Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25.272
Filter
Add more filters

Publication year range
1.
Cell ; 187(9): 2124-2126, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38670069

ABSTRACT

Many types of tumor cells alter metabolic pathways to meet their energy and biosynthetic demands for proliferation or stress adaptation. In this issue of Cell, Kong et al. find that the glycolytic metabolite methylglyoxal causes cancer-associated mutant single-base substitution features by inducing BRCA2 proteolysis, leading to functional haploinsufficiency of BRCA2.


Subject(s)
BRCA2 Protein , Glycolysis , Haploinsufficiency , Humans , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , Pyruvaldehyde/metabolism , Mutation
2.
Cell ; 187(9): 2269-2287.e16, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38608703

ABSTRACT

Knudson's "two-hit" paradigm posits that carcinogenesis requires inactivation of both copies of an autosomal tumor suppressor gene. Here, we report that the glycolytic metabolite methylglyoxal (MGO) transiently bypasses Knudson's paradigm by inactivating the breast cancer suppressor protein BRCA2 to elicit a cancer-associated, mutational single-base substitution (SBS) signature in nonmalignant mammary cells or patient-derived organoids. Germline monoallelic BRCA2 mutations predispose to these changes. An analogous SBS signature, again without biallelic BRCA2 inactivation, accompanies MGO accumulation and DNA damage in Kras-driven, Brca2-mutant murine pancreatic cancers and human breast cancers. MGO triggers BRCA2 proteolysis, temporarily disabling BRCA2's tumor suppressive functions in DNA repair and replication, causing functional haploinsufficiency. Intermittent MGO exposure incites episodic SBS mutations without permanent BRCA2 inactivation. Thus, a metabolic mechanism wherein MGO-induced BRCA2 haploinsufficiency transiently bypasses Knudson's two-hit requirement could link glycolysis activation by oncogenes, metabolic disorders, or dietary challenges to mutational signatures implicated in cancer evolution.


Subject(s)
BRCA2 Protein , Breast Neoplasms , Glycolysis , Pyruvaldehyde , Animals , BRCA2 Protein/metabolism , BRCA2 Protein/genetics , Mice , Humans , Female , Pyruvaldehyde/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Haploinsufficiency , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Mutation , DNA Damage , DNA Repair , Cell Line, Tumor
3.
Cell ; 186(4): 748-763.e15, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36758548

ABSTRACT

Although many prokaryotes have glycolysis alternatives, it's considered as the only energy-generating glucose catabolic pathway in eukaryotes. Here, we managed to create a hybrid-glycolysis yeast. Subsequently, we identified an inositol pyrophosphatase encoded by OCA5 that could regulate glycolysis and respiration by adjusting 5-diphosphoinositol 1,2,3,4,6-pentakisphosphate (5-InsP7) levels. 5-InsP7 levels could regulate the expression of genes involved in glycolysis and respiration, representing a global mechanism that could sense ATP levels and regulate central carbon metabolism. The hybrid-glycolysis yeast did not produce ethanol during growth under excess glucose and could produce 2.68 g/L free fatty acids, which is the highest reported production in shake flask of Saccharomyces cerevisiae. This study demonstrated the significance of hybrid-glycolysis yeast and determined Oca5 as an inositol pyrophosphatase controlling the balance between glycolysis and respiration, which may shed light on the role of inositol pyrophosphates in regulating eukaryotic metabolism.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Diphosphates/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Inositol Phosphates/genetics , Inositol Phosphates/metabolism , Glycolysis/genetics , Respiration , Pyrophosphatases/metabolism , Glucose/metabolism
4.
Annu Rev Biochem ; 90: 31-55, 2021 06 20.
Article in English | MEDLINE | ID: mdl-34153217

ABSTRACT

My graduate and postdoctoral training in metabolism and enzymology eventually led me to study the short- and long-term regulation of glucose and lipid metabolism. In the early phase of my career, my trainees and I identified, purified, and characterized a variety of phosphofructokinase enzymes from mammalian tissues. These studies led us to discover fructose 2,6-P2, the most potent activator of phosphofructokinase and glycolysis. The discovery of fructose 2,6-P2 led to the identification and characterization of the tissue-specific bifunctional enzyme 6-phosphofructo-2-kinase:fructose 2,6-bisphosphatase. We discovered a glucose signaling mechanism by which the liver maintains glucose homeostasis by regulating the activities of this bifunctional enzyme. With a rise in glucose, a signaling metabolite, xylulose 5-phosphate, triggers rapid activation of a specific protein phosphatase (PP2ABδC), which dephosphorylates the bifunctional enzyme, thereby increasing fructose 2,6-P2 levels and upregulating glycolysis. These endeavors paved the way for us to initiate the later phase of my career in which we discovered a new transcription factor termed the carbohydrate response element binding protein (ChREBP). Now ChREBP is recognized as the masterregulator controlling conversion of excess carbohydrates to storage of fat in the liver. ChREBP functions as a central metabolic coordinator that responds to nutrients independently of insulin. The ChREBP transcription factor facilitates metabolic adaptation to excess glucose, leading to obesity and its associated diseases.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Biochemistry/history , Fructosediphosphates/metabolism , Phosphofructokinase-2/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gluconeogenesis/physiology , Glucose/metabolism , Glycolysis , History, 20th Century , History, 21st Century , Humans , Male , Mice , Phosphofructokinase-2/chemistry , Phosphofructokinases/chemistry , Phosphofructokinases/metabolism , Phosphorylation , United States
5.
Cell ; 184(15): 3899-3914.e16, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34237254

ABSTRACT

The impact of the microbiome on HIV disease is widely acknowledged although the mechanisms downstream of fluctuations in microbial composition remain speculative. We detected rapid, dynamic changes in translocated microbial constituents during two years after cART initiation. An unbiased systems biology approach revealed two distinct pathways driven by changes in the abundance ratio of Serratia to other bacterial genera. Increased CD4 T cell numbers over the first year were associated with high Serratia abundance, pro-inflammatory innate cytokines, and metabolites that drive Th17 gene expression signatures and restoration of mucosal integrity. Subsequently, decreased Serratia abundance and downregulation of innate cytokines allowed re-establishment of systemic T cell homeostasis promoting restoration of Th1 and Th2 gene expression signatures. Analyses of three other geographically distinct cohorts of treated HIV infection established a more generalized principle that changes in diversity and composition of translocated microbial species influence systemic inflammation and consequently CD4 T cell recovery.


Subject(s)
Gastrointestinal Microbiome , HIV Infections/immunology , HIV Infections/microbiology , Antiretroviral Therapy, Highly Active , Biodiversity , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokines/blood , Cohort Studies , Glycolysis , HIV Infections/blood , HIV Infections/drug therapy , Humans , Inflammation/genetics , Inflammation/pathology , Mitochondria/metabolism , Monocytes/metabolism , Nucleic Acids/blood , Principal Component Analysis , Serratia/physiology , Th1 Cells/immunology , Th2 Cells/immunology , Transcription, Genetic , Uganda , Viral Load/immunology
6.
Cell ; 184(19): 5031-5052.e26, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34534465

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with poor patient survival. Toward understanding the underlying molecular alterations that drive PDAC oncogenesis, we conducted comprehensive proteogenomic analysis of 140 pancreatic cancers, 67 normal adjacent tissues, and 9 normal pancreatic ductal tissues. Proteomic, phosphoproteomic, and glycoproteomic analyses were used to characterize proteins and their modifications. In addition, whole-genome sequencing, whole-exome sequencing, methylation, RNA sequencing (RNA-seq), and microRNA sequencing (miRNA-seq) were performed on the same tissues to facilitate an integrated proteogenomic analysis and determine the impact of genomic alterations on protein expression, signaling pathways, and post-translational modifications. To ensure robust downstream analyses, tumor neoplastic cellularity was assessed via multiple orthogonal strategies using molecular features and verified via pathological estimation of tumor cellularity based on histological review. This integrated proteogenomic characterization of PDAC will serve as a valuable resource for the community, paving the way for early detection and identification of novel therapeutic targets.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Proteogenomics , Adenocarcinoma/diagnosis , Adult , Aged , Aged, 80 and over , Algorithms , Carcinoma, Pancreatic Ductal/diagnosis , Cohort Studies , Endothelial Cells/metabolism , Epigenesis, Genetic , Female , Gene Dosage , Genome, Human , Glycolysis , Glycoproteins/biosynthesis , Humans , Male , Middle Aged , Molecular Targeted Therapy , Pancreatic Neoplasms/diagnosis , Phenotype , Phosphoproteins/metabolism , Phosphorylation , Prognosis , Protein Kinases/metabolism , Proteome/metabolism , Substrate Specificity , Transcriptome/genetics
7.
Cell ; 184(16): 4168-4185.e21, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34216539

ABSTRACT

Metabolism is a major regulator of immune cell function, but it remains difficult to study the metabolic status of individual cells. Here, we present Compass, an algorithm to characterize cellular metabolic states based on single-cell RNA sequencing and flux balance analysis. We applied Compass to associate metabolic states with T helper 17 (Th17) functional variability (pathogenic potential) and recovered a metabolic switch between glycolysis and fatty acid oxidation, akin to known Th17/regulatory T cell (Treg) differences, which we validated by metabolic assays. Compass also predicted that Th17 pathogenicity was associated with arginine and downstream polyamine metabolism. Indeed, polyamine-related enzyme expression was enhanced in pathogenic Th17 and suppressed in Treg cells. Chemical and genetic perturbation of polyamine metabolism inhibited Th17 cytokines, promoted Foxp3 expression, and remodeled the transcriptome and epigenome of Th17 cells toward a Treg-like state. In vivo perturbations of the polyamine pathway altered the phenotype of encephalitogenic T cells and attenuated tissue inflammation in CNS autoimmunity.


Subject(s)
Autoimmunity/immunology , Models, Biological , Th17 Cells/immunology , Acetyltransferases/metabolism , Adenosine Triphosphate/metabolism , Aerobiosis/drug effects , Algorithms , Animals , Autoimmunity/drug effects , Chromatin/metabolism , Citric Acid Cycle/drug effects , Cytokines/metabolism , Eflornithine/pharmacology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Epigenome , Fatty Acids/metabolism , Glycolysis/drug effects , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice, Inbred C57BL , Mitochondrial Membrane Transport Proteins/metabolism , Oxidation-Reduction/drug effects , Putrescine/metabolism , Single-Cell Analysis , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th17 Cells/drug effects , Transcriptome/genetics
8.
Cell ; 184(17): 4480-4494.e15, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34320407

ABSTRACT

In neutrophils, nicotinamide adenine dinucleotide phosphate (NADPH) generated via the pentose phosphate pathway fuels NADPH oxidase NOX2 to produce reactive oxygen species for killing invading pathogens. However, excessive NOX2 activity can exacerbate inflammation, as in acute respiratory distress syndrome (ARDS). Here, we use two unbiased chemical proteomic strategies to show that small-molecule LDC7559, or a more potent designed analog NA-11, inhibits the NOX2-dependent oxidative burst in neutrophils by activating the glycolytic enzyme phosphofructokinase-1 liver type (PFKL) and dampening flux through the pentose phosphate pathway. Accordingly, neutrophils treated with NA-11 had reduced NOX2-dependent outputs, including neutrophil cell death (NETosis) and tissue damage. A high-resolution structure of PFKL confirmed binding of NA-11 to the AMP/ADP allosteric activation site and explained why NA-11 failed to agonize phosphofructokinase-1 platelet type (PFKP) or muscle type (PFKM). Thus, NA-11 represents a tool for selective activation of PFKL, the main phosphofructokinase-1 isoform expressed in immune cells.


Subject(s)
Phagocytosis , Phosphofructokinase-1, Liver Type/metabolism , Respiratory Burst , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/metabolism , Allosteric Regulation/drug effects , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Glycolysis/drug effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Microbial Viability/drug effects , Models, Molecular , NADPH Oxidases/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Phagocytosis/drug effects , Phosphate-Binding Proteins/metabolism , Phosphofructokinase-1, Liver Type/antagonists & inhibitors , Phosphofructokinase-1, Liver Type/ultrastructure , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/isolation & purification , Respiratory Burst/drug effects , Tetradecanoylphorbol Acetate/pharmacology
9.
Nat Immunol ; 24(3): 452-462, 2023 03.
Article in English | MEDLINE | ID: mdl-36823405

ABSTRACT

Exposure of lipopolysaccharide triggers macrophage pro-inflammatory polarization accompanied by metabolic reprogramming, characterized by elevated aerobic glycolysis and a broken tricarboxylic acid cycle. However, in contrast to lipopolysaccharide, CD40 signal is able to drive pro-inflammatory and anti-tumorigenic polarization by some yet undefined metabolic programming. Here we show that CD40 activation triggers fatty acid oxidation (FAO) and glutamine metabolism to promote ATP citrate lyase-dependent epigenetic reprogramming of pro-inflammatory genes and anti-tumorigenic phenotypes in macrophages. Mechanistically, glutamine usage reinforces FAO-induced pro-inflammatory and anti-tumorigenic activation by fine-tuning the NAD+/NADH ratio via glutamine-to-lactate conversion. Genetic ablation of important metabolic enzymes involved in CD40-mediated metabolic reprogramming abolishes agonistic anti-CD40-induced antitumor responses and reeducation of tumor-associated macrophages. Together these data show that metabolic reprogramming, which includes FAO and glutamine metabolism, controls the activation of pro-inflammatory and anti-tumorigenic polarization, and highlight a therapeutic potential of metabolic preconditioning of tumor-associated macrophages before agonistic anti-CD40 treatments.


Subject(s)
Fatty Acids , Glutamine , Glutamine/metabolism , Fatty Acids/metabolism , Lipopolysaccharides/metabolism , Glycolysis , Macrophages/metabolism , Macrophage Activation
10.
Annu Rev Immunol ; 31: 259-83, 2013.
Article in English | MEDLINE | ID: mdl-23298210

ABSTRACT

T cell activation leads to dramatic shifts in cell metabolism to protect against pathogens and to orchestrate the action of other immune cells. Quiescent T cells require predominantly ATP-generating processes, whereas proliferating effector T cells require high metabolic flux through growth-promoting pathways. Further, functionally distinct T cell subsets require distinct energetic and biosynthetic pathways to support their specific functional needs. Pathways that control immune cell function and metabolism are intimately linked, and changes in cell metabolism at both the cell and system levels have been shown to enhance or suppress specific T cell functions. As a result of these findings, cell metabolism is now appreciated as a key regulator of T cell function specification and fate. This review discusses the role of cellular metabolism in T cell development, activation, differentiation, and function to highlight the clinical relevance and opportunities for therapeutic interventions that may be used to disrupt immune pathogenesis.


Subject(s)
T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Gene Regulatory Networks/genetics , Gene Regulatory Networks/immunology , Glycolysis/genetics , Glycolysis/immunology , Humans , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Phosphorylation/genetics , Phosphorylation/immunology , T-Lymphocyte Subsets/cytology
11.
Cell ; 182(4): 933-946.e14, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32780992

ABSTRACT

Methanol, being electron rich and derivable from methane or CO2, is a potentially renewable one-carbon (C1) feedstock for microorganisms. Although the ribulose monophosphate (RuMP) cycle used by methylotrophs to assimilate methanol differs from the typical sugar metabolism by only three enzymes, turning a non-methylotrophic organism to a synthetic methylotroph that grows to a high cell density has been challenging. Here we reprogrammed E. coli using metabolic robustness criteria followed by laboratory evolution to establish a strain that can efficiently utilize methanol as the sole carbon source. This synthetic methylotroph alleviated a so far uncharacterized hurdle, DNA-protein crosslinking (DPC), by insertion sequence (IS)-mediated copy number variations (CNVs) and balanced the metabolic flux by mutations. Being capable of growing at a rate comparable with natural methylotrophs in a wide range of methanol concentrations, this synthetic methylotrophic strain illustrates genome editing and evolution for microbial tropism changes and expands the scope of biological C1 conversion.


Subject(s)
Escherichia coli/metabolism , Metabolic Engineering , Methanol/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carbon/metabolism , Citric Acid Cycle/genetics , DNA Copy Number Variations , Directed Molecular Evolution , Escherichia coli/genetics , Formaldehyde/metabolism , Glycolysis , Mutagenesis , Ribosemonophosphates/metabolism
12.
Cell ; 182(3): 641-654.e20, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32615085

ABSTRACT

Targeting glycolysis has been considered therapeutically intractable owing to its essential housekeeping role. However, the context-dependent requirement for individual glycolytic steps has not been fully explored. We show that CRISPR-mediated targeting of glycolysis in T cells in mice results in global loss of Th17 cells, whereas deficiency of the glycolytic enzyme glucose phosphate isomerase (Gpi1) selectively eliminates inflammatory encephalitogenic and colitogenic Th17 cells, without substantially affecting homeostatic microbiota-specific Th17 cells. In homeostatic Th17 cells, partial blockade of glycolysis upon Gpi1 inactivation was compensated by pentose phosphate pathway flux and increased mitochondrial respiration. In contrast, inflammatory Th17 cells experience a hypoxic microenvironment known to limit mitochondrial respiration, which is incompatible with loss of Gpi1. Our study suggests that inhibiting glycolysis by targeting Gpi1 could be an effective therapeutic strategy with minimum toxicity for Th17-mediated autoimmune diseases, and, more generally, that metabolic redundancies can be exploited for selective targeting of disease processes.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Glucose-6-Phosphate Isomerase/metabolism , Glycolysis/genetics , Oxidative Phosphorylation , Pentose Phosphate Pathway/physiology , Th17 Cells/metabolism , Animals , Cell Hypoxia/genetics , Cell Hypoxia/immunology , Chimera/genetics , Chromatography, Gas , Chromatography, Liquid , Clostridium Infections/immunology , Cytokines/deficiency , Cytokines/genetics , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Glucose-6-Phosphate Isomerase/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Glycolysis/immunology , Homeostasis/genetics , Homeostasis/immunology , Inflammation/genetics , Inflammation/immunology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/microbiology , Pentose Phosphate Pathway/genetics , Pentose Phosphate Pathway/immunology , RNA-Seq , Single-Cell Analysis , Th17 Cells/immunology , Th17 Cells/pathology
13.
Cell ; 182(6): 1490-1507.e19, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32916131

ABSTRACT

Metabolic reprogramming is a key feature of many cancers, but how and when it contributes to tumorigenesis remains unclear. Here we demonstrate that metabolic reprogramming induced by mitochondrial fusion can be rate-limiting for immortalization of tumor-initiating cells (TICs) and trigger their irreversible dedication to tumorigenesis. Using single-cell transcriptomics, we find that Drosophila brain tumors contain a rapidly dividing stem cell population defined by upregulation of oxidative phosphorylation (OxPhos). We combine targeted metabolomics and in vivo genetic screening to demonstrate that OxPhos is required for tumor cell immortalization but dispensable in neural stem cells (NSCs) giving rise to tumors. Employing an in vivo NADH/NAD+ sensor, we show that NSCs precisely increase OxPhos during immortalization. Blocking OxPhos or mitochondrial fusion stalls TICs in quiescence and prevents tumorigenesis through impaired NAD+ regeneration. Our work establishes a unique connection between cellular metabolism and immortalization of tumor-initiating cells.


Subject(s)
Brain Neoplasms/metabolism , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Mitochondrial Dynamics , NAD/metabolism , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , Oxidative Phosphorylation , Animals , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Transformation, Neoplastic/pathology , Citric Acid Cycle/genetics , Computational Biology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Glycolysis/genetics , Mass Spectrometry , Metabolomics , Microscopy, Electron, Transmission , Multigene Family , Neural Stem Cells/pathology , Oxygen Consumption/genetics , RNA Interference , Reactive Oxygen Species/metabolism , Single-Cell Analysis , Transcriptome/genetics
14.
Nat Immunol ; 23(3): 386-398, 2022 03.
Article in English | MEDLINE | ID: mdl-35190717

ABSTRACT

The mechanisms underlying the heightened protection mediated by central memory CD8+ T (TCM) cells remain unclear. Here we show that the transcription factor Tcf1 was required in resting TCM cells to generate secondary effector CD8+ T cells and to clear pathogens during recall responses. Recall stimulation of CD8+ TCM cells caused extensive reprogramming of the transcriptome and chromatin accessibility, leading to rapid induction of glycolytic enzymes, cell cycle regulators and transcriptional regulators, including Id3. This cluster of genes did not require Tcf1 in resting CD8+ TCM cells, but depended on Tcf1 for optimal induction and chromatin opening in recall-stimulated CD8+ TCM cells. Tcf1 bound extensively to these recall-induced gene loci in resting CD8+ TCM cells and mediated chromatin interactions that positioned these genes in architectural proximity with poised enhancers. Thus, Tcf1 preprogramed a transcriptional program that supported the bioenergetic and proliferative needs of CD8+ TCM cells in case of a secondary challenge.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Chromatin/metabolism , Glycolysis/genetics , Immunologic Memory/genetics , Mice , Mice, Inbred C57BL
15.
Cell ; 177(2): 384-398.e11, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30853218

ABSTRACT

The signaling organelles of the innate immune system consist of oligomeric protein complexes known as supramolecular organizing centers (SMOCs). Examples of SMOCs include myddosomes and inflammasomes, which respectively induce transcription-dependent and -independent inflammatory responses. The common use of oligomeric structures as signaling platforms suggests multifunctionality, but each SMOC has a singular biochemically defined function. Here, we report that the myddosome is a multifunctional organizing center. In addition to promoting inflammatory transcription factor activation, the myddosome drives the rapid induction of glycolysis. We identify the kinase TBK1 as a myddosome component that promotes glycolysis, but not nuclear factor κB (NF-κB) activation. Synthetic immunology approaches further diversified SMOC activities, as we created interferon- or necroptosis-inducing myddosomes, inflammasomes that induce interferon responses instead of pyroptosis, and a SMOC-like nanomachine that induces interferon expression in response to a chemical ligand. These discoveries demonstrate the flexibility of immune signaling organelles, which permits the design of user-defined innate immune responses.


Subject(s)
Immunity, Innate/immunology , Immunity, Innate/physiology , Signal Transduction/immunology , Animals , Glycolysis/immunology , Inflammasomes , Mice , Mice, Inbred C57BL , Multifunctional Enzymes/immunology , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Organelles/physiology , Protein Serine-Threonine Kinases/metabolism , Toll-Like Receptors
16.
Cell ; 179(5): 1222-1238.e17, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31730859

ABSTRACT

Mitochondrial dysfunction is associated with a spectrum of human conditions, ranging from rare, inborn errors of metabolism to the aging process. To identify pathways that modify mitochondrial dysfunction, we performed genome-wide CRISPR screens in the presence of small-molecule mitochondrial inhibitors. We report a compendium of chemical-genetic interactions involving 191 distinct genetic modifiers, including 38 that are synthetic sick/lethal and 63 that are suppressors. Genes involved in glycolysis (PFKP), pentose phosphate pathway (G6PD), and defense against lipid peroxidation (GPX4) scored high as synthetic sick/lethal. A surprisingly large fraction of suppressors are pathway intrinsic and encode mitochondrial proteins. A striking example of such "intra-organelle" buffering is the alleviation of a chemical defect in complex V by simultaneous inhibition of complex I, which benefits cells by rebalancing redox cofactors, increasing reductive carboxylation, and promoting glycolysis. Perhaps paradoxically, certain forms of mitochondrial dysfunction may best be buffered with "second site" inhibitors to the organelle.


Subject(s)
Genes, Modifier , Mitochondria/genetics , Mitochondria/pathology , Autoantigens/metabolism , Cell Death/drug effects , Cytosol/drug effects , Cytosol/metabolism , Electron Transport Complex I/metabolism , Epistasis, Genetic/drug effects , Ferroptosis/drug effects , Ferroptosis/genetics , Genome , Glutathione Peroxidase/metabolism , Glycolysis/drug effects , Glycolysis/genetics , Humans , K562 Cells , Mitochondria/drug effects , Oligomycins/toxicity , Oxidation-Reduction , Oxidative Phosphorylation/drug effects , Pentose Phosphate Pathway/drug effects , Pentose Phosphate Pathway/genetics , Reactive Oxygen Species/metabolism , Ribonucleoproteins/metabolism , SS-B Antigen
17.
Cell ; 178(5): 1072-1087.e14, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442401

ABSTRACT

Nutritional status potentially influences immune responses; however, how nutritional signals regulate cellular dynamics and functionality remains obscure. Herein, we report that temporary fasting drastically reduces the number of lymphocytes by ∼50% in Peyer's patches (PPs), the inductive site of the gut immune response. Subsequent refeeding seemingly restored the number of lymphocytes, but whose cellular composition was conspicuously altered. A large portion of germinal center and IgA+ B cells were lost via apoptosis during fasting. Meanwhile, naive B cells migrated from PPs to the bone marrow during fasting and then back to PPs during refeeding when stromal cells sensed nutritional signals and upregulated CXCL13 expression to recruit naive B cells. Furthermore, temporal fasting before oral immunization with ovalbumin abolished the induction of antigen-specific IgA, failed to induce oral tolerance, and eventually exacerbated food antigen-induced diarrhea. Thus, nutritional signals are critical in maintaining gut immune homeostasis.


Subject(s)
B-Lymphocytes/physiology , Immunity, Mucosal , Animals , Antigens/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bone Marrow/immunology , Bone Marrow/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Fasting , Gene Expression Regulation , Glycolysis , Immunoglobulin A/metabolism , Male , Mice , Mice, Inbred BALB C , Nutritional Status , Ovalbumin/immunology , Peyer's Patches/immunology , Peyer's Patches/metabolism , Peyer's Patches/pathology , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Signal Transduction , Stromal Cells/cytology , Stromal Cells/metabolism , TOR Serine-Threonine Kinases/metabolism
18.
Cell ; 177(4): 1035-1049.e19, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31031003

ABSTRACT

We performed the first proteogenomic study on a prospectively collected colon cancer cohort. Comparative proteomic and phosphoproteomic analysis of paired tumor and normal adjacent tissues produced a catalog of colon cancer-associated proteins and phosphosites, including known and putative new biomarkers, drug targets, and cancer/testis antigens. Proteogenomic integration not only prioritized genomically inferred targets, such as copy-number drivers and mutation-derived neoantigens, but also yielded novel findings. Phosphoproteomics data associated Rb phosphorylation with increased proliferation and decreased apoptosis in colon cancer, which explains why this classical tumor suppressor is amplified in colon tumors and suggests a rationale for targeting Rb phosphorylation in colon cancer. Proteomics identified an association between decreased CD8 T cell infiltration and increased glycolysis in microsatellite instability-high (MSI-H) tumors, suggesting glycolysis as a potential target to overcome the resistance of MSI-H tumors to immune checkpoint blockade. Proteogenomics presents new avenues for biological discoveries and therapeutic development.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/therapy , Proteogenomics/methods , Apoptosis/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CD8-Positive T-Lymphocytes , Cell Proliferation/genetics , Colonic Neoplasms/metabolism , Genomics/methods , Glycolysis , Humans , Microsatellite Instability , Mutation , Phosphorylation , Prospective Studies , Proteomics/methods , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism
19.
Cell ; 178(1): 176-189.e15, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31155231

ABSTRACT

RLR-mediated type I IFN production plays a pivotal role in elevating host immunity for viral clearance and cancer immune surveillance. Here, we report that glycolysis, which is inactivated during RLR activation, serves as a barrier to impede type I IFN production upon RLR activation. RLR-triggered MAVS-RIG-I recognition hijacks hexokinase binding to MAVS, leading to the impairment of hexokinase mitochondria localization and activation. Lactate serves as a key metabolite responsible for glycolysis-mediated RLR signaling inhibition by directly binding to MAVS transmembrane (TM) domain and preventing MAVS aggregation. Notably, lactate restoration reverses increased IFN production caused by lactate deficiency. Using pharmacological and genetic approaches, we show that lactate reduction by lactate dehydrogenase A (LDHA) inactivation heightens type I IFN production to protect mice from viral infection. Our study establishes a critical role of glycolysis-derived lactate in limiting RLR signaling and identifies MAVS as a direct sensor of lactate, which functions to connect energy metabolism and innate immunity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DEAD Box Protein 58/antagonists & inhibitors , DEAD Box Protein 58/metabolism , Lactic Acid/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Animals , Female , Glycolysis , HEK293 Cells , Humans , Interferon-beta/metabolism , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RAW 264.7 Cells , Receptors, Immunologic , Signal Transduction/drug effects , Transfection
20.
Cell ; 178(2): 330-345.e22, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31257027

ABSTRACT

For tumors to progress efficiently, cancer cells must overcome barriers of oxidative stress. Although dietary antioxidant supplementation or activation of endogenous antioxidants by NRF2 reduces oxidative stress and promotes early lung tumor progression, little is known about its effect on lung cancer metastasis. Here, we show that long-term supplementation with the antioxidants N-acetylcysteine and vitamin E promotes KRAS-driven lung cancer metastasis. The antioxidants stimulate metastasis by reducing levels of free heme and stabilizing the transcription factor BACH1. BACH1 activates transcription of Hexokinase 2 and Gapdh and increases glucose uptake, glycolysis rates, and lactate secretion, thereby stimulating glycolysis-dependent metastasis of mouse and human lung cancer cells. Targeting BACH1 normalized glycolysis and prevented antioxidant-induced metastasis, while increasing endogenous BACH1 expression stimulated glycolysis and promoted metastasis, also in the absence of antioxidants. We conclude that BACH1 stimulates glycolysis-dependent lung cancer metastasis and that BACH1 is activated under conditions of reduced oxidative stress.


Subject(s)
Antioxidants/pharmacology , Basic-Leucine Zipper Transcription Factors/metabolism , Glycolysis/drug effects , Lung Neoplasms/pathology , Animals , Antioxidants/administration & dosage , Basic-Leucine Zipper Transcription Factors/genetics , Cell Movement/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Heme/metabolism , Hexokinase/antagonists & inhibitors , Hexokinase/genetics , Hexokinase/metabolism , Humans , Kaplan-Meier Estimate , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , NF-E2-Related Factor 2/metabolism , Neoplasm Metastasis , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL