Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 56(9): 2021-2035.e8, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37516105

ABSTRACT

Environmental nutrient availability influences T cell metabolism, impacting T cell function and shaping immune outcomes. Here, we identified ketone bodies (KBs)-including ß-hydroxybutyrate (ßOHB) and acetoacetate (AcAc)-as essential fuels supporting CD8+ T cell metabolism and effector function. ßOHB directly increased CD8+ T effector (Teff) cell cytokine production and cytolytic activity, and KB oxidation (ketolysis) was required for Teff cell responses to bacterial infection and tumor challenge. CD8+ Teff cells preferentially used KBs over glucose to fuel the tricarboxylic acid (TCA) cycle in vitro and in vivo. KBs directly boosted the respiratory capacity and TCA cycle-dependent metabolic pathways that fuel CD8+ T cell function. Mechanistically, ßOHB was a major substrate for acetyl-CoA production in CD8+ T cells and regulated effector responses through effects on histone acetylation. Together, our results identify cell-intrinsic ketolysis as a metabolic and epigenetic driver of optimal CD8+ T cell effector responses.


Subject(s)
CD8-Positive T-Lymphocytes , Histones , 3-Hydroxybutyric Acid/metabolism , 3-Hydroxybutyric Acid/pharmacology , Acetylation , Histones/metabolism , Ketone Bodies , Animals , Mice
2.
Nat Immunol ; 20(10): 1311-1321, 2019 10.
Article in English | MEDLINE | ID: mdl-31527833

ABSTRACT

Whether screening the metabolic activity of immune cells facilitates discovery of molecular pathology remains unknown. Here we prospectively screened the extracellular acidification rate as a measure of glycolysis and the oxygen consumption rate as a measure of mitochondrial respiration in B cells from patients with primary antibody deficiency. The highest oxygen consumption rate values were detected in three study participants with persistent polyclonal B cell lymphocytosis (PPBL). Exome sequencing identified germline mutations in SDHA, which encodes succinate dehydrogenase subunit A, in all three patients with PPBL. SDHA gain-of-function led to an accumulation of fumarate in PPBL B cells, which engaged the KEAP1-Nrf2 system to drive the transcription of genes encoding inflammatory cytokines. In a single patient trial, blocking the activity of the cytokine interleukin-6 in vivo prevented systemic inflammation and ameliorated clinical disease. Overall, our study has identified pathological mitochondrial retrograde signaling as a disease modifier in primary antibody deficiency.


Subject(s)
B-Lymphocytes/immunology , Electron Transport Complex II/genetics , Inflammation/metabolism , Lymphocytosis/immunology , Mitochondria/metabolism , Mutation/genetics , Anti-Inflammatory Agents/pharmacology , Cell Respiration , Cells, Cultured , Fumarates/metabolism , Glycolysis , Humans , Inflammation/genetics , Interleukin-6/antagonists & inhibitors , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Oxygen Consumption , Prospective Studies , Signal Transduction , Exome Sequencing
3.
Mol Cell ; 83(11): 1872-1886.e5, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37172591

ABSTRACT

Deregulated inflammation is a critical feature driving the progression of tumors harboring mutations in the liver kinase B1 (LKB1), yet the mechanisms linking LKB1 mutations to deregulated inflammation remain undefined. Here, we identify deregulated signaling by CREB-regulated transcription coactivator 2 (CRTC2) as an epigenetic driver of inflammatory potential downstream of LKB1 loss. We demonstrate that LKB1 mutations sensitize both transformed and non-transformed cells to diverse inflammatory stimuli, promoting heightened cytokine and chemokine production. LKB1 loss triggers elevated CRTC2-CREB signaling downstream of the salt-inducible kinases (SIKs), increasing inflammatory gene expression in LKB1-deficient cells. Mechanistically, CRTC2 cooperates with the histone acetyltransferases CBP/p300 to deposit histone acetylation marks associated with active transcription (i.e., H3K27ac) at inflammatory gene loci, promoting cytokine expression. Together, our data reveal a previously undefined anti-inflammatory program, regulated by LKB1 and reinforced through CRTC2-dependent histone modification signaling, that links metabolic and epigenetic states to cell-intrinsic inflammatory potential.


Subject(s)
Histones , Protein Serine-Threonine Kinases , Humans , Histones/genetics , Histones/metabolism , Acetylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Cytokines/metabolism , Inflammation/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Mol Cell ; 82(16): 2918-2921, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35985300

ABSTRACT

Zhang et al. (2022) show that TCR signaling promotes the phosphorylation and activation of glycogen phosphorylase B (PYGB) in CD8+ memory T (Tmem) cells. PYGB-dependent glycogen mobilization provides a carbon source to support glycolysis and early Tmem cell recall responses.


Subject(s)
Glycogen , Memory T Cells , Glycogen/metabolism , Glycolysis , Signal Transduction
5.
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
6.
Cell ; 153(6): 1239-51, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23746840

ABSTRACT

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


Subject(s)
Glycolysis , Lymphocyte Activation , Oxidative Phosphorylation , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , 3' Untranslated Regions , Animals , Cell Proliferation , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Interferon-gamma/genetics , Listeria monocytogenes , Listeriosis/immunology , Mice , Mice, Inbred C57BL , Protein Biosynthesis , T-Lymphocytes/immunology
7.
Cell ; 153(5): 1064-79, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706743

ABSTRACT

Metabolic adaptation is essential for cell survival during nutrient deprivation. We report that eukaryotic elongation factor 2 kinase (eEF2K), which is activated by AMP-kinase (AMPK), confers cell survival under acute nutrient depletion by blocking translation elongation. Tumor cells exploit this pathway to adapt to nutrient deprivation by reactivating the AMPK-eEF2K axis. Adaptation of transformed cells to nutrient withdrawal is severely compromised in cells lacking eEF2K. Moreover, eEF2K knockdown restored sensitivity to acute nutrient deprivation in highly resistant human tumor cell lines. In vivo, overexpression of eEF2K rendered murine tumors remarkably resistant to caloric restriction. Expression of eEF2K strongly correlated with overall survival in human medulloblastoma and glioblastoma multiforme. Finally, C. elegans strains deficient in efk-1, the eEF2K ortholog, were severely compromised in their response to nutrient depletion. Our data highlight a conserved role for eEF2K in protecting cells from nutrient deprivation and in conferring tumor cell adaptation to metabolic stress. PAPERCLIP:


Subject(s)
Caenorhabditis elegans/metabolism , Elongation Factor 2 Kinase/metabolism , Neoplasms/physiopathology , Peptide Chain Elongation, Translational , Signal Transduction , AMP-Activated Protein Kinases/metabolism , Animals , Brain Neoplasms/physiopathology , Caenorhabditis elegans/genetics , Cell Survival , Cell Transformation, Neoplastic , Elongation Factor 2 Kinase/genetics , Food Deprivation , Glioblastoma/physiopathology , HeLa Cells , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasm Transplantation , Peptide Elongation Factor 2/metabolism , Transplantation, Heterologous
8.
Mol Cell ; 75(2): 340-356.e10, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31253575

ABSTRACT

The microRNAs encoded by the miR-17∼92 polycistron are commonly overexpressed in cancer and orchestrate a wide range of oncogenic functions. Here, we identify a mechanism for miR-17∼92 oncogenic function through the disruption of endogenous microRNA (miRNA) processing. We show that, upon oncogenic overexpression of the miR-17∼92 primary transcript (pri-miR-17∼92), the microprocessor complex remains associated with partially processed intermediates that aberrantly accumulate. These intermediates reflect a series of hierarchical and conserved steps in the early processing of the pri-miR-17∼92 transcript. Encumbrance of the microprocessor by miR-17∼92 intermediates leads to the broad but selective downregulation of co-expressed polycistronic miRNAs, including miRNAs derived from tumor-suppressive miR-34b/c and from the Dlk1-Dio3 polycistrons. We propose that the identified steps of polycistronic miR-17∼92 biogenesis contribute to the oncogenic re-wiring of gene regulation networks. Our results reveal previously unappreciated functional paradigms for polycistronic miRNAs in cancer.


Subject(s)
Carcinogenesis/genetics , MicroRNAs/genetics , RNA Processing, Post-Transcriptional/genetics , Calcium-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Iodide Peroxidase/genetics , Membrane Proteins/genetics , MicroRNAs/biosynthesis , Nucleic Acid Conformation
9.
Nat Immunol ; 15(4): 323-32, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24562310

ABSTRACT

The ligation of Toll-like receptors (TLRs) leads to rapid activation of dendritic cells (DCs). However, the metabolic requirements that support this process remain poorly defined. We found that DC glycolytic flux increased within minutes of exposure to TLR agonists and that this served an essential role in supporting the de novo synthesis of fatty acids for the expansion of the endoplasmic reticulum and Golgi required for the production and secretion of proteins that are integral to DC activation. Signaling via the kinases TBK1, IKKɛ and Akt was essential for the TLR-induced increase in glycolysis by promoting the association of the glycolytic enzyme HK-II with mitochondria. In summary, we identified the rapid induction of glycolysis as an integral component of TLR signaling that is essential for the anabolic demands of the activation and function of DCs.


Subject(s)
Dendritic Cells/immunology , Glycolysis , I-kappa B Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/immunology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Fatty Acids/biosynthesis , Glycolysis/drug effects , Glycolysis/genetics , Glycolysis/immunology , Hexokinase/metabolism , I-kappa B Kinase/genetics , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptors/agonists
10.
Immunity ; 46(5): 730-742, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514674

ABSTRACT

Tissue-resident immune cells must balance survival in peripheral tissues with the capacity to respond rapidly upon infection or tissue damage, and in turn couple these responses with intrinsic metabolic control and conditions in the tissue microenvironment. The serine/threonine kinase mammalian/mechanistic target of rapamycin (mTOR) is a central integrator of extracellular and intracellular growth signals and cellular metabolism and plays important roles in both innate and adaptive immune responses. This review discusses the function of mTOR signaling in the differentiation and function of tissue-resident immune cells, with focus on the role of mTOR as a metabolic sensor and its impact on metabolic regulation in innate and adaptive immune cells. We also discuss the impact of metabolic constraints in tissues on immune homeostasis and disease, and how manipulating mTOR activity with drugs such as rapamycin can modulate immunity in these contexts.


Subject(s)
Immunity , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Adaptive Immunity , Animals , Energy Metabolism , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immunity, Innate , Organ Specificity/immunology
11.
Immunity ; 44(6): 1312-24, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27212436

ABSTRACT

How systemic metabolic alterations during acute infections impact immune cell function remains poorly understood. We found that acetate accumulates in the serum within hours of systemic bacterial infections and that these increased acetate concentrations are required for optimal memory CD8(+) T cell function in vitro and in vivo. Mechanistically, upon uptake by memory CD8(+) T cells, stress levels of acetate expanded the cellular acetyl-coenzyme A pool via ATP citrate lyase and promoted acetylation of the enzyme GAPDH. This context-dependent post-translational modification enhanced GAPDH activity, catalyzing glycolysis and thus boosting rapid memory CD8(+) T cell responses. Accordingly, in a murine Listeria monocytogenes model, transfer of acetate-augmented memory CD8(+) T cells exerted superior immune control compared to control cells. Our results demonstrate that increased systemic acetate concentrations are functionally integrated by CD8(+) T cells and translate into increased glycolytic and functional capacity. The immune system thus directly relates systemic metabolism with immune alertness.


Subject(s)
Acetates/metabolism , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Listeria monocytogenes/immunology , Listeriosis/immunology , ATP Citrate (pro-S)-Lyase/metabolism , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , CD8-Positive T-Lymphocytes/transplantation , Cells, Cultured , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating) , Glycolysis , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Processing, Post-Translational , Stress, Physiological/immunology
12.
Immunity ; 42(1): 41-54, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25607458

ABSTRACT

Naive T cells undergo metabolic reprogramming to support the increased energetic and biosynthetic demands of effector T cell function. However, how nutrient availability influences T cell metabolism and function remains poorly understood. Here we report plasticity in effector T cell metabolism in response to changing nutrient availability. Activated T cells were found to possess a glucose-sensitive metabolic checkpoint controlled by the energy sensor AMP-activated protein kinase (AMPK) that regulated mRNA translation and glutamine-dependent mitochondrial metabolism to maintain T cell bioenergetics and viability. T cells lacking AMPKα1 displayed reduced mitochondrial bioenergetics and cellular ATP in response to glucose limitation in vitro or pathogenic challenge in vivo. Finally, we demonstrated that AMPKα1 is essential for T helper 1 (Th1) and Th17 cell development and primary T cell responses to viral and bacterial infections in vivo. Our data highlight AMPK-dependent regulation of metabolic homeostasis as a key regulator of T cell-mediated adaptive immunity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/metabolism , AMP-Activated Protein Kinases/genetics , Adaptation, Physiological/immunology , Animals , Cells, Cultured , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Energy Metabolism , Glucose/metabolism , Glutamine/metabolism , Humans , Immunomodulation , Lymphocyte Activation/genetics , Metabolomics , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Protein Biosynthesis/genetics
13.
Immunity ; 40(5): 635-7, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24837098

ABSTRACT

Reprogramming cellular metabolism helps support T cell growth and effector function upon activation. In this issue of Immunity, Nakaya et al. (2014) report that the glutamine transporter ASCT2 regulates T cell metabolism and mTOR kinase signaling to shape inflammatory T helper cell responses.


Subject(s)
Amino Acid Transport System ASC/immunology , Glutamine/metabolism , Multiprotein Complexes/metabolism , Receptors, Antigen, T-Cell/immunology , TOR Serine-Threonine Kinases/metabolism , Animals , Humans , Mechanistic Target of Rapamycin Complex 1 , Minor Histocompatibility Antigens
14.
RNA Biol ; 20(1): 186-197, 2023 01.
Article in English | MEDLINE | ID: mdl-37095747

ABSTRACT

Here, we provide an in-depth analysis of the usefulness of single-sample metabolite/RNA extraction for multi-'omics readout. Using pulverized frozen livers of mice injected with lymphocytic choriomeningitis virus (LCMV) or vehicle (Veh), we isolated RNA prior (RNA) or following metabolite extraction (MetRNA). RNA sequencing (RNAseq) data were evaluated for differential expression analysis and dispersion, and differential metabolite abundance was determined. Both RNA and MetRNA clustered together by principal component analysis, indicating that inter-individual differences were the largest source of variance. Over 85% of LCMV versus Veh differentially expressed genes were shared between extraction methods, with the remaining 15% evenly and randomly divided between groups. Differentially expressed genes unique to the extraction method were attributed to randomness around the 0.05 FDR cut-off and stochastic changes in variance and mean expression. In addition, analysis using the mean absolute difference showed no difference in the dispersion of transcripts between extraction methods. Altogether, our data show that prior metabolite extraction preserves RNAseq data quality, which enables us to confidently perform integrated pathway enrichment analysis on metabolomics and RNAseq data from a single sample. This analysis revealed pyrimidine metabolism as the most LCMV-impacted pathway. Combined analysis of genes and metabolites in the pathway exposed a pattern in the degradation of pyrimidine nucleotides leading to uracil generation. In support of this, uracil was among the most differentially abundant metabolites in serum upon LCMV infection. Our data suggest that hepatic uracil export is a novel phenotypic feature of acute infection and highlight the usefulness of our integrated single-sample multi-'omics approach.


Subject(s)
Metabolomics , Virus Diseases , Animals , Mice , Sequence Analysis, RNA , Liver , RNA
15.
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
16.
Proc Natl Acad Sci U S A ; 116(18): 8975-8984, 2019 04 30.
Article in English | MEDLINE | ID: mdl-30988188

ABSTRACT

T cell help in humoral immunity includes interactions of B cells with activated extrafollicular CD4+ and follicular T helper (Tfh) cells. Each can promote antibody responses but Tfh cells play critical roles during germinal center (GC) reactions. After restimulation of their antigen receptor (TCR) by B cells, helper T cells act on B cells via CD40 ligand and secreted cytokines that guide Ig class switching. Hypoxia is a normal feature of GC, raising questions about molecular mechanisms governing the relationship between hypoxia response mechanisms and T cell help to antibody responses. Hypoxia-inducible factors (HIF) are prominent among mechanisms that mediate cellular responses to limited oxygen but also are induced by lymphocyte activation. We now show that loss of HIF-1α or of both HIF-1α and HIF-2α in CD4+ T cells compromised essential functions in help during antibody responses. HIF-1α depletion from CD4+ T cells reduced frequencies of antigen-specific GC B cells, Tfh cells, and overall antigen-specific Ab after immunization with sheep red blood cells. Compound deficiency of HIF-1α and HIF-2α led to humoral defects after hapten-carrier immunization. Further, HIF promoted CD40L expression while restraining the FoxP3-positive CD4+ cells in the CXCR5+ follicular regulatory population. Glycolysis increases T helper cytokine expression, and HIF promoted glycolysis in T helper cells via TCR or cytokine stimulation, as well as their production of cytokines that direct antibody class switching. Indeed, IFN-γ elaboration by HIF-deficient in vivo-generated Tfh cells was impaired. Collectively, the results indicate that HIF transcription factors are vital components of the mechanisms of help during humoral responses.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , Antibody Formation , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Hypoxia/immunology , Cell Hypoxia/physiology , Cytokines/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Humans , Hypoxia/metabolism , Immunity, Humoral , Immunization , Lymphocyte Activation/immunology , Lymphocyte Activation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CXCR5/metabolism , Sheep , T-Lymphocytes, Helper-Inducer/immunology
17.
Immunol Rev ; 283(1): 213-231, 2018 05.
Article in English | MEDLINE | ID: mdl-29664569

ABSTRACT

CD8+ T cells are key members of the adaptive immune response against infections and cancer. As we discuss in this review, these cells can present diverse metabolic requirements, which have been intensely studied during the past few years. Our current understanding suggests that aerobic glycolysis is a hallmark of activated CD8+ T cells, while naive and memory (Tmem ) cells often rely on oxidative phosphorylation, and thus mitochondrial metabolism is a crucial determinant of CD8+ Tmem cell development. Moreover, it has been proposed that CD8+ Tmem cells have a specific requirement for the oxidation of long-chain fatty acids (LC-FAO), a process modulated in lymphocytes by the enzyme CPT1A. However, this notion relies heavily on the metabolic analysis of in vitro cultures and on chemical inhibition of CPT1A. Therefore, we introduce more recent studies using genetic models to demonstrate that CPT1A-mediated LC-FAO is dispensable for the development of CD8+ T cell memory and protective immunity, and question the use of chemical inhibitors to target this enzyme. We discuss insights obtained from those and other studies analyzing the metabolic characteristics of CD8+ Tmem cells, and emphasize how T cells exhibit flexibility in their choice of metabolic fuel.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Fatty Acids/metabolism , Immunity, Cellular , Lipid Metabolism , Alcohol Oxidoreductases/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Energy Metabolism , Humans , Lymphocyte Activation/immunology , Mitochondria/metabolism , Signal Transduction
18.
Proc Natl Acad Sci U S A ; 115(10): E2202-E2209, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463754

ABSTRACT

The translation of mRNAs into proteins serves as a critical regulatory event in gene expression. In the context of cancer, deregulated translation is a hallmark of transformation, promoting the proliferation, survival, and metastatic capabilities of cancer cells. The best-studied factor involved in the translational control of cancer is the eukaryotic translation initiation factor 4E (eIF4E). We and others have shown that eIF4E availability and phosphorylation promote metastasis in mouse models of breast cancer by selectively augmenting the translation of mRNAs involved in invasion and metastasis. However, the impact of translational control in cell types within the tumor microenvironment (TME) is unknown. Here, we demonstrate that regulatory events affecting translation in cells of the TME impact cancer progression. Mice bearing a mutation in the phosphorylation site of eIF4E (S209A) in cells comprising the TME are resistant to the formation of lung metastases in a syngeneic mammary tumor model. This is associated with reduced survival of prometastatic neutrophils due to decreased expression of the antiapoptotic proteins BCL2 and MCL1. Furthermore, we demonstrate that pharmacological inhibition of eIF4E phosphorylation prevents metastatic progression in vivo, supporting the development of phosphorylation inhibitors for clinical use.


Subject(s)
Breast Neoplasms/pathology , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Neutrophils/metabolism , Protein Biosynthesis , Tumor Microenvironment , Amino Acid Motifs , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Eukaryotic Initiation Factor-4E/chemistry , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasm Metastasis , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Genes Dev ; 25(10): 1041-51, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21576264

ABSTRACT

Tumor cells gain a survival/growth advantage by adapting their metabolism to respond to environmental stress, a process known as metabolic transformation. The best-known aspect of metabolic transformation is the Warburg effect, whereby cancer cells up-regulate glycolysis under aerobic conditions. However, other mechanisms mediating metabolic transformation remain undefined. Here we report that carnitine palmitoyltransferase 1C (CPT1C), a brain-specific metabolic enzyme, may participate in metabolic transformation. CPT1C expression correlates inversely with mammalian target of rapamycin (mTOR) pathway activation, contributes to rapamycin resistance in murine primary tumors, and is frequently up-regulated in human lung tumors. Tumor cells constitutively expressing CPT1C show increased fatty acid (FA) oxidation, ATP production, and resistance to glucose deprivation or hypoxia. Conversely, cancer cells lacking CPT1C produce less ATP and are more sensitive to metabolic stress. CPT1C depletion via siRNA suppresses xenograft tumor growth and metformin responsiveness in vivo. CPT1C can be induced by hypoxia or glucose deprivation and is regulated by AMPKα. Cpt1c-deficient murine embryonic stem (ES) cells show sensitivity to hypoxia and glucose deprivation and altered FA homeostasis. Our results indicate that cells can use a novel mechanism involving CPT1C and FA metabolism to protect against metabolic stress. CPT1C may thus be a new therapeutic target for the treatment of hypoxic tumors.


Subject(s)
Carnitine O-Palmitoyltransferase/metabolism , Stress, Physiological/physiology , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/physiology , Carnitine O-Palmitoyltransferase/deficiency , Carnitine O-Palmitoyltransferase/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cells, Cultured , Drug Resistance, Neoplasm/genetics , Embryonic Stem Cells/enzymology , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Hypoxia/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , RNA, Messenger/metabolism , Reproducibility of Results , Stress, Physiological/genetics , TOR Serine-Threonine Kinases/metabolism , Transplantation, Heterologous , Up-Regulation
20.
PLoS Biol ; 13(12): e1002309, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26625127

ABSTRACT

Metformin is a biguanide widely prescribed to treat Type II diabetes that has gained interest as an antineoplastic agent. Recent work suggests that metformin directly antagonizes cancer cell growth through its actions on complex I of the mitochondrial electron transport chain (ETC). However, the mechanisms by which metformin arrests cancer cell proliferation remain poorly defined. Here we demonstrate that the metabolic checkpoint kinases AMP-activated protein kinase (AMPK) and LKB1 are not required for the antiproliferative effects of metformin. Rather, metformin inhibits cancer cell proliferation by suppressing mitochondrial-dependent biosynthetic activity. We show that in vitro metformin decreases the flow of glucose- and glutamine-derived metabolic intermediates into the Tricarboxylic Acid (TCA) cycle, leading to reduced citrate production and de novo lipid biosynthesis. Tumor cells lacking functional mitochondria maintain lipid biosynthesis in the presence of metformin via glutamine-dependent reductive carboxylation, and display reduced sensitivity to metformin-induced proliferative arrest. Our data indicate that metformin inhibits cancer cell proliferation by suppressing the production of mitochondrial-dependent metabolic intermediates required for cell growth, and that metabolic adaptations that bypass mitochondrial-dependent biosynthesis may provide a mechanism of tumor cell resistance to biguanide activity.


Subject(s)
Antineoplastic Agents/pharmacology , Citric Acid Cycle/drug effects , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Mitochondria/drug effects , Neoplasms/drug therapy , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Drug Resistance, Neoplasm , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Embryo, Mammalian/cytology , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Humans , Lipid Metabolism/drug effects , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL