Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.896
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 37: 325-347, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30676821

ABSTRACT

ATP, NAD+, and nucleic acids are abundant purines that, in addition to having critical intracellular functions, have evolved extracellular roles as danger signals released in response to cell lysis, apoptosis, degranulation, or membrane pore formation. In general ATP and NAD+ have excitatory and adenosine has anti-inflammatory effects on immune cells. This review focuses on recent advances in our understanding of purine release mechanisms, ectoenzymes that metabolize purines (CD38, CD39, CD73, ENPP1, and ENPP2/autotaxin), and signaling by key P2 purinergic receptors (P2X7, P2Y2, and P2Y12). In addition to metabolizing ATP or NAD+, some purinergic ectoenzymes metabolize other inflammatory modulators, notably lysophosphatidic acid and cyclic GMP-AMP (cGAMP). Also discussed are extracellular signaling effects of NAD+ mediated by ADP-ribosylation, and epigenetic effects of intracellular adenosine mediated by modification of S-adenosylmethionine-dependent DNA methylation.


Subject(s)
Inflammation/immunology , Purines/metabolism , Receptors, Purinergic/metabolism , ADP-Ribosylation , Adenosine Triphosphate/metabolism , Animals , DNA Methylation , Humans , Inflammation/genetics , Inflammation/metabolism , Lysophospholipids/metabolism , Signal Transduction
2.
Cell ; 184(17): 4531-4546.e26, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34314702

ABSTRACT

Defects in translation lead to changes in the expression of proteins that can serve as drivers of cancer formation. Here, we show that cytosolic NAD+ synthesis plays an essential role in ovarian cancer by regulating translation and maintaining protein homeostasis. Expression of NMNAT-2, a cytosolic NAD+ synthase, is highly upregulated in ovarian cancers. NMNAT-2 supports the catalytic activity of the mono(ADP-ribosyl) transferase (MART) PARP-16, which mono(ADP-ribosyl)ates (MARylates) ribosomal proteins. Depletion of NMNAT-2 or PARP-16 leads to inhibition of MARylation, increased polysome association and enhanced translation of specific mRNAs, aggregation of their translated protein products, and reduced growth of ovarian cancer cells. Furthermore, MARylation of the ribosomal proteins, such as RPL24 and RPS6, inhibits polysome assembly by stabilizing eIF6 binding to ribosomes. Collectively, our results demonstrate that ribosome MARylation promotes protein homeostasis in cancers by fine-tuning the levels of protein synthesis and preventing toxic protein aggregation.


Subject(s)
ADP-Ribosylation , Ovarian Neoplasms/metabolism , Protein Biosynthesis , Proteostasis , Ribosomes/metabolism , 3' Untranslated Regions/genetics , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , Endoplasmic Reticulum Stress , Fallopian Tubes/metabolism , Female , Humans , Mice, Inbred NOD , Mice, SCID , NAD/metabolism , Nicotinamide-Nucleotide Adenylyltransferase , Nucleic Acid Conformation , Ovarian Neoplasms/pathology , Poly(ADP-ribose) Polymerases/metabolism , Polyribosomes/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Ribosomal Proteins/metabolism
3.
Cell ; 173(1): 74-89.e20, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29570999

ABSTRACT

A decline in capillary density and blood flow with age is a major cause of mortality and morbidity. Understanding why this occurs is key to future gains in human health. NAD precursors reverse aspects of aging, in part, by activating sirtuin deacylases (SIRT1-SIRT7) that mediate the benefits of exercise and dietary restriction (DR). We show that SIRT1 in endothelial cells is a key mediator of pro-angiogenic signals secreted from myocytes. Treatment of mice with the NAD+ booster nicotinamide mononucleotide (NMN) improves blood flow and increases endurance in elderly mice by promoting SIRT1-dependent increases in capillary density, an effect augmented by exercise or increasing the levels of hydrogen sulfide (H2S), a DR mimetic and regulator of endothelial NAD+ levels. These findings have implications for improving blood flow to organs and tissues, increasing human performance, and reestablishing a virtuous cycle of mobility in the elderly.


Subject(s)
Aging , Hydrogen Sulfide/metabolism , NAD/metabolism , Animals , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Mice , Mice, Knockout , Microvessels/metabolism , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Neovascularization, Physiologic , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Physical Conditioning, Animal , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Notch/metabolism , Signal Transduction , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Sirtuin 1/metabolism , Vascular Endothelial Growth Factor A/metabolism
4.
Cell ; 170(4): 664-677.e11, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28802039

ABSTRACT

The process of aging and circadian rhythms are intimately intertwined, but how peripheral clocks involved in metabolic homeostasis contribute to aging remains unknown. Importantly, caloric restriction (CR) extends lifespan in several organisms and rewires circadian metabolism. Using young versus old mice, fed ad libitum or under CR, we reveal reprogramming of the circadian transcriptome in the liver. These age-dependent changes occur in a highly tissue-specific manner, as demonstrated by comparing circadian gene expression in the liver versus epidermal and skeletal muscle stem cells. Moreover, de novo oscillating genes under CR show an enrichment in SIRT1 targets in the liver. This is accompanied by distinct circadian hepatic signatures in NAD+-related metabolites and cyclic global protein acetylation. Strikingly, this oscillation in acetylation is absent in old mice while CR robustly rescues global protein acetylation. Our findings indicate that the clock operates at the crossroad between protein acetylation, liver metabolism, and aging.


Subject(s)
Aging/metabolism , Circadian Rhythm , Liver/metabolism , Metabolic Networks and Pathways , Acetyl Coenzyme A/metabolism , Acetylation , Aging/pathology , Animals , Caloric Restriction , Histones/metabolism , Liver/pathology , Mice , NAD/metabolism , Proteins/metabolism , Sirtuin 1/metabolism , Stem Cells/metabolism , Transcriptome
5.
Mol Cell ; 84(4): 744-759.e6, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38266638

ABSTRACT

Serine metabolism is involved in the fate decisions of immune cells; however, whether and how de novo serine synthesis shapes innate immune cell function remain unknown. Here, we first demonstrated that inflammatory macrophages have high expression of phosphoglycerate dehydrogenase (PHGDH, the rate-limiting enzyme of de novo serine synthesis) via nuclear factor κB signaling. Notably, the pharmacological inhibition or genetic modulation of PHGDH limits macrophage interleukin (IL)-1ß production through NAD+ accumulation and subsequent NAD+-dependent SIRT1 and SIRT3 expression and activity. Mechanistically, PHGDH not only sustains IL-1ß expression through H3K9/27 acetylation-mediated transcriptional activation of Toll-like receptor 4 but also supports IL-1ß maturation via NLRP3-K21/22/24/ASC-K21/22/24 acetylation-mediated activation of the NLRP3 inflammasome. Moreover, mice with myeloid-specific depletion of Phgdh show alleviated inflammatory responses in lipopolysaccharide-induced systemic inflammation. This study reveals a network by which a metabolic enzyme, involved in de novo serine synthesis, mediates post-translational modifications and epigenetic regulation to orchestrate IL-1ß production, providing a potential inflammatory disease target.


Subject(s)
NAD , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Mice , Acetylation , Epigenesis, Genetic , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages/metabolism , NAD/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Processing, Post-Translational , Serine/metabolism
6.
Mol Cell ; 84(11): 2185-2202.e12, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38788717

ABSTRACT

Retrons are toxin-antitoxin systems protecting bacteria against bacteriophages via abortive infection. The Retron-Eco1 antitoxin is formed by a reverse transcriptase (RT) and a non-coding RNA (ncRNA)/multi-copy single-stranded DNA (msDNA) hybrid that neutralizes an uncharacterized toxic effector. Yet, the molecular mechanisms underlying phage defense remain unknown. Here, we show that the N-glycosidase effector, which belongs to the STIR superfamily, hydrolyzes NAD+ during infection. Cryoelectron microscopy (cryo-EM) analysis shows that the msDNA stabilizes a filament that cages the effector in a low-activity state in which ADPr, a NAD+ hydrolysis product, is covalently linked to the catalytic E106 residue. Mutations shortening the msDNA induce filament disassembly and the effector's toxicity, underscoring the msDNA role in immunity. Furthermore, we discovered a phage-encoded Retron-Eco1 inhibitor (U56) that binds ADPr, highlighting the intricate interplay between retron systems and phage evolution. Our work outlines the structural basis of Retron-Eco1 defense, uncovering ADPr's pivotal role in immunity.


Subject(s)
Bacteriophages , Cryoelectron Microscopy , NAD , NAD/metabolism , Bacteriophages/genetics , Bacteriophages/metabolism , Bacteriophages/immunology , Hydrolysis , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/immunology , Toxin-Antitoxin Systems/genetics , Escherichia coli/virology , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli/metabolism
7.
Annu Rev Biochem ; 85: 431-54, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-26844395

ABSTRACT

Recent developments indicate that macrodomains, an ancient and diverse protein domain family, are key players in the recognition, interpretation, and turnover of ADP-ribose (ADPr) signaling. Crucial to this is the ability of macrodomains to recognize ADPr either directly, in the form of a metabolic derivative, or as a modification covalently bound to proteins. Thus, macrodomains regulate a wide variety of cellular and organismal processes, including DNA damage repair, signal transduction, and immune response. Their importance is further indicated by the fact that dysregulation or mutation of a macrodomain is associated with several diseases, including cancer, developmental defects, and neurodegeneration. In this review, we summarize the current insights into macrodomain evolution and how this evolution influenced their structural and functional diversification. We highlight some aspects of macrodomain roles in pathobiology as well as their emerging potential as therapeutic targets.


Subject(s)
DNA Repair , Escherichia coli Proteins/chemistry , Neoplasms/enzymology , Poly(ADP-ribose) Polymerases/chemistry , Protein Processing, Post-Translational , Repressor Proteins/chemistry , Virus Diseases/enzymology , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Animals , DNA Damage , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Evolution, Molecular , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Multigene Family , Neoplasms/chemistry , Neoplasms/genetics , Neoplasms/pathology , Phylogeny , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Domains , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Structural Homology, Protein , Virus Diseases/genetics , Virus Diseases/pathology , Virus Diseases/virology
8.
Immunity ; 55(6): 1067-1081.e8, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35659337

ABSTRACT

Immunoregulatory B cells impede antitumor immunity through unknown features and mechanisms. We report the existence of leucine-tRNA-synthase-2 (LARS2)-expressing B cell (LARS B) subset with a transforming growth factor-ß1 (TGF-ß1)-dominant regulatory feature in both mouse and human progressive colorectal cancer (CRC). Of note, LARS B cells exhibited a leucine nutrient preference and displayed active mitochondrial aminoacyl-tRNA biosynthesis. They were located outside the tertiary lymphoid structure and correlated with colorectal hyperplasia and shortened survival in CRC patients. A leucine diet induced LARS B cell generation, whereas LARS B cell deletion by Lars2 gene ablation or leucine blockage repressed CRC immunoevasion. Mechanistically, LARS2 programmed mitochondrial nicotinamide adenine dinucleotide (NAD+) regeneration and oxidative metabolism, thus determining the regulatory feature of LARS B cells in which the NAD-dependent protein deacetylase sirtuin-1 (SIRT1) was involved. We propose a leucine-dieting scheme to inhibit LARS B cells, which is safe and useful for CRC therapy.


Subject(s)
Amino Acyl-tRNA Synthetases , Colorectal Neoplasms , Animals , Humans , Leucine , Mice , Mitochondria/metabolism , NAD/metabolism , RNA, Transfer
9.
Mol Cell ; 83(8): 1340-1349.e7, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37084714

ABSTRACT

The glycerol-3-phosphate shuttle (G3PS) is a major NADH shuttle that regenerates reducing equivalents in the cytosol and produces energy in the mitochondria. Here, we demonstrate that G3PS is uncoupled in kidney cancer cells where the cytosolic reaction is ∼4.5 times faster than the mitochondrial reaction. The high flux through cytosolic glycerol-3-phosphate dehydrogenase (GPD) is required to maintain redox balance and support lipid synthesis. Interestingly, inhibition of G3PS by knocking down mitochondrial GPD (GPD2) has no effect on mitochondrial respiration. Instead, loss of GPD2 upregulates cytosolic GPD on a transcriptional level and promotes cancer cell proliferation by increasing glycerol-3-phosphate supply. The proliferative advantage of GPD2 knockdown tumor can be abolished by pharmacologic inhibition of lipid synthesis. Taken together, our results suggest that G3PS is not required to run as an intact NADH shuttle but is instead truncated to support complex lipid synthesis in kidney cancer.


Subject(s)
Glycerol-3-Phosphate Dehydrogenase (NAD+) , Kidney Neoplasms , Lipids , Humans , Glycerol/metabolism , Glycerol-3-Phosphate Dehydrogenase (NAD+)/genetics , Glycerol-3-Phosphate Dehydrogenase (NAD+)/metabolism , Glycerolphosphate Dehydrogenase/genetics , Glycerolphosphate Dehydrogenase/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Lipids/biosynthesis , NAD/metabolism , Oxidation-Reduction , Phosphates/metabolism
10.
Annu Rev Biochem ; 84: 227-63, 2015.
Article in English | MEDLINE | ID: mdl-25747399

ABSTRACT

Protein ADP-ribosylation is an ancient posttranslational modification with high biochemical complexity. It alters the function of modified proteins or provides a scaffold for the recruitment of other proteins and thus regulates several cellular processes. ADP-ribosylation is governed by ADP-ribosyltransferases and a subclass of sirtuins (writers), is sensed by proteins that contain binding modules (readers) that recognize specific parts of the ADP-ribosyl posttranslational modification, and is removed by ADP-ribosylhydrolases (erasers). The large amount of experimental data generated and technical progress made in the last decade have significantly advanced our knowledge of the function of ADP-ribosylation at the molecular level. This review summarizes the current knowledge of nuclear ADP-ribosylation reactions and their role in chromatin plasticity, cell differentiation, and epigenetics and discusses current progress and future perspectives.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Chromatin/metabolism , Epigenesis, Genetic , Protein Processing, Post-Translational , ADP Ribose Transferases/metabolism , Animals , Cell Differentiation , Humans , N-Glycosyl Hydrolases/metabolism , Protein Structure, Tertiary , Proteins/chemistry , Proteins/metabolism
11.
Immunity ; 54(1): 68-83.e6, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33238133

ABSTRACT

While antibiotics are intended to specifically target bacteria, most are known to affect host cell physiology. In addition, some antibiotic classes are reported as immunosuppressive for reasons that remain unclear. Here, we show that Linezolid, a ribosomal-targeting antibiotic (RAbo), effectively blocked the course of a T cell-mediated autoimmune disease. Linezolid and other RAbos were strong inhibitors of T helper-17 cell effector function in vitro, showing that this effect was independent of their antibiotic activity. Perturbing mitochondrial translation in differentiating T cells, either with RAbos or through the inhibition of mitochondrial elongation factor G1 (mEF-G1) progressively compromised the integrity of the electron transport chain. Ultimately, this led to deficient oxidative phosphorylation, diminishing nicotinamide adenine dinucleotide concentrations and impairing cytokine production in differentiating T cells. In accordance, mice lacking mEF-G1 in T cells were protected from experimental autoimmune encephalomyelitis, demonstrating that this pathway is crucial in maintaining T cell function and pathogenicity.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Linezolid/therapeutic use , Mitochondria/metabolism , Peptides, Cyclic/therapeutic use , Ribosomes/metabolism , Th17 Cells/physiology , Animals , Autoimmunity/drug effects , Cell Differentiation , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Targeted Therapy , Multiple Sclerosis/drug therapy , NAD/metabolism , Oxidative Phosphorylation , Peptide Elongation Factor G/genetics , Peptide Elongation Factor G/metabolism
12.
Mol Cell ; 82(17): 3299-3311.e8, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35868311

ABSTRACT

NAD+ kinases (NADKs) are metabolite kinases that phosphorylate NAD+ molecules to make NADP+, a limiting substrate for the generation of reducing power NADPH. NADK2 sustains mitochondrial NADPH production that enables proline biosynthesis and antioxidant defense. However, its molecular architecture and mechanistic regulation remain undescribed. Here, we report the crystal structure of human NADK2, revealing a substrate-driven mode of activation. We find that NADK2 presents an unexpected dimeric organization instead of the typical tetrameric assemblage observed for other NADKs. A specific extended segment (aa 325-365) is crucial for NADK2 dimerization and activity. Moreover, we characterize numerous acetylation events, including those on Lys76 and Lys304, which reside near the active site and inhibit NADK2 activity without disrupting dimerization, thereby reducing mitochondrial NADP(H) production, proline synthesis, and cell growth. These findings reveal important molecular insight into the structure and regulation of a vital enzyme in mitochondrial NADPH and proline metabolism.


Subject(s)
Lysine , NAD , Acetylation , Catalytic Domain , Humans , Lysine/metabolism , Mitochondrial Proteins/metabolism , NAD/metabolism , NADP/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proline/metabolism
13.
Genes Dev ; 36(3-4): 149-166, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35115380

ABSTRACT

The process of tissue regeneration occurs in a developmentally timed manner, yet the role of circadian timing is not understood. Here, we identify a role for the adult muscle stem cell (MuSC)-autonomous clock in the control of muscle regeneration following acute ischemic injury. We observed greater muscle repair capacity following injury during the active/wake period as compared with the inactive/rest period in mice, and loss of Bmal1 within MuSCs leads to impaired muscle regeneration. We demonstrate that Bmal1 loss in MuSCs leads to reduced activated MuSC number at day 3 postinjury, indicating a failure to properly expand the myogenic precursor pool. In cultured primary myoblasts, we observed that loss of Bmal1 impairs cell proliferation in hypoxia (a condition that occurs in the first 1-3 d following tissue injury in vivo), as well as subsequent myofiber differentiation. Loss of Bmal1 in both cultured myoblasts and in vivo activated MuSCs leads to reduced glycolysis and premature activation of prodifferentiation gene transcription and epigenetic remodeling. Finally, hypoxic cell proliferation and myofiber formation in Bmal1-deficient myoblasts are restored by increasing cytosolic NAD+ Together, we identify the MuSC clock as a pivotal regulator of oxygen-dependent myoblast cell fate and muscle repair through the control of the NAD+-driven response to injury.


Subject(s)
ARNTL Transcription Factors , NAD , Satellite Cells, Skeletal Muscle , ARNTL Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Hypoxia , Mice , Muscle Development/genetics , Muscle, Skeletal , Myoblasts
14.
Genes Dev ; 36(9-10): 601-617, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35654456

ABSTRACT

The differentiation of embryonic stem cells (ESCs) into a lineage-committed state is a dynamic process involving changes in cellular metabolism, epigenetic modifications, post-translational modifications, gene expression, and RNA processing. Here we integrated data from metabolomic, proteomic, and transcriptomic assays to characterize how alterations in NAD+ metabolism during the differentiation of mouse ESCs lead to alteration of the PARP1-mediated ADP-ribosylated (ADPRylated) proteome and mRNA isoform specialization. Our metabolomic analyses indicate that mESCs use distinct NAD+ biosynthetic pathways in different cell states: the de novo pathway in the pluripotent state, and the salvage and Preiss-Handler pathways as differentiation progresses. We observed a dramatic induction of PARP1 catalytic activity driven by enhanced nuclear NAD+ biosynthesis during the early stages of mESC differentiation (e.g., within 12 h of LIF removal). PARP1-modified proteins in mESCs are enriched for biological processes related to stem cell maintenance, transcriptional regulation, and RNA processing. The PARP1 substrates include core spliceosome components, such as U2AF35 and U2AF65, whose splicing functions are modulated by PARP1-mediated site-specific ADP-ribosylation. Finally, we observed that splicing is dysregulated genome-wide in Parp1 knockout mESCs. Together, these results demonstrate a role for the NAD+-PARP1 axis in the maintenance of mESC state, specifically in the splicing program during differentiation.


Subject(s)
NAD , Poly(ADP-ribose) Polymerases , ADP-Ribosylation , Animals , Embryonic Stem Cells/metabolism , Mice , NAD/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Proteomics
15.
Physiol Rev ; 102(1): 339-341, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34494892

ABSTRACT

During the COVID-19 pandemic, efforts have been made worldwide to develop effective therapies to address the devastating immune-mediated effects of SARS-CoV-2. With the exception of monoclonal antibody-mediated therapeutics and preventive approaches such as mass immunization, most experimental or repurposed drugs have failed in large randomized clinical trials (https://www.who.int/publications/i/item/therapeutics-and-covid-19-living-guideline). The worldwide spread of SARS-CoV-2 virus revealed specific susceptibilities to the virus among the elderly and individuals with age-related syndromes. These populations were more likely to experience a hyperimmune response characterized by a treatment-resistant acute lung pathology accompanied by multiple organ failure. These observations underscore the interplay between the virus, the biology of aging, and outcomes observed in the most severe cases of SARS-CoV-2 infection. The ectoenzyme CD38 has been implicated in the process of "inflammaging" in aged tissues. In a current publication, Horenstein et al. present evidence to support the hypothesis that CD38 plays a central role in altered immunometabolism resulting from COVID-19 infection. The authors discuss a critical but underappreciated trifecta of CD38-mediated NAD+ metabolism, aging, and COVID-19 immune response and speculate that the CD38/NAD+ axis is a promising therapeutic target for this disease.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , COVID-19/physiopathology , Membrane Glycoproteins/metabolism , SARS-CoV-2 , ADP-ribosyl Cyclase 1/genetics , Aging , Gene Expression Regulation, Enzymologic , Humans , Membrane Glycoproteins/genetics , NAD/metabolism
16.
Mol Cell ; 81(4): 691-707.e6, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33382985

ABSTRACT

Aerobic glycolysis, or preferential fermentation of glucose-derived pyruvate to lactate despite available oxygen, is associated with proliferation across many organisms and conditions. To better understand that association, we examined the metabolic consequence of activating the pyruvate dehydrogenase complex (PDH) to increase pyruvate oxidation at the expense of fermentation. We find that increasing PDH activity impairs cell proliferation by reducing the NAD+/NADH ratio. This change in NAD+/NADH is caused by increased mitochondrial membrane potential that impairs mitochondrial electron transport and NAD+ regeneration. Uncoupling respiration from ATP synthesis or increasing ATP hydrolysis restores NAD+/NADH homeostasis and proliferation even when glucose oxidation is increased. These data suggest that when demand for NAD+ to support oxidation reactions exceeds the rate of ATP turnover in cells, NAD+ regeneration by mitochondrial respiration becomes constrained, promoting fermentation, despite available oxygen. This argues that cells engage in aerobic glycolysis when the demand for NAD+ is in excess of the demand for ATP.


Subject(s)
Adenosine Triphosphate/metabolism , Glucose/metabolism , Glycolysis , NAD/metabolism , A549 Cells , Adenosine Triphosphate/genetics , Aerobiosis , Glucose/genetics , HeLa Cells , Humans , NAD/genetics , Oxidation-Reduction
17.
Mol Cell ; 81(18): 3848-3865.e19, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34547241

ABSTRACT

Metabolic rewiring and redox balance play pivotal roles in cancer. Cellular senescence is a barrier for tumorigenesis circumvented in cancer cells by poorly understood mechanisms. We report a multi-enzymatic complex that reprograms NAD metabolism by transferring reducing equivalents from NADH to NADP+. This hydride transfer complex (HTC) is assembled by malate dehydrogenase 1, malic enzyme 1, and cytosolic pyruvate carboxylase. HTC is found in phase-separated bodies in the cytosol of cancer or hypoxic cells and can be assembled in vitro with recombinant proteins. HTC is repressed in senescent cells but induced by p53 inactivation. HTC enzymes are highly expressed in mouse and human prostate cancer models, and their inactivation triggers senescence. Exogenous expression of HTC is sufficient to bypass senescence, rescue cells from complex I inhibitors, and cooperate with oncogenic RAS to transform primary cells. Altogether, we provide evidence for a new multi-enzymatic complex that reprograms metabolism and overcomes cellular senescence.


Subject(s)
Cellular Senescence/physiology , NAD/metabolism , Aging/metabolism , Aging/physiology , Animals , Cell Line, Tumor , Cellular Senescence/genetics , Cytosol , Glucose/metabolism , Humans , Hydrogen/chemistry , Hydrogen/metabolism , Malate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , NAD/physiology , Oxidation-Reduction , Pyruvate Carboxylase/metabolism , Pyruvic Acid/metabolism
18.
Mol Cell ; 81(2): 340-354.e5, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33450210

ABSTRACT

In addition to its role as an electron transporter, mitochondrial nicotinamide adenine dinucleotide (NAD+) is an important co-factor for enzymatic reactions, including ADP-ribosylation. Although mitochondria harbor the most intra-cellular NAD+, mitochondrial ADP-ribosylation remains poorly understood. Here we provide evidence for mitochondrial ADP-ribosylation, which was identified using various methodologies including immunofluorescence, western blot, and mass spectrometry. We show that mitochondrial ADP-ribosylation reversibly increases in response to respiratory chain inhibition. Conversely, H2O2-induced oxidative stress reciprocally induces nuclear and reduces mitochondrial ADP-ribosylation. Elevated mitochondrial ADP-ribosylation, in turn, dampens H2O2-triggered nuclear ADP-ribosylation and increases MMS-induced ARTD1 chromatin retention. Interestingly, co-treatment of cells with the mitochondrial uncoupler FCCP decreases PARP inhibitor efficacy. Together, our results suggest that mitochondrial ADP-ribosylation is a dynamic cellular process that impacts nuclear ADP-ribosylation and provide evidence for a NAD+-mediated mitochondrial-nuclear crosstalk.


Subject(s)
ADP-Ribosylation , Cell Nucleus/enzymology , Mitochondria/enzymology , NAD/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , ADP-Ribosylation/drug effects , Animals , Antimycin A/analogs & derivatives , Antimycin A/pharmacology , Cell Line , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Chromatin/chemistry , Chromatin/metabolism , Electron Transport/drug effects , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Methacrylates/pharmacology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/genetics , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/enzymology , Oligomycins/pharmacology , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/enzymology , Poly (ADP-Ribose) Polymerase-1/genetics , Rotenone/pharmacology , Thiazoles/pharmacology
19.
Trends Biochem Sci ; 49(6): 477-479, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677919

ABSTRACT

A recently characterized RNA modification is NAD+-modified RNAs (NAD-RNAs). Various enzymes decap NAD-RNAs, and Wang and Yu et al. now describe another, namely Toll/interleukin-1 receptor (TIR) domain-containing proteins of bacteria and Archaea. TIR decapping products are a specific variant of cyclic ADP ribose (ADPR)-RNAs (v-cADPR-RNAs), opening a new window to the NAD-RNA world.


Subject(s)
NAD , NAD/metabolism , Humans , Protein Domains , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1/chemistry , RNA/metabolism , RNA/chemistry
20.
Trends Biochem Sci ; 49(2): 97-98, 2024 02.
Article in English | MEDLINE | ID: mdl-37980188

ABSTRACT

Bacteriophages have been a treasure trove for the discovery of fundamental biological principles and the expansion of our enzymatic toolkit since the dawn of molecular biology. In a recent study by Wolfram-Schauerte et al. these ubiquitous bacteria-infecting viruses reveal yet another new biological concept: post-translational modification through covalent RNA-protein linkages.


Subject(s)
Bacteriophages , Bacteriophages/genetics , RNA , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL