Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.696
Filter
Add more filters

Publication year range
1.
Cell ; 179(7): 1483-1498.e22, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31813625

ABSTRACT

Metabolism has been shown to control peripheral immunity, but little is known about its role in central nervous system (CNS) inflammation. Through a combination of proteomic, metabolomic, transcriptomic, and perturbation studies, we found that sphingolipid metabolism in astrocytes triggers the interaction of the C2 domain in cytosolic phospholipase A2 (cPLA2) with the CARD domain in mitochondrial antiviral signaling protein (MAVS), boosting NF-κB-driven transcriptional programs that promote CNS inflammation in experimental autoimmune encephalomyelitis (EAE) and, potentially, multiple sclerosis. cPLA2 recruitment to MAVS also disrupts MAVS-hexokinase 2 (HK2) interactions, decreasing HK enzymatic activity and the production of lactate involved in the metabolic support of neurons. Miglustat, a drug used to treat Gaucher and Niemann-Pick disease, suppresses astrocyte pathogenic activities and ameliorates EAE. Collectively, these findings define a novel immunometabolic mechanism that drives pro-inflammatory astrocyte activities, outlines a new role for MAVS in CNS inflammation, and identifies candidate targets for therapeutic intervention.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Phospholipases A2, Secretory/metabolism , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/pharmacology , 1-Deoxynojirimycin/therapeutic use , Adaptor Proteins, Signal Transducing/genetics , Animals , Astrocytes/drug effects , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Hexokinase/metabolism , Humans , Lactic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phospholipases A2, Secretory/genetics
2.
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
3.
Nat Immunol ; 22(2): 193-204, 2021 02.
Article in English | MEDLINE | ID: mdl-33398181

ABSTRACT

Metabolic reprograming toward aerobic glycolysis is a pivotal mechanism shaping immune responses. Here we show that deficiency in NF-κB-inducing kinase (NIK) impairs glycolysis induction, rendering CD8+ effector T cells hypofunctional in the tumor microenvironment. Conversely, ectopic expression of NIK promotes CD8+ T cell metabolism and effector function, thereby profoundly enhancing antitumor immunity and improving the efficacy of T cell adoptive therapy. NIK regulates T cell metabolism via a NF-κB-independent mechanism that involves stabilization of hexokinase 2 (HK2), a rate-limiting enzyme of the glycolytic pathway. NIK prevents autophagic degradation of HK2 through controlling cellular reactive oxygen species levels, which in turn involves modulation of glucose-6-phosphate dehydrogenase (G6PD), an enzyme that mediates production of the antioxidant NADPH. We show that the G6PD-NADPH redox system is important for HK2 stability and metabolism in activated T cells. These findings establish NIK as a pivotal regulator of T cell metabolism and highlight a post-translational mechanism of metabolic regulation.


Subject(s)
CD8-Positive T-Lymphocytes/enzymology , Colonic Neoplasms/enzymology , Energy Metabolism , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/enzymology , Melanoma, Experimental/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Cytotoxicity, Immunologic , Enzyme Stability , Female , Glucosephosphate Dehydrogenase/metabolism , Glycolysis , Hexokinase/genetics , Hexokinase/metabolism , Immunotherapy, Adoptive , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/transplantation , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Mice, Knockout , NADP/metabolism , Phenotype , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Microenvironment , NF-kappaB-Inducing Kinase
4.
Immunity ; 57(5): 1019-1036.e9, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38677292

ABSTRACT

Group 3 innate lymphoid cells (ILC3) are the major subset of gut-resident ILC with essential roles in infections and tissue repair, but how they adapt to the gut environment to maintain tissue residency is unclear. We report that Tox2 is critical for gut ILC3 maintenance and function. Gut ILC3 highly expressed Tox2, and depletion of Tox2 markedly decreased ILC3 in gut but not at central sites, resulting in defective control of Citrobacter rodentium infection. Single-cell transcriptional profiling revealed decreased expression of Hexokinase-2 in Tox2-deficient gut ILC3. Consistent with the requirement for hexokinases in glycolysis, Tox2-/- ILC3 displayed decreased ability to utilize glycolysis for protein translation. Ectopic expression of Hexokinase-2 rescued Tox2-/- gut ILC3 defects. Hypoxia and interleukin (IL)-17A each induced Tox2 expression in ILC3, suggesting a mechanism by which ILC3 adjusts to fluctuating environments by programming glycolytic metabolism. Our results reveal the requirement for Tox2 to support the metabolic adaptation of ILC3 within the gastrointestinal tract.


Subject(s)
Citrobacter rodentium , Enterobacteriaceae Infections , Glycolysis , HMGB Proteins , Immunity, Innate , Lymphocytes , Mice, Knockout , Animals , Mice , Adaptation, Physiological/immunology , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Hexokinase/metabolism , Hexokinase/genetics , Interleukin-17/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred C57BL , Trans-Activators/metabolism , Trans-Activators/genetics , HMGB Proteins/genetics , HMGB Proteins/immunology , HMGB Proteins/metabolism
5.
Mol Cell ; 84(14): 2593-2595, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39059367

ABSTRACT

In this issue of Molecular Cell, Pilic et al.1 show that hexokinase, the first enzyme of glycolysis, forms perimitochondrial rings that prevent mitochondrial fragmentation when ATP levels drop.


Subject(s)
Glucose , Hexokinase , Mitochondria , Mitochondrial Dynamics , Hexokinase/metabolism , Hexokinase/genetics , Mitochondria/metabolism , Mitochondria/enzymology , Glucose/metabolism , Adenosine Triphosphate/metabolism , Humans , Animals , Glycolysis
6.
Mol Cell ; 84(14): 2732-2746.e5, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38981483

ABSTRACT

Metabolic enzymes can adapt during energy stress, but the consequences of these adaptations remain understudied. Here, we discovered that hexokinase 1 (HK1), a key glycolytic enzyme, forms rings around mitochondria during energy stress. These HK1-rings constrict mitochondria at contact sites with the endoplasmic reticulum (ER) and mitochondrial dynamics protein (MiD51). HK1-rings prevent mitochondrial fission by displacing the dynamin-related protein 1 (Drp1) from mitochondrial fission factor (Mff) and mitochondrial fission 1 protein (Fis1). The disassembly of HK1-rings during energy restoration correlated with mitochondrial fission. Mechanistically, we identified that the lack of ATP and glucose-6-phosphate (G6P) promotes the formation of HK1-rings. Mutations that affect the formation of HK1-rings showed that HK1-rings rewire cellular metabolism toward increased TCA cycle activity. Our findings highlight that HK1 is an energy stress sensor that regulates the shape, connectivity, and metabolic activity of mitochondria. Thus, the formation of HK1-rings may affect mitochondrial function in energy-stress-related pathologies.


Subject(s)
Dynamins , Energy Metabolism , Hexokinase , Mitochondria , Mitochondrial Dynamics , Mitochondrial Proteins , Hexokinase/metabolism , Hexokinase/genetics , Humans , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria/enzymology , Dynamins/metabolism , Dynamins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Animals , Adenosine Triphosphate/metabolism , Stress, Physiological , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Citric Acid Cycle , Glucose-6-Phosphate/metabolism , Mice , HeLa Cells , HEK293 Cells , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Mutation
7.
Cell ; 166(3): 624-636, 2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27374331

ABSTRACT

Degradation of Gram-positive bacterial cell wall peptidoglycan in macrophage and dendritic cell phagosomes leads to activation of the NLRP3 inflammasome, a cytosolic complex that regulates processing and secretion of interleukin (IL)-1ß and IL-18. While many inflammatory responses to peptidoglycan are mediated by detection of its muramyl dipeptide component in the cytosol by NOD2, we report here that NLRP3 inflammasome activation is caused by release of N-acetylglucosamine that is detected in the cytosol by the glycolytic enzyme hexokinase. Inhibition of hexokinase by N-acetylglucosamine causes its dissociation from mitochondria outer membranes, and we found that this is sufficient to activate the NLRP3 inflammasome. In addition, we observed that glycolytic inhibitors and metabolic conditions affecting hexokinase function and localization induce inflammasome activation. While previous studies have demonstrated that signaling by pattern recognition receptors can regulate metabolic processes, this study shows that a metabolic enzyme can act as a pattern recognition receptor. PAPERCLIP.


Subject(s)
Hexokinase/metabolism , Inflammasomes/metabolism , Peptidoglycan/metabolism , Receptors, Immunologic/metabolism , Acetylation , Acetylglucosamine/metabolism , Animals , Bacillus anthracis/metabolism , Cell Wall/metabolism , Dendritic Cells/metabolism , Glycolysis , Humans , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Models, Biological , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Potassium/metabolism
8.
Mol Cell ; 83(6): 829-831, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36931254

ABSTRACT

Hexokinase 2 (HK2) plays a multifaceted role in the regulation of cellular activities. A new study by Hu et al.1 delineated a critical role of HK2 in governing glycolytic flux and mitochondrial activity, thereby modulating microglial functions in maladaptive inflammation in brain diseases.


Subject(s)
Hexokinase , Microglia , Hexokinase/genetics , Hexokinase/metabolism , Microglia/metabolism , Gatekeeping , Mitochondria/metabolism , Glycolysis/physiology , Glucose/metabolism
9.
Cell ; 162(6): 1217-28, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26321681

ABSTRACT

Activated T cells engage aerobic glycolysis and anabolic metabolism for growth, proliferation, and effector functions. We propose that a glucose-poor tumor microenvironment limits aerobic glycolysis in tumor-infiltrating T cells, which suppresses tumoricidal effector functions. We discovered a new role for the glycolytic metabolite phosphoenolpyruvate (PEP) in sustaining T cell receptor-mediated Ca(2+)-NFAT signaling and effector functions by repressing sarco/ER Ca(2+)-ATPase (SERCA) activity. Tumor-specific CD4 and CD8 T cells could be metabolically reprogrammed by increasing PEP production through overexpression of phosphoenolpyruvate carboxykinase 1 (PCK1), which bolstered effector functions. Moreover, PCK1-overexpressing T cells restricted tumor growth and prolonged the survival of melanoma-bearing mice. This study uncovers new metabolic checkpoints for T cell activity and demonstrates that metabolic reprogramming of tumor-reactive T cells can enhance anti-tumor T cell responses, illuminating new forms of immunotherapy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Melanoma/therapy , Monitoring, Immunologic , Phosphoenolpyruvate/metabolism , Tumor Microenvironment , Animals , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Glycolysis , Hexokinase/metabolism , Immunotherapy , Mice , NFATC Transcription Factors/metabolism , Receptors, Antigen, T-Cell/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction , Transforming Growth Factor beta/immunology
10.
Mol Cell ; 82(7): 1261-1277.e9, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35305311

ABSTRACT

The product of hexokinase (HK) enzymes, glucose-6-phosphate, can be metabolized through glycolysis or directed to alternative metabolic routes, such as the pentose phosphate pathway (PPP) to generate anabolic intermediates. HK1 contains an N-terminal mitochondrial binding domain (MBD), but its physiologic significance remains unclear. To elucidate the effect of HK1 mitochondrial dissociation on cellular metabolism, we generated mice lacking the HK1 MBD (ΔE1HK1). These mice produced a hyper-inflammatory response when challenged with lipopolysaccharide. Additionally, there was decreased glucose flux below the level of GAPDH and increased upstream flux through the PPP. The glycolytic block below GAPDH is mediated by the binding of cytosolic HK1 with S100A8/A9, resulting in GAPDH nitrosylation through iNOS. Additionally, human and mouse macrophages from conditions of low-grade inflammation, such as aging and diabetes, displayed increased cytosolic HK1 and reduced GAPDH activity. Our data indicate that HK1 mitochondrial binding alters glucose metabolism through regulation of GAPDH.


Subject(s)
Glucose , Hexokinase/metabolism , Animals , Glucose/metabolism , Glycolysis , Hexokinase/genetics , Mice , Mitochondria/metabolism , Pentose Phosphate Pathway
11.
Cell ; 148(3): 543-55, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22304920

ABSTRACT

The transcription factor ATF2 elicits oncogenic activities in melanoma and tumor suppressor activities in nonmalignant skin cancer. Here, we identify that ATF2 tumor suppressor function is determined by its ability to localize at the mitochondria, where it alters membrane permeability following genotoxic stress. The ability of ATF2 to reach the mitochondria is determined by PKCε, which directs ATF2 nuclear localization. Genotoxic stress attenuates PKCε effect on ATF2; enables ATF2 nuclear export and localization at the mitochondria, where it perturbs the HK1-VDAC1 complex; increases mitochondrial permeability; and promotes apoptosis. Significantly, high levels of PKCε, as seen in melanoma cells, block ATF2 nuclear export and function at the mitochondria, thereby attenuating apoptosis following exposure to genotoxic stress. In melanoma tumor samples, high PKCε levels associate with poor prognosis. Overall, our findings provide the framework for understanding how subcellular localization enables ATF2 oncogenic or tumor suppressor functions.


Subject(s)
Activating Transcription Factor 2/metabolism , Apoptosis , Melanoma/metabolism , Mitochondria/metabolism , Protein Kinase C-epsilon/metabolism , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cytosol/metabolism , DNA Damage , Fibroblasts/metabolism , Hexokinase/metabolism , Humans , Prognosis , Protein Transport , Voltage-Dependent Anion Channel 1/metabolism
12.
Proc Natl Acad Sci U S A ; 121(2): e2306454120, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38170752

ABSTRACT

Mitochondrial and lysosomal functions are intimately linked and are critical for cellular homeostasis, as evidenced by the fact that cellular senescence, aging, and multiple prominent diseases are associated with concomitant dysfunction of both organelles. However, it is not well understood how the two important organelles are regulated. Transcription factor EB (TFEB) is the master regulator of lysosomal function and is also implicated in regulating mitochondrial function; however, the mechanism underlying the maintenance of both organelles remains to be fully elucidated. Here, by comprehensive transcriptome analysis and subsequent chromatin immunoprecipitation-qPCR, we identified hexokinase domain containing 1 (HKDC1), which is known to function in the glycolysis pathway as a direct TFEB target. Moreover, HKDC1 was upregulated in both mitochondrial and lysosomal stress in a TFEB-dependent manner, and its function was critical for the maintenance of both organelles under stress conditions. Mechanistically, the TFEB-HKDC1 axis was essential for PINK1 (PTEN-induced kinase 1)/Parkin-dependent mitophagy via its initial step, PINK1 stabilization. In addition, the functions of HKDC1 and voltage-dependent anion channels, with which HKDC1 interacts, were essential for the clearance of damaged lysosomes and maintaining mitochondria-lysosome contact. Interestingly, HKDC1 regulated mitophagy and lysosomal repair independently of its prospective function in glycolysis. Furthermore, loss function of HKDC1 accelerated DNA damage-induced cellular senescence with the accumulation of hyperfused mitochondria and damaged lysosomes. Our results show that HKDC1, a factor downstream of TFEB, maintains both mitochondrial and lysosomal homeostasis, which is critical to prevent cellular senescence.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Hexokinase , Hexokinase/genetics , Hexokinase/metabolism , Prospective Studies , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Mitochondria/metabolism , Lysosomes/metabolism , Protein Kinases/metabolism , Cellular Senescence/genetics , Homeostasis , Autophagy/genetics
13.
PLoS Genet ; 20(7): e1011357, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39074078

ABSTRACT

Hexokinase (HK) catalyzes the first irreversible rate-limiting step in glycolysis that converts glucose to glucose-6-phosphate. HK1 is ubiquitously expressed in the brain, erythrocytes, and other tissues where glycolysis serves as the major source of ATP production. Spermatogenic cell-specific type 1 hexokinase (HK1S) is expressed in sperm but its physiological role in male mice is still unknown. In this study, we generate Hk1s knockout mice using the CRISPR/Cas9 system to study the gene function in vivo. Hk1s mRNA is exclusively expressed in testes starting from postnatal day 18 and continuing to adulthood. HK1S protein is specifically localized in the outer surface of the sperm fibrous sheath (FS). Depletion of Hk1s leads to infertility in male mice and reduces sperm glycolytic pathway activity, yet they have normal motile parameters and ATP levels. In addition, by using in vitro fertilization (IVF), Hk1s deficient sperms are unable to fertilize cumulus-intact or cumulus-free oocytes, but can normally fertilize zona pellucida-free oocytes. Moreover, Hk1s deficiency impairs sperm migration into the oviduct, reduces acrosome reaction, and prevents capacitation-associated increases in tyrosine phosphorylation, which are probable causes of infertility. Taken together, our results reveal that HK1S plays a critical role in sperm function and male fertility in mice.


Subject(s)
Fertility , Hexokinase , Infertility, Male , Mice, Knockout , Sperm Capacitation , Spermatozoa , Tyrosine , Animals , Hexokinase/genetics , Hexokinase/metabolism , Male , Mice , Phosphorylation , Spermatozoa/metabolism , Sperm Capacitation/genetics , Infertility, Male/genetics , Infertility, Male/metabolism , Fertility/genetics , Tyrosine/metabolism , Female , Testis/metabolism , Sperm Motility/genetics , Glycolysis , Spermatogenesis/genetics
14.
Development ; 150(20)2023 10 15.
Article in English | MEDLINE | ID: mdl-37842778

ABSTRACT

As photoautotrophic organisms, plants produce an incredible spectrum of pigments, anti-herbivory compounds, structural materials and energic intermediates. These biosynthetic routes help plants grow, reproduce and mitigate stress. HEXOKINASE1 (HXK1), a metabolic enzyme and glucose sensor, catalyzes the phosphorylation of hexoses, a key introductory step for many of these pathways. However, previous studies have largely focused on the glucose sensing and signaling functions of HXK1, and the importance of the enzyme's catalytic function is only recently being connected to plant development. In this brief Spotlight, we describe the developmental significance of plant HXK1 and its role in plant metabolic pathways, specifically in glucose-6-phosphate production. Furthermore, we describe the emerging connections between metabolism and development and suggest that HXK1 signaling and catalytic activity regulate discrete areas of plant development.


Subject(s)
Glucose-6-Phosphate , Hexokinase , Plant Development , Glucose/metabolism , Hexokinase/genetics , Hexokinase/metabolism , Phosphorylation , Plants/metabolism
15.
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
16.
Proc Natl Acad Sci U S A ; 120(8): e2209177120, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36787364

ABSTRACT

Microglial phagocytosis is an energetically demanding process that plays a critical role in the removal of toxic protein aggregates in Alzheimer's disease (AD). Recent evidence indicates that a switch in energy production from mitochondrial respiration to glycolysis disrupts this important protective microglial function and may provide therapeutic targets for AD. Here, we demonstrate that the translocator protein (TSPO) and a member of its mitochondrial complex, hexokinase-2 (HK), play critical roles in microglial respiratory-glycolytic metabolism and phagocytosis. Pharmacological and genetic loss-of-function experiments showed that TSPO is critical for microglial respiratory metabolism and energy supply for phagocytosis, and its expression is enriched in phagocytic microglia of AD mice. Meanwhile, HK controlled glycolytic metabolism and phagocytosis via mitochondrial binding or displacement. In cultured microglia, TSPO deletion impaired mitochondrial respiration and increased mitochondrial recruitment of HK, inducing a switch to glycolysis and reducing phagocytosis. To determine the functional significance of mitochondrial HK recruitment, we developed an optogenetic tool for reversible control of HK localization. Displacement of mitochondrial HK inhibited glycolysis and improved phagocytosis in TSPO-knockout microglia. Mitochondrial HK recruitment also coordinated the inflammatory switch to glycolysis that occurs in response to lipopolysaccharide in normal microglia. Interestingly, cytosolic HK increased phagocytosis independent of its metabolic activity, indicating an immune signaling function. Alzheimer's beta amyloid drastically stimulated mitochondrial HK recruitment in cultured microglia, which may contribute to microglial dysfunction in AD. Thus, targeting mitochondrial HK may offer an immunotherapeutic approach to promote phagocytic microglial function in AD.


Subject(s)
Alzheimer Disease , Animals , Mice , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Hexokinase/genetics , Hexokinase/metabolism , Microglia/metabolism , Phagocytosis , Mitochondria/metabolism
17.
PLoS Genet ; 19(5): e1010745, 2023 05.
Article in English | MEDLINE | ID: mdl-37196001

ABSTRACT

Glucose is the preferred carbon source for most eukaryotes, and the first step in its metabolism is phosphorylation to glucose-6-phosphate. This reaction is catalyzed by hexokinases or glucokinases. The yeast Saccharomyces cerevisiae encodes three such enzymes, Hxk1, Hxk2, and Glk1. In yeast and mammals, some isoforms of this enzyme are found in the nucleus, suggesting a possible moonlighting function beyond glucose phosphorylation. In contrast to mammalian hexokinases, yeast Hxk2 has been proposed to shuttle into the nucleus in glucose-replete conditions, where it reportedly moonlights as part of a glucose-repressive transcriptional complex. To achieve its role in glucose repression, Hxk2 reportedly binds the Mig1 transcriptional repressor, is dephosphorylated at serine 15 and requires an N-terminal nuclear localization sequence (NLS). We used high-resolution, quantitative, fluorescent microscopy of live cells to determine the conditions, residues, and regulatory proteins required for Hxk2 nuclear localization. Countering previous yeast studies, we find that Hxk2 is largely excluded from the nucleus under glucose-replete conditions but is retained in the nucleus under glucose-limiting conditions. We find that the Hxk2 N-terminus does not contain an NLS but instead is necessary for nuclear exclusion and regulating multimerization. Amino acid substitutions of the phosphorylated residue, serine 15, disrupt Hxk2 dimerization but have no effect on its glucose-regulated nuclear localization. Alanine substation at nearby lysine 13 affects dimerization and maintenance of nuclear exclusion in glucose-replete conditions. Modeling and simulation provide insight into the molecular mechanisms of this regulation. In contrast to earlier studies, we find that the transcriptional repressor Mig1 and the protein kinase Snf1 have little effect on Hxk2 localization. Instead, the protein kinase Tda1 regulates Hxk2 localization. RNAseq analyses of the yeast transcriptome dispels the idea that Hxk2 moonlights as a transcriptional regulator of glucose repression, demonstrating that Hxk2 has a negligible role in transcriptional regulation in both glucose-replete and limiting conditions. Our studies define a new model of cis- and trans-acting regulators of Hxk2 dimerization and nuclear localization. Based on our data, the nuclear translocation of Hxk2 in yeast occurs in glucose starvation conditions, which aligns well with the nuclear regulation of mammalian orthologs. Our results lay the foundation for future studies of Hxk2 nuclear activity.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Glucose/metabolism , Hexokinase/genetics , Hexokinase/metabolism , Protein Kinases/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Serine/genetics , Serine/metabolism , Transcription Factors/metabolism
18.
J Biol Chem ; 300(1): 105518, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38042489

ABSTRACT

Bacillus Calmette-Guérin (BCG) vaccination induces a type of immune memory known as "trained immunity", characterized by the immunometabolic and epigenetic changes in innate immune cells. However, the molecular mechanism underlying the strategies for inducing and/or boosting trained immunity in alveolar macrophages remains unknown. Here, we found that mucosal vaccination with the recombinant strain rBCGPPE27 significantly augmented the trained immune response in mice, facilitating a superior protective response against Mycobacterium tuberculosis and non-related bacterial reinfection in mice when compared to BCG. Mucosal immunization with rBCGPPE27 enhanced innate cytokine production by alveolar macrophages associated with promoted glycolytic metabolism, typical of trained immunity. Deficiency of the mammalian target of rapamycin complex 2 and hexokinase 1 abolished the immunometabolic and epigenetic rewiring in mouse alveolar macrophages after mucosal rBCGPPE27 vaccination. Most noteworthy, utilizing rBCGPPE27's higher-up trained effects: The single mucosal immunization with rBCGPPE27-adjuvanted coronavirus disease (CoV-2) vaccine raised the rapid development of virus-specific immunoglobulin G antibodies, boosted pseudovirus neutralizing antibodies, and augmented T helper type 1-biased cytokine release by vaccine-specific T cells, compared to BCG/CoV-2 vaccine. These findings revealed that mucosal recombinant BCG vaccine induces lung-resident memory macrophages and enhances trained immunity via reprogramming mTORC2- and HK-1-mediated aerobic glycolysis, providing new vaccine strategies for improving tuberculosis (TB) or coronavirus variant vaccinations, and targeting innate immunity via mucosal surfaces.


Subject(s)
BCG Vaccine , Hexokinase , Immunologic Memory , Lung , Macrophages, Alveolar , Mechanistic Target of Rapamycin Complex 2 , Mycobacterium tuberculosis , Trained Immunity , Animals , Mice , BCG Vaccine/immunology , Cytokines/metabolism , Lung/immunology , Macrophages, Alveolar/immunology , Mycobacterium tuberculosis/immunology , Vaccines, Synthetic/immunology , Mechanistic Target of Rapamycin Complex 2/metabolism , Hexokinase/metabolism
19.
J Biol Chem ; 300(3): 105684, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38272231

ABSTRACT

Eukaryotic elongation factor 1A1 (EEF1A1) is canonically involved in protein synthesis but also has noncanonical functions in diverse cellular processes. Previously, we identified EEF1A1 as a mediator of lipotoxicity and demonstrated that chemical inhibition of EEF1A1 activity reduced mouse liver lipid accumulation. These findings suggested a link between EEF1A1 and metabolism. Therefore, we investigated its role in regulating metabolic substrate preference. EEF1A1-deficient Chinese hamster ovary (2E2) cells displayed reduced media lactate accumulation. These effects were also observed with EEF1A1 knockdown in human hepatocyte-like HepG2 cells and in WT Chinese hamster ovary and HepG2 cells treated with selective EEF1A inhibitors, didemnin B, or plitidepsin. Extracellular flux analyses revealed decreased glycolytic ATP production and increased mitochondrial-to-glycolytic ATP production ratio in 2E2 cells, suggesting a more oxidative metabolic phenotype. Correspondingly, fatty acid oxidation was increased in 2E2 cells. Both 2E2 cells and HepG2 cells treated with didemnin B exhibited increased neutral lipid content, which may be required to support elevated oxidative metabolism. RNA-seq revealed a >90-fold downregulation of a rate-limiting glycolytic enzyme, hexokinase 2, which we confirmed through immunoblotting and enzyme activity assays. Pathway enrichment analysis identified downregulations in TNFA signaling via NFKB and MYC targets. Correspondingly, nuclear abundances of RELB and MYC were reduced in 2E2 cells. Thus, EEF1A1 deficiency may perturb glycolysis by limiting NFKB- and MYC-mediated gene expression, leading to decreased hexokinase expression and activity. This is the first evidence of a role for a translation elongation factor, EEF1A1, in regulating metabolic substrate utilization in mammalian cells.


Subject(s)
Hexokinase , Peptide Elongation Factor 1 , Animals , Cricetinae , Humans , Adenosine Triphosphate , Cell Line , Cricetulus , Hexokinase/genetics , Hexokinase/metabolism , Lipids , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/chemistry , Peptide Elongation Factor 1/metabolism , Glycolysis , Oxidation-Reduction , Cell Movement , Cell Proliferation , Lipid Metabolism
20.
FASEB J ; 38(6): e23556, 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38498348

ABSTRACT

PARP-1 over-activation results in cell death via excessive PAR generation in different cell types, including neurons following brain ischemia. Glycolysis, mitochondrial function, and redox balance are key cellular processes altered in brain ischemia. Studies show that PAR generated after PARP-1 over-activation can bind hexokinase-1 (HK-1) and result in glycolytic defects and subsequent mitochondrial dysfunction. HK-1 is the neuronal hexokinase and catalyzes the first reaction of glycolysis, converting glucose to glucose-6-phosphate (G6P), a common substrate for glycolysis, and the pentose phosphate pathway (PPP). PPP is critical in maintaining NADPH and GSH levels via G6P dehydrogenase activity. Therefore, defects in HK-1 will not only decrease cellular bioenergetics but will also cause redox imbalance due to the depletion of GSH. In brain ischemia, whether PAR-mediated inhibition of HK-1 results in bioenergetics defects and redox imbalance is not known. We used oxygen-glucose deprivation (OGD) in mouse cortical neurons to mimic brain ischemia in neuronal cultures and observed that PARP-1 activation via PAR formation alters glycolysis, mitochondrial function, and redox homeostasis in neurons. We used pharmacological inhibition of PARP-1 and adenoviral-mediated overexpression of wild-type HK-1 (wtHK-1) and PAR-binding mutant HK-1 (pbmHK-1). Our data show that PAR inhibition or overexpression of HK-1 significantly improves glycolysis, mitochondrial function, redox homeostasis, and cell survival in mouse cortical neurons exposed to OGD. These results suggest that PAR binding and inhibition of HK-1 during OGD drive bioenergetic defects in neurons due to inhibition of glycolysis and impairment of mitochondrial function.


Subject(s)
Brain Ischemia , Oxygen , Mice , Animals , Oxygen/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Hexokinase/genetics , Hexokinase/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/metabolism , Glucose/metabolism , Brain Ischemia/metabolism , Glycolysis , Neurons/metabolism , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL