Cultured , Child , Child, Preschool , Female , Gene Knockout Techniques , Humans , Lymphocyte Activation/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction/drug effects , Oxidative Phosphorylation/drug effects , T-Lymphocytes, Regulatory/metabolism
17.
Cell Rep ; 21(1): 1-9, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978464

ABSTRACT

Reactive oxygen species (ROS) are continuously produced as a by-product of mitochondrial metabolism and eliminated via antioxidant systems. Regulation of mitochondrially produced ROS is required for proper cellular function, adaptation to metabolic stress, and bypassing cellular senescence. Here, we report non-canonical regulation of the cellular energy sensor AMP-activated protein kinase (AMPK) by mitochondrial ROS (mROS) that functions to maintain cellular metabolic homeostasis. We demonstrate that mitochondrial ROS are a physiological activator of AMPK and that AMPK activation triggers a PGC-1α-dependent antioxidant response that limits mitochondrial ROS production. Cells lacking AMPK activity display increased mitochondrial ROS levels and undergo premature senescence. Finally, we show that AMPK-PGC-1α-dependent control of mitochondrial ROS regulates HIF-1α stabilization and that mitochondrial ROS promote the Warburg effect in cells lacking AMPK signaling. These data highlight a key function for AMPK in sensing and resolving mitochondrial ROS for stress resistance and maintaining cellular metabolic balance.


Subject(s)
AMP-Activated Protein Kinases/genetics , Homeostasis/genetics , Metabolic Networks and Pathways/genetics , Mitochondria/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Reactive Oxygen Species/metabolism , AMP-Activated Protein Kinases/deficiency , Animals , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Cellular Senescence/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Transgenic , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/deficiency , Primary Cell Culture , Protein Stability , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Uncoupling Protein 2/genetics , Uncoupling Protein 2/metabolism , Uncoupling Protein 3/genetics , Uncoupling Protein 3/metabolism , Glutathione Peroxidase GPX1
18.
Cell Metab ; 25(2): 345-357, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28111214

ABSTRACT

During immune challenge, T lymphocytes engage pathways of anabolic metabolism to support clonal expansion and the development of effector functions. Here we report a critical role for the non-essential amino acid serine in effector T cell responses. Upon activation, T cells upregulate enzymes of the serine, glycine, one-carbon (SGOC) metabolic network, and rapidly increase processing of serine into one-carbon metabolism. We show that extracellular serine is required for optimal T cell expansion even in glucose concentrations sufficient to support T cell activation, bioenergetics, and effector function. Restricting dietary serine impairs pathogen-driven expansion of T cells in vivo, without affecting overall immune cell homeostasis. Mechanistically, serine supplies glycine and one-carbon units for de novo nucleotide biosynthesis in proliferating T cells, and one-carbon units from formate can rescue T cells from serine deprivation. Our data implicate serine as a key immunometabolite that directly modulates adaptive immunity by controlling T cell proliferative capacity.


Subject(s)
Metabolome , Serine/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Carbon/metabolism , Cell Cycle Checkpoints , Cell Proliferation , Diet , Energy Metabolism , Extracellular Space/metabolism , Glycine , Listeria monocytogenes/immunology , Metabolic Networks and Pathways , Mice, Inbred C57BL , Purine Nucleotides/biosynthesis
19.
Cell Rep ; 16(7): 1915-28, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27498867

ABSTRACT

A central hallmark of cancer cells is the reprogramming of cellular metabolism to meet the bioenergetic and biosynthetic demands of malignant growth. Here, we report that the miR-17∼92 microRNA (miRNA) cluster is an oncogenic driver of tumor metabolic reprogramming. Loss of miR-17∼92 in Myc(+) tumor cells leads to a global decrease in tumor cell metabolism, affecting both glycolytic and mitochondrial metabolism, whereas increased miR-17∼92 expression is sufficient to drive increased nutrient usage by tumor cells. We mapped the metabolic control element of miR-17∼92 to the miR-17 seed family, which influences cellular metabolism and mammalian target of rapamycin complex 1 (mTORC1) signaling through negative regulation of the LKB1 tumor suppressor. miR-17-dependent tuning of LKB1 levels regulates both the metabolic potential of Myc(+) lymphomas and tumor growth in vivo. Our results establish metabolic reprogramming as a central function of the oncogenic miR-17∼92 miRNA cluster that drives the progression of MYC-dependent tumors.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Lymphocytes/metabolism , Lymphoma/metabolism , MicroRNAs/genetics , AMP-Activated Protein Kinase Kinases , Animals , Base Sequence , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Glycolysis/genetics , Heterografts , Humans , Lymphocyte Transfusion , Lymphocytes/pathology , Lymphoma/genetics , Lymphoma/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , MicroRNAs/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Oxidative Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL