1
|
Lautrup S, Zhang SQ, Funayama S, Lirussi L, Visnovska T, Cheung HH, Niere M, Tian Y, Nilsen HL, Selbæk G, Saarela J, Maezawa Y, Yokote K, Nilsson P, Chan WY, Kato H, Ziegler M, Bohr VA, Fang EF. Decreased mitochondrial NAD+ in WRN deficient cells links to dysfunctional proliferation. Aging (Albany NY) 2025; 17:937-959. [PMID: 40179319 PMCID: PMC12074813 DOI: 10.18632/aging.206236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025]
Abstract
Werner syndrome (WS), caused by mutations in the RecQ helicase WERNER (WRN) gene, is a classical accelerated aging disease with patients suffering from several metabolic dysfunctions without a cure. While, as we previously reported, depleted NAD+ causes accumulation of damaged mitochondria, leading to compromised metabolism, how mitochondrial NAD+ changes in WS and the impact on WS pathologies were unknown. We show that loss of WRN increases senescence in mesenchymal stem cells (MSCs) likely related to dysregulation of metabolic and aging pathways. In line with this, NAD+ augmentation, via supplementation with nicotinamide riboside, reduces senescence and improves mitochondrial metabolic profiles in MSCs with WRN knockout (WRN-/-) and in primary fibroblasts derived from WS patients compared to controls. Moreover, WRN deficiency results in decreased mitochondrial NAD+ (measured indirectly via mitochondrially-expressed PARP activity), and altered expression of key salvage pathway enzymes, including NMNAT1 and NAMPT; ChIP-seq data analysis unveils a potential co-regulatory axis between WRN and the NMNATs, likely important for chromatin stability and DNA metabolism. However, restoration of mitochondrial or cellular NAD+ is not sufficient to reinstall cellular proliferation in immortalized cells with siRNA-mediated knockdown of WRN, highlighting an indispensable role of WRN in proliferation even in an NAD+ affluent environment. Further cell and animal studies are needed to deepen our understanding of the underlying mechanisms, facilitating related drug development.
Collapse
Affiliation(s)
- Sofie Lautrup
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog 1478, Norway
| | - Shi-Qi Zhang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog 1478, Norway
| | - Shinichiro Funayama
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba 260-0856, Japan
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba 260-8677, Japan
| | - Lisa Lirussi
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog 1478, Norway
- Department of Microbiology, Oslo University Hospital, Oslo 0450, Norway
| | - Tina Visnovska
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog 1478, Norway
| | - Hoi-Hung Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Marc Niere
- Department of Biomedicine, University of Bergen, Bergen 5009, Norway
| | - Yuyao Tian
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Hilde Loge Nilsen
- Department of Microbiology, Oslo University Hospital, Oslo 0450, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
| | - Geir Selbæk
- Institute of Clinical Medicine, University of Oslo, Oslo 0372, Norway
- The Norwegian National Centre for Aging and Health, Vestfold Hospital Trust, Tønsberg 3103, Norway
- Department of Geriatric Medicine, Oslo University Hospital, Oslo 0450, Norway
| | - Janna Saarela
- Centre for Molecular Medicine Norway (NCMM), University of Oslo, Oslo 0372, Norway
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Medical Genetics, Oslo University Hospital, Oslo 0450, Norway
| | - Yoshiro Maezawa
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba 260-0856, Japan
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba 260-8677, Japan
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba 260-0856, Japan
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba 260-8677, Japan
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Solna 17164, Sweden
| | - Wai-Yee Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Hisaya Kato
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba 260-0856, Japan
- Department of Diabetes, Metabolism and Endocrinology, Chiba University Hospital, Chiba 260-8677, Japan
| | - Mathias Ziegler
- Department of Biomedicine, University of Bergen, Bergen 5009, Norway
- Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena 07745, Germany
| | - Vilhelm A. Bohr
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen 1172, Denmark
| | - Evandro F. Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog 1478, Norway
- The Norwegian Centre on Healthy Ageing (NO-Age) and The Norwegian National Anti-Alzheimer’s Disease (NO-AD) Networks, Oslo 0372, Norway
| |
Collapse
|
2
|
Chen L, Wang P, Huang G, Cheng W, Liu K, Yu Q. Quantitative dynamics of intracellular NMN by genetically encoded biosensor. Biosens Bioelectron 2025; 267:116842. [PMID: 39418868 DOI: 10.1016/j.bios.2024.116842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/19/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024]
Abstract
Nicotinamide mononucleotide (NMN) is the direct precursor and a major booster of NAD+ with increasing applications in NAD+- and aging-related pathologies. However, measuring live cell NMN dynamics was not possible, leaving key questions in NMN uptake and intracellular regulation unanswered. Here we developed genetically encoded bioluminescent and fluorescent sensors to quantify subcellular NMN in live cells by engineering specific NMN-responsive protein scaffolds fused to luciferase and fluorescent proteins. The sensor dissected the multimechanistic uptake of exogenous NMN and nicotinamide riboside (NR) in live cells and further measured the NMN levels across different subcellular compartments, as well as the perturbed NMN/NAD+ ratios by external supplements. Moreover, we measured the NMN regulation by NAD(H) hydrolase Nudts and peroxisomal carrier Pxmp2 and identified Slc25a45 as a potential mitochondrial NMN regulator for its unique fingerprint on the local NMN/NAD+ ratio. Collectively, the genetically encoded sensors provide a useful tool for visualizing NMN metabolism.
Collapse
Affiliation(s)
- Liuqing Chen
- Sino-European Center of Biomedicine and Health, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Pei Wang
- Sino-European Center of Biomedicine and Health, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Guan Huang
- Sino-European Center of Biomedicine and Health, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Wenxiang Cheng
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Kaijing Liu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China; State Key Laboratory of Oncology in South China, Sun Yat-sen University, 510060, Guangzhou, China.
| | - Qiuliyang Yu
- Sino-European Center of Biomedicine and Health, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China.
| |
Collapse
|
3
|
Høyland LE, VanLinden MR, Niere M, Strømland Ø, Sharma S, Dietze J, Tolås I, Lucena E, Bifulco E, Sverkeli LJ, Cimadamore-Werthein C, Ashrafi H, Haukanes KF, van der Hoeven B, Dölle C, Davidsen C, Pettersen IKN, Tronstad KJ, Mjøs SA, Hayat F, Makarov MV, Migaud ME, Heiland I, Ziegler M. Subcellular NAD + pools are interconnected and buffered by mitochondrial NAD . Nat Metab 2024; 6:2319-2337. [PMID: 39702414 DOI: 10.1038/s42255-024-01174-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 11/05/2024] [Indexed: 12/21/2024]
Abstract
The coenzyme NAD+ is consumed by signalling enzymes, including poly-ADP-ribosyltransferases (PARPs) and sirtuins. Ageing is associated with a decrease in cellular NAD+ levels, but how cells cope with persistently decreased NAD+ concentrations is unclear. Here, we show that subcellular NAD+ pools are interconnected, with mitochondria acting as a rheostat to maintain NAD+ levels upon excessive consumption. To evoke chronic, compartment-specific overconsumption of NAD+, we engineered cell lines stably expressing PARP activity in mitochondria, the cytosol, endoplasmic reticulum or peroxisomes, resulting in a decline of cellular NAD+ concentrations by up to 50%. Isotope-tracer flux measurements and mathematical modelling show that the lowered NAD+ concentration kinetically restricts NAD+ consumption to maintain a balance with the NAD+ biosynthesis rate, which remains unchanged. Chronic NAD+ deficiency is well tolerated unless mitochondria are directly targeted. Mitochondria maintain NAD+ by import through SLC25A51 and reversibly cleave NAD+ to nicotinamide mononucleotide and ATP when NMNAT3 is present. Thus, these organelles can maintain an additional, virtual NAD+ pool. Our results are consistent with a well-tolerated ageing-related NAD+ decline as long as the vulnerable mitochondrial pool is not directly affected.
Collapse
Affiliation(s)
- Lena E Høyland
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Marc Niere
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Suraj Sharma
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Neuro-SysMed Center, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Jörn Dietze
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ingvill Tolås
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Biological Sciences, NTNU Ålesund, Ålesund, Norway
| | - Eva Lucena
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ersilia Bifulco
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Lars J Sverkeli
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Camila Cimadamore-Werthein
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Hanan Ashrafi
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | | | - Christian Dölle
- Neuro-SysMed Center, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's Disease, University of Bergen, Bergen, Norway
| | - Cédric Davidsen
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | | | - Karl J Tronstad
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Svein A Mjøs
- Department of Chemistry, University of Bergen, Bergen, Norway
| | - Faisal Hayat
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Mikhail V Makarov
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Marie E Migaud
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Ines Heiland
- Neuro-SysMed Center, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Mathias Ziegler
- Department of Biomedicine, University of Bergen, Bergen, Norway.
| |
Collapse
|
4
|
Ren C, Zhang S, Chen Y, Deng K, Kuang M, Gong Z, Zhang K, Wang P, Huang P, Zhou Z, Gong A. Exploring nicotinamide adenine dinucleotide precursors across biosynthesis pathways: Unraveling their role in the ovary. FASEB J 2024; 38:e23804. [PMID: 39037422 DOI: 10.1096/fj.202400453r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/31/2024] [Accepted: 06/26/2024] [Indexed: 07/23/2024]
Abstract
Natural Nicotinamide Adenine Dinucleotide (NAD+) precursors have attracted much attention due to their positive effects in promoting ovarian health. However, their target tissue, synthesis efficiency, advantages, and disadvantages are still unclear. This review summarizes the distribution of NAD+ at the tissue, cellular and subcellular levels, discusses its biosynthetic pathways and the latest findings in ovary, include: (1) NAD+ plays distinct roles both intracellularly and extracellularly, adapting its distribution in response to requirements. (2) Different precursors differs in target tissues, synthetic efficiency, biological utilization, and adverse effects. Importantly: tryptophan is primarily utilized in the liver and kidneys, posing metabolic risks in excess; nicotinamide (NAM) is indispensable for maintaining NAD+ levels; nicotinic acid (NA) constructs a crucial bridge between intestinal microbiota and the host with diverse functions; nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN) increase NAD+ systemically and can be influenced by delivery route, tissue specificity, and transport efficiency. (3) The biosynthetic pathways of NAD+ are intricately intertwined. They provide multiple sources and techniques for NAD+ synthesis, thereby reducing the dependence on a single molecule to maintain cellular NAD+ levels. However, an excess of a specific precursor potentially influencing other pathways. In addition, Protein expression analysis suggest that ovarian tissues may preferentially utilize NAM and NMN. These findings summarize the specific roles and potential of NAD+ precursors in enhancing ovarian health. Future research should delve into the molecular mechanisms and intervention strategies of different precursors, aiming to achieve personalized prevention or treatment of ovarian diseases, and reveal their clinical application value.
Collapse
Affiliation(s)
- Caifang Ren
- School of Medicine, Jiangsu University, Zhenjiang, China
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Shuang Zhang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yanyan Chen
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Kaiping Deng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Meiqian Kuang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zihao Gong
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ke Zhang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Panqi Wang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Pan Huang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhengrong Zhou
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Aihua Gong
- School of Medicine, Jiangsu University, Zhenjiang, China
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| |
Collapse
|
5
|
Sauter R, Sharma S, Heiland I. Accounting for NAD Concentrations in Genome-Scale Metabolic Models Captures Important Metabolic Alterations in NAD-Depleted Systems. Biomolecules 2024; 14:602. [PMID: 38786009 PMCID: PMC11117748 DOI: 10.3390/biom14050602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is a ubiquitous molecule found within all cells, acting as a crucial coenzyme in numerous metabolic reactions. It plays a vital role in energy metabolism, cellular signaling, and DNA repair. Notably, NAD levels decline naturally with age, and this decline is associated with the development of various age-related diseases. Despite this established link, current genome-scale metabolic models, which offer powerful tools for understanding cellular metabolism, do not account for the dynamic changes in NAD concentration. This impedes our understanding of a fluctuating NAD level's impact on cellular metabolism and its contribution to age-related pathologies. To bridge this gap in our knowledge, we have devised a novel method that integrates altered NAD concentration into genome-scale models of human metabolism. This approach allows us to accurately reflect the changes in fatty acid metabolism, glycolysis, and oxidative phosphorylation observed experimentally in an engineered human cell line with a compromised level of subcellular NAD.
Collapse
Affiliation(s)
- Roland Sauter
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, 9019 Tromsø, Norway;
| | - Suraj Sharma
- Department of Biomedicine, University of Bergen, 5020 Bergen, Norway;
| | - Ines Heiland
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, 9019 Tromsø, Norway;
- Department of Clinical Medicine, University of Bergen, 5020 Bergen, Norway
| |
Collapse
|
6
|
Iqbal T, Nakagawa T. The therapeutic perspective of NAD + precursors in age-related diseases. Biochem Biophys Res Commun 2024; 702:149590. [PMID: 38340651 DOI: 10.1016/j.bbrc.2024.149590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is the fundamental molecule that performs numerous biological reactions and is crucial for maintaining cellular homeostasis. Studies have found that NAD+ decreases with age in certain tissues, and age-related NAD+ depletion affects physiological functions and contributes to various aging-related diseases. Supplementation of NAD+ precursor significantly elevates NAD+ levels in murine tissues, effectively mitigates metabolic syndrome, enhances cardiovascular health, protects against neurodegeneration, and boosts muscular strength. Despite the versatile therapeutic functions of NAD+ in animal studies, the efficacy of NAD+ precursors in clinical studies have been limited compared with that in the pre-clinical study. Clinical studies have demonstrated that NAD+ precursor treatment efficiently increases NAD+ levels in various tissues, though their clinical proficiency is insufficient to ameliorate the diseases. However, the latest studies regarding NAD+ precursors and their metabolism highlight the significant role of gut microbiota. The studies found that orally administered NAD+ intermediates interact with the gut microbiome. These findings provide compelling evidence for future trials to further explore the involvement of gut microbiota in NAD+ metabolism. Also, the reduced form of NAD+ precursor shows their potential to raise NAD+, though preclinical studies have yet to discover their efficacy. This review sheds light on NAD+ therapeutic efficiency in preclinical and clinical studies and the effect of the gut microbiota on NAD+ metabolism.
Collapse
Affiliation(s)
- Tooba Iqbal
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan; Research Center for Pre-Disease Science, University of Toyama, Toyama, Japan.
| |
Collapse
|
7
|
Lautrup S, Hou Y, Fang EF, Bohr VA. Roles of NAD + in Health and Aging. Cold Spring Harb Perspect Med 2024; 14:a041193. [PMID: 37848251 PMCID: PMC10759992 DOI: 10.1101/cshperspect.a041193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
NAD+, the essential metabolite involved in multiple reactions such as the regulation of cellular metabolism, energy production, DNA repair, mitophagy and autophagy, inflammation, and neuronal function, has been the subject of intense research in the field of aging and disease over the last decade. NAD+ levels decline with aging and in some age-related diseases, and reduction in NAD+ affects all the hallmarks of aging. Here, we present an overview of the discovery of NAD+, the cellular pathways of producing and consuming NAD+, and discuss how imbalances in the production rate and cellular request of NAD+ likely contribute to aging and age-related diseases including neurodegeneration. Preclinical studies have revealed great potential for NAD+ precursors in promotion of healthy aging and improvement of neurodegeneration. This has led to the initiation of several clinical trials with NAD+ precursors to treat accelerated aging, age-associated dysfunctions, and diseases including Alzheimer's and Parkinson's. NAD supplementation has great future potential clinically, and these studies will also provide insight into the mechanisms of aging.
Collapse
Affiliation(s)
- Sofie Lautrup
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - Yujun Hou
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Evandro F Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| | - Vilhelm A Bohr
- DNA Repair Section, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
- Danish Center for Healthy Aging, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
8
|
Waddell J, Khatoon R, Kristian T. Cellular and Mitochondrial NAD Homeostasis in Health and Disease. Cells 2023; 12:1329. [PMID: 37174729 PMCID: PMC10177113 DOI: 10.3390/cells12091329] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/26/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
The mitochondrion has a unique position among other cellular organelles due to its dynamic properties and symbiotic nature, which is reflected in an active exchange of metabolites and cofactors between the rest of the intracellular compartments. The mitochondrial energy metabolism is greatly dependent on nicotinamide adenine dinucleotide (NAD) as a cofactor that is essential for both the activity of respiratory and TCA cycle enzymes. The NAD level is determined by the rate of NAD synthesis, the activity of NAD-consuming enzymes, and the exchange rate between the individual subcellular compartments. In this review, we discuss the NAD synthesis pathways, the NAD degradation enzymes, and NAD subcellular localization, as well as NAD transport mechanisms with a focus on mitochondria. Finally, the effect of the pathologic depletion of mitochondrial NAD pools on mitochondrial proteins' post-translational modifications and its role in neurodegeneration will be reviewed. Understanding the physiological constraints and mechanisms of NAD maintenance and the exchange between subcellular compartments is critical given NAD's broad effects and roles in health and disease.
Collapse
Affiliation(s)
- Jaylyn Waddell
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Rehana Khatoon
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Tibor Kristian
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Veterans Affairs Maryland Health Center System, 10 North Greene Street, Baltimore, MD 21201, USA
| |
Collapse
|
9
|
Chen L, Chen M, Luo M, Li Y, Liao B, Hu M, Yu Q. Ratiometric NAD + Sensors Reveal Subcellular NAD + Modulators. ACS Sens 2023; 8:1518-1528. [PMID: 36931900 DOI: 10.1021/acssensors.2c02565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
Mapping NAD+ dynamics in live cells and human is essential for translating NAD+ interventions into effective therapies. Yet, genetically encoded NAD+ sensors with better specificity and pH resistance are still needed for the cost-effective monitoring of NAD+ in both subcellular compartments and clinical samples. Here, we introduce multicolor, resonance energy transfer-based NAD+ sensors covering nano- to millimolar concentration ranges for clinical NAD+ measurement and subcellular NAD+ visualization. The sensors captured the blood NAD+ increase induced by NMN supplementation and revealed the distinct subcellular effects of NAD+ precursors and modulators. The sensors then enabled high-throughput screenings for mitochondrial and nuclear NAD+ modulators and identified α-GPC, a cognition-related metabolite that induces NAD+ redistribution from mitochondria to the nucleus relative to the total adenine nucleotides, which was further confirmed by NAD+ FRET microscopy.
Collapse
Affiliation(s)
- Liuqing Chen
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,Shenzhen Key Laboratory for the Intelligent Microbial Manufacturing of Medicines, Shenzhen 518055, China
| | - Meiting Chen
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mupeng Luo
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yong Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Bagen Liao
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, Guangzhou 510150, China
| | - Min Hu
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, Guangzhou 510150, China
| | - Qiuliyang Yu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,Shenzhen Key Laboratory for the Intelligent Microbial Manufacturing of Medicines, Shenzhen 518055, China
| |
Collapse
|
10
|
Muskalla L, Güldenpfennig A, Hottiger MO. Subcellular Quantitation of ADP-Ribosylation by High-Content Microscopy. Methods Mol Biol 2022; 2609:101-109. [PMID: 36515832 DOI: 10.1007/978-1-0716-2891-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ADP-ribosylation is a posttranslational modification with many functions ranging from the DNA damage response to transcriptional regulation. While nuclear ADP-ribosylation has been extensively studied in the context of genotoxic stress mediated by PARP1, signaling by other members of the family and in other cellular compartments is still not as well understood. In recent years, however, progress has been made with the development of new tools for detection of ADP-ribosylation by immunofluorescence, which allows for a spatial differentiation of signal intensity for different cellular compartments. Here, we present our method for the detection and quantification of compartment-specific ADP-ribosylation by immunofluorescence and show why the engineered macrodomain eAf5121 might be the best tool to date.
Collapse
Affiliation(s)
- Lukas Muskalla
- Department of Molecular Mechanisms of Disease, Vetsuisse Faculty and Faculty of Science, University of Zurich, Zurich, Switzerland.,Life Science Zurich Graduate School, Cancer Biology PhD program, University of Zurich, Zurich, Switzerland
| | - Anka Güldenpfennig
- Department of Molecular Mechanisms of Disease, Vetsuisse Faculty and Faculty of Science, University of Zurich, Zurich, Switzerland.,Life Science Zurich Graduate School, Molecular Life Science PhD program, University of Zurich, Zurich, Switzerland
| | - Michael O Hottiger
- Department of Molecular Mechanisms of Disease, Vetsuisse Faculty and Faculty of Science, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
11
|
Chen W, Liu S, Yang Y, Zhang Z, Zhao Y. Spatiotemporal Monitoring of NAD+ Metabolism with Fluorescent Biosensors. Mech Ageing Dev 2022; 204:111657. [DOI: 10.1016/j.mad.2022.111657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 03/09/2022] [Accepted: 03/09/2022] [Indexed: 01/07/2023]
|
12
|
McReynolds MR, Chellappa K, Chiles E, Jankowski C, Shen Y, Chen L, Descamps HC, Mukherjee S, Bhat YR, Lingala SR, Chu Q, Botolin P, Hayat F, Doke T, Susztak K, Thaiss CA, Lu W, Migaud ME, Su X, Rabinowitz JD, Baur JA. NAD + flux is maintained in aged mice despite lower tissue concentrations. Cell Syst 2021; 12:1160-1172.e4. [PMID: 34559996 DOI: 10.1016/j.cels.2021.09.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 06/08/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022]
Abstract
NAD+ is an essential coenzyme for all living cells. NAD+ concentrations decline with age, but whether this reflects impaired production or accelerated consumption remains unclear. We employed isotope tracing and mass spectrometry to probe age-related changes in NAD+ metabolism across tissues. In aged mice, we observed modest tissue NAD+ depletion (median decrease ∼30%). Circulating NAD+ precursors were not significantly changed, and isotope tracing showed the unimpaired synthesis of nicotinamide from tryptophan. In most tissues of aged mice, turnover of the smaller tissue NAD+ pool was modestly faster such that absolute NAD+ biosynthetic flux was maintained, consistent with more active NAD+-consuming enzymes. Calorie restriction partially mitigated age-associated NAD+ decline by decreasing consumption. Acute inflammatory stress induced by LPS decreased NAD+ by impairing synthesis in both young and aged mice. Thus, the decline in NAD+ with normal aging is relatively subtle and occurs despite maintained NAD+ production, likely due to increased consumption.
Collapse
Affiliation(s)
- Melanie R McReynolds
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Karthikeyani Chellappa
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric Chiles
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Connor Jankowski
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Yihui Shen
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Li Chen
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Hélène C Descamps
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarmistha Mukherjee
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yashaswini R Bhat
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Siddharth R Lingala
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qingwei Chu
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Botolin
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Faisal Hayat
- Department of Pharmacology, Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Tomohito Doke
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
| | - Christoph A Thaiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wenyun Lu
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Marie E Migaud
- Department of Pharmacology, Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Xiaoyang Su
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Chemistry, Princeton University, Princeton, NJ, USA.
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Strømland Ø, Diab J, Ferrario E, Sverkeli LJ, Ziegler M. The balance between NAD + biosynthesis and consumption in ageing. Mech Ageing Dev 2021; 199:111569. [PMID: 34509469 DOI: 10.1016/j.mad.2021.111569] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/18/2021] [Accepted: 09/08/2021] [Indexed: 01/07/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a vital coenzyme in redox reactions. NAD+ is also important in cellular signalling as it is consumed by PARPs, SARM1, sirtuins and CD38. Cellular NAD+ levels regulate several essential processes including DNA repair, immune cell function, senescence, and chromatin remodelling. Maintenance of these cellular processes is important for healthy ageing and lifespan. Interestingly, the levels of NAD+ decline during ageing in several organisms, including humans. Declining NAD+ levels have been linked to several age-related diseases including various metabolic diseases and cognitive decline. Decreasing tissue NAD+ concentrations have been ascribed to an imbalance between biosynthesis and consumption of the dinucleotide, resulting from, for instance, reduced levels of the rate limiting enzyme NAMPT along with an increased activation state of the NAD+-consuming enzymes PARPs and CD38. The progression of some age-related diseases can be halted or reversed by therapeutic augmentation of NAD+ levels. NAD+ metabolism has therefore emerged as a potential target to ameliorate age-related diseases. The present review explores how ageing affects NAD+ metabolism and current approaches to reverse the age-dependent decline of NAD+.
Collapse
Affiliation(s)
- Øyvind Strømland
- Department of Biomedicine, University of Bergen, Bergen, 5009, Norway
| | - Joseph Diab
- Department of Biomedicine, University of Bergen, Bergen, 5009, Norway
| | - Eugenio Ferrario
- Department of Biomedicine, University of Bergen, Bergen, 5009, Norway
| | - Lars J Sverkeli
- Department of Biomedicine, University of Bergen, Bergen, 5009, Norway; Department of Biological Sciences, University of Bergen, Bergen, 5020, Norway
| | - Mathias Ziegler
- Department of Biomedicine, University of Bergen, Bergen, 5009, Norway.
| |
Collapse
|
14
|
Welcome to the Family: Identification of the NAD + Transporter of Animal Mitochondria as Member of the Solute Carrier Family SLC25. Biomolecules 2021; 11:biom11060880. [PMID: 34198503 PMCID: PMC8231866 DOI: 10.3390/biom11060880] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/01/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023] Open
Abstract
Subcellular compartmentation is a fundamental property of eukaryotic cells. Communication and metabolic and regulatory interconnectivity between organelles require that solutes can be transported across their surrounding membranes. Indeed, in mammals, there are hundreds of genes encoding solute carriers (SLCs) which mediate the selective transport of molecules such as nucleotides, amino acids, and sugars across biological membranes. Research over many years has identified the localization and preferred substrates of a large variety of SLCs. Of particular interest has been the SLC25 family, which includes carriers embedded in the inner membrane of mitochondria to secure the supply of these organelles with major metabolic intermediates and coenzymes. The substrate specificity of many of these carriers has been established in the past. However, the route by which animal mitochondria are supplied with NAD+ had long remained obscure. Only just recently, the existence of a human mitochondrial NAD+ carrier was firmly established. With the realization that SLC25A51 (or MCART1) represents the major mitochondrial NAD+ carrier in mammals, a long-standing mystery in NAD+ biology has been resolved. Here, we summarize the functional importance and structural features of this carrier as well as the key observations leading to its discovery.
Collapse
|
15
|
Hopp AK, Hottiger MO. Uncovering the Invisible: Mono-ADP-ribosylation Moved into the Spotlight. Cells 2021; 10:680. [PMID: 33808662 PMCID: PMC8003356 DOI: 10.3390/cells10030680] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
Adenosine diphosphate (ADP)-ribosylation is a nicotinamide adenine dinucleotide (NAD+)-dependent post-translational modification that is found on proteins as well as on nucleic acids. While ARTD1/PARP1-mediated poly-ADP-ribosylation has extensively been studied in the past 60 years, comparably little is known about the physiological function of mono-ADP-ribosylation and the enzymes involved in its turnover. Promising technological advances have enabled the development of innovative tools to detect NAD+ and NAD+/NADH (H for hydrogen) ratios as well as ADP-ribosylation. These tools have significantly enhanced our current understanding of how intracellular NAD dynamics contribute to the regulation of ADP-ribosylation as well as to how mono-ADP-ribosylation integrates into various cellular processes. Here, we discuss the recent technological advances, as well as associated new biological findings and concepts.
Collapse
Affiliation(s)
| | - Michael O. Hottiger
- Department of Molecular Mechanisms of Disease (DMMD), University of Zurich, 8057 Zurich, Switzerland;
| |
Collapse
|
16
|
Köritzer J, Blenn C, Bürkle A, Beneke S. Mitochondria are devoid of poly(ADP-ribose)polymerase-1, but harbor its product oligo(ADP-ribose). J Cell Biochem 2021; 122:507-523. [PMID: 33417272 DOI: 10.1002/jcb.29887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/28/2022]
Abstract
There are conflicting data about localization of poly(ADP-ribose)polymerase-1 and its product poly(ADP-ribose) in mitochondria. To finally clarify the discussion, we investigated with biochemical and cell biological methods the potential presence of poly(ADP-ribose) polymerase-1 in these organelles. Our data show that endogenous and overexpressed poly(ADP-ribose)polymerase 1 is only localized to the nucleus with a clear exclusion of cytosolic compartments. In addition, highly purified mitochondria devoid of nuclear contaminations do not contain poly(ADP-ribose)polymerase-1. Although no poly(ADP-ribose)polymerase-1 enzyme is detectable in mitochondria, a shorter variant of its product poly(ADP-ribose) is present, associated specifically with a small subset of mitochondrial proteins as revealed by immunoprecipitation and protein fingerprint analysis. These proteins are located at key-points of the Krebs-cycle, are chaperones involved in mitochondrial functionality and quality-control, and are RNA-binding proteins important for transcript stability, respectively. Of note, despite the fact that especially poly(ADP-ribose)polymerase-1 is its own major target for modification, we could not detect this enzyme by mass spectrometry in these organelles. These data suggests a new way of targeted nuclear-mitochondrial signaling, mediated by nuclear poly(ADP-ribosyl)ation dependent on poly(ADP-ribose)polymerase-1.
Collapse
Affiliation(s)
- Julia Köritzer
- Molecular Toxicology Group, University of Konstanz, Konstanz, Germany
| | - Christian Blenn
- Institute of Pharmacology and Toxicology, University of Zurich/Vetsuisse, Zurich, Switzerland
| | - Alexander Bürkle
- Molecular Toxicology Group, University of Konstanz, Konstanz, Germany
| | - Sascha Beneke
- Molecular Toxicology Group, University of Konstanz, Konstanz, Germany.,Human and Environmental Toxicology Group, University of Konstanz, Konstanz, Germany
| |
Collapse
|
17
|
Navas LE, Carnero A. NAD + metabolism, stemness, the immune response, and cancer. Signal Transduct Target Ther 2021; 6:2. [PMID: 33384409 PMCID: PMC7775471 DOI: 10.1038/s41392-020-00354-w] [Citation(s) in RCA: 267] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/11/2020] [Accepted: 09/27/2020] [Indexed: 02/07/2023] Open
Abstract
NAD+ was discovered during yeast fermentation, and since its discovery, its important roles in redox metabolism, aging, and longevity, the immune system and DNA repair have been highlighted. A deregulation of the NAD+ levels has been associated with metabolic diseases and aging-related diseases, including neurodegeneration, defective immune responses, and cancer. NAD+ acts as a cofactor through its interplay with NADH, playing an essential role in many enzymatic reactions of energy metabolism, such as glycolysis, oxidative phosphorylation, fatty acid oxidation, and the TCA cycle. NAD+ also plays a role in deacetylation by sirtuins and ADP ribosylation during DNA damage/repair by PARP proteins. Finally, different NAD hydrolase proteins also consume NAD+ while converting it into ADP-ribose or its cyclic counterpart. Some of these proteins, such as CD38, seem to be extensively involved in the immune response. Since NAD cannot be taken directly from food, NAD metabolism is essential, and NAMPT is the key enzyme recovering NAD from nicotinamide and generating most of the NAD cellular pools. Because of the complex network of pathways in which NAD+ is essential, the important role of NAD+ and its key generating enzyme, NAMPT, in cancer is understandable. In the present work, we review the role of NAD+ and NAMPT in the ways that they may influence cancer metabolism, the immune system, stemness, aging, and cancer. Finally, we review some ongoing research on therapeutic approaches.
Collapse
Affiliation(s)
- Lola E Navas
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain.,CIBER de Cancer, Sevilla, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Sevilla, Spain. .,CIBER de Cancer, Sevilla, Spain.
| |
Collapse
|
18
|
Luongo TS, Eller JM, Lu MJ, Niere M, Raith F, Perry C, Bornstein MR, Oliphint P, Wang L, McReynolds MR, Migaud ME, Rabinowitz JD, Johnson FB, Johnsson K, Ziegler M, Cambronne XA, Baur JA. SLC25A51 is a mammalian mitochondrial NAD + transporter. Nature 2020; 588:174-179. [PMID: 32906142 PMCID: PMC7718333 DOI: 10.1038/s41586-020-2741-7] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 09/01/2020] [Indexed: 12/11/2022]
Abstract
Mitochondria require nicotinamide adenine dinucleotide (NAD+) in order to carry out the fundamental processes that fuel respiration and mediate cellular energy transduction. Mitochondrial NAD+ transporters have been identified in yeast and plants 1,2 but their very existence is controversial in mammals 3–5. Here we demonstrate that mammalian mitochondria are capable of taking up intact NAD+ and identify SLC25A51 (an essential 6,7 mitochondrial protein of previously unknown function, also known as MCART1) as a mammalian mitochondrial NAD+ transporter. Loss of SLC25A51 decreases mitochondrial but not whole-cell NAD+ content, impairs mitochondrial respiration, and blocks the uptake of NAD+ into isolated mitochondria. Conversely, overexpression of SLC25A51 or a nearly identical paralog, SLC25A52, increases mitochondrial NAD+ levels and restores NAD+ uptake into yeast mitochondria lacking endogenous NAD+ transporters. Together, these findings identify SLC25A51 as the first transporter capable of importing NAD+ into mammalian mitochondria.
Collapse
Affiliation(s)
- Timothy S Luongo
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jared M Eller
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Mu-Jie Lu
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Marc Niere
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Fabio Raith
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany.,Faculty of Chemistry and Earth Sciences, University of Heidelberg, Heidelberg, Germany
| | - Caroline Perry
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marc R Bornstein
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul Oliphint
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Lin Wang
- Lewis-Sigler Institute for Integrative Genomics, Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Melanie R McReynolds
- Lewis-Sigler Institute for Integrative Genomics, Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Marie E Migaud
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - F Brad Johnson
- Department of Pathology and Laboratory Medicine, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kai Johnsson
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany.,Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Mathias Ziegler
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Xiaolu A Cambronne
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA.
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Minireview Exploring the Biological Cycle of Vitamin B3 and Its Influence on Oxidative Stress: Further Molecular and Clinical Aspects. Molecules 2020; 25:molecules25153323. [PMID: 32707945 PMCID: PMC7436124 DOI: 10.3390/molecules25153323] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/13/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Vitamin B3, or niacin, is one of the most important compounds of the B-vitamin complex. Recent reports have demonstrated the involvement of vitamin B3 in a number of pivotal functions which ensure that homeostasis is maintained. In addition, the intriguing nature of its synthesis and the underlying mechanism of action of vitamin B3 have encouraged further studies aimed at deepening our understanding of the close link between the exogenous supply of B3 and how it activates dependent enzymes. This crucial role can be attributed to the gut microflora and its ability to shape human behavior and development by mediating the bioavailability of metabolites. Recent studies have indicated a possible interconnection between the novel coronavirus and commensal bacteria. As such, we have attempted to explain how the gastrointestinal deficiencies displayed by SARS-CoV-2-infected patients arise. It seems that the stimulation of a proinflammatory cascade and the production of large amounts of reactive oxygen species culminates in the subsequent loss of host eubiosis. Studies of the relationhip between ROS, SARS-CoV-2, and gut flora are sparse in the current literature. As an integrated component, oxidative stress (OS) has been found to negatively influence host eubiosis, in vitro fertilization outcomes, and oocyte quality, but to act as a sentinel against infections. In conclusion, research suggests that in the future, a healthy diet may be considered a reliable tool for maintaining and optimizing our key internal parameters.
Collapse
|
20
|
Cambronne XA, Kraus WL. Location, Location, Location: Compartmentalization of NAD + Synthesis and Functions in Mammalian Cells. Trends Biochem Sci 2020; 45:858-873. [PMID: 32595066 DOI: 10.1016/j.tibs.2020.05.010] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/06/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
The numerous biological roles of NAD+ are organized and coordinated via its compartmentalization within cells. The spatial and temporal partitioning of this intermediary metabolite is intrinsic to understanding the impact of NAD+ on cellular signaling and metabolism. We review evidence supporting the compartmentalization of steady-state NAD+ levels in cells, as well as how the modulation of NAD+ synthesis dynamically regulates signaling by controlling subcellular NAD+ concentrations. We further discuss potential benefits to the cell of compartmentalizing NAD+, and methods for measuring subcellular NAD+ levels.
Collapse
Affiliation(s)
- Xiaolu A Cambronne
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA.
| | - W Lee Kraus
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
21
|
Van Veldhoven PP, de Schryver E, Young SG, Zwijsen A, Fransen M, Espeel M, Baes M, Van Ael E. Slc25a17 Gene Trapped Mice: PMP34 Plays a Role in the Peroxisomal Degradation of Phytanic and Pristanic Acid. Front Cell Dev Biol 2020; 8:144. [PMID: 32266253 PMCID: PMC7106852 DOI: 10.3389/fcell.2020.00144] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/20/2020] [Indexed: 12/04/2022] Open
Abstract
Mice lacking PMP34, a peroxisomal membrane transporter encoded by Slc25a17, did not manifest any obvious phenotype on a Swiss Webster genetic background, even with various treatments designed to unmask impaired peroxisomal functioning. Peroxisomal α- and β-oxidation rates in PMP34 deficient fibroblasts or liver slices were not or only modestly affected and in bile, no abnormal bile acid intermediates were detected. Peroxisomal content of cofactors like CoA, ATP, NAD+, thiamine-pyrophosphate and pyridoxal-phosphate, based on direct or indirect data, appeared normal as were tissue plasmalogen and very long chain fatty acid levels. However, upon dietary phytol administration, the knockout mice displayed hepatomegaly, liver inflammation, and an induction of peroxisomal enzymes. This phenotype was partially mediated by PPARα. Hepatic triacylglycerols and cholesterylesters were elevated and both phytanic acid and pristanic acid accumulated in the liver lipids, in females to higher extent than in males. In addition, pristanic acid degradation products were detected, as wells as the CoA-esters of all these branched fatty acids. Hence, PMP34 is important for the degradation of phytanic/pristanic acid and/or export of their metabolites. Whether this is caused by a shortage of peroxisomal CoA affecting the intraperoxisomal formation of pristanoyl-CoA (and perhaps of phytanoyl-CoA), or the SCPx-catalyzed thiolytic cleavage during pristanic acid β-oxidation, could not be proven in this model, but the phytol-derived acyl-CoA profile is compatible with the latter possibility. On the other hand, the normal functioning of other peroxisomal pathways, and especially bile acid formation, seems to exclude severe transport problems or a shortage of CoA, and other cofactors like FAD, NAD(P)+, TPP. Based on our findings, PMP34 deficiency in humans is unlikely to be a life threatening condition but could cause elevated phytanic/pristanic acid levels in older adults.
Collapse
Affiliation(s)
| | - Evelyn de Schryver
- LIPIT, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stephen G. Young
- Departments of Medicine and Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - An Zwijsen
- Laboratory of Developmental Signaling, Department Human Genetics, VIB-KU Leuven, Leuven, Belgium
| | - Marc Fransen
- LIPIT, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Marc Espeel
- Department of Anatomy, Embryology, Histology and Medical Physics, Ghent University, Ghent, Belgium
| | - Myriam Baes
- Laboratory of Cell Metabolism, Faculty of Pharmaceutical Sciences, KU Leuven, Leuven, Belgium
| | - Elke Van Ael
- LIPIT, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
22
|
Gilmour BC, Gudmundsrud R, Frank J, Hov A, Lautrup S, Aman Y, Røsjø H, Brenner C, Ziegler M, Tysnes OB, Tzoulis C, Omland T, Søraas A, Holmøy T, Bergersen LH, Storm-Mathisen J, Nilsen H, Fang EF. Targeting NAD + in translational research to relieve diseases and conditions of metabolic stress and ageing. Mech Ageing Dev 2020; 186:111208. [PMID: 31953124 DOI: 10.1016/j.mad.2020.111208] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 02/07/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) plays a fundamental role in life and health through the regulation of energy biogenesis, redox homeostasis, cell metabolism, and the arbitration of cell survival via linkages to apoptosis and autophagic pathways. The importance of NAD+ in ageing and healthy longevity has been revealed from laboratory animal studies and early-stage clinical testing. While basic researchers and clinicians have investigated the molecular mechanisms and translation potential of NAD+, there are still major gaps in applying laboratory science to design the most effective trials. This mini-review was based on the programme and discussions of the 3rd NO-Age Symposium held at the Akershus University Hospital, Norway on the 28th October 2019. This symposium brought together leading basic researchers on NAD+ and clinicians who are leading or are going to perform NAD+ augmentation-related clinical studies. This meeting covered talks about NAD+ synthetic pathways, subcellular homeostasis of NAD+, the benefits of NAD+ augmentation from maternal milk to offspring, current clinical trials of the NAD+ precursor nicotinamide riboside (NR) on Ataxia-Telangiectasia (A-T), Parkinson's disease (PD), post-sepsis fatigue, as well as other potential NR-based clinical trials. Importantly, a consensus is emerging with respect to the design of clinical trials in order to measure meaningful parameters and ensure safety.
Collapse
Affiliation(s)
- Brian C Gilmour
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| | | | - Johannes Frank
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - Amund Hov
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| | - Sofie Lautrup
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - Yahyah Aman
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - Helge Røsjø
- Division of Research and Innovation, Akershus University Hospital, 1478 Lørenskog, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mathias Ziegler
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Ole-Bjørn Tysnes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Charalampos Tzoulis
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Torbjørn Omland
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cardiology, Division of Medicine, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Arne Søraas
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Trygve Holmøy
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway; Department of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Linda H Bergersen
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway; The Brain and Muscle Energy Group, Electron Microscopy Laboratory, Department of Oral Biology, University of Oslo, NO-0316 Oslo, Norway; Synaptic Neurochemistry and Amino Acid Transporters Labs, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences (IMB) and Healthy Brain Ageing Centre (SERTA), University of Oslo, NO-0317 Oslo, Norway; Center for Healthy Ageing, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Jon Storm-Mathisen
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway; Synaptic Neurochemistry and Amino Acid Transporters Labs, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences (IMB) and Healthy Brain Ageing Centre (SERTA), University of Oslo, NO-0317 Oslo, Norway
| | - Hilde Nilsen
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway; The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| | - Evandro F Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway; The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway.
| |
Collapse
|
23
|
Zhou H, Nguyen L, Arnesano C, Ando Y, Raval M, Rodgers JT, Fraser S, Lu R, Shen K. Non-invasive Optical Biomarkers Distinguish and Track the Metabolic Status of Single Hematopoietic Stem Cells. iScience 2020; 23:100831. [PMID: 31982780 PMCID: PMC6994633 DOI: 10.1016/j.isci.2020.100831] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/11/2019] [Accepted: 01/07/2020] [Indexed: 12/28/2022] Open
Abstract
Metabolism is a key regulator of hematopoietic stem cell (HSC) functions. There is a lack of real-time, non-invasive approaches to evaluate metabolism in single HSCs. Using fluorescence lifetime imaging microscopy, we developed a set of metabolic optical biomarkers (MOBs) from the auto-fluorescent properties of metabolic coenzymes NAD(P)H and FAD. The MOBs revealed the enhanced glycolysis, low oxidative metabolism, and distinct mitochondrial localization of HSCs. Importantly, the fluorescence lifetime of enzyme-bound NAD(P)H (τbound) can non-invasively monitor the glycolytic/lactate dehydrogenase activity in single HSCs. As a proof of concept for metabolism-based cell sorting, we further identified HSCs within the Lineage-cKit+Sca1+ (KLS) hematopoietic stem/progenitor population using MOBs and a machine-learning algorithm. Moreover, we revealed the dynamic changes of MOBs, and the association of longer τbound with enhanced glycolysis under HSC stemness-maintaining conditions during HSC culture. Our work thus provides a new paradigm to identify and track the metabolism of single HSCs non-invasively and in real time. Metabolic optical biomarkers non-invasively distinguish HSCs from early progenitors NAD(P)H τbound reflects lactate dehydrogenase activity in single fresh/cultured HSCs pHi correlates with τbound in hematopoietic populations, with HSCs being the highest Optical biomarkers track metabolic changes and response to drugs in cultured HSCs
Collapse
Affiliation(s)
- Hao Zhou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Lisa Nguyen
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cosimo Arnesano
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA; Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Yuta Ando
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Manmeet Raval
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Joseph T Rodgers
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Scott Fraser
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA; Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA; Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Rong Lu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; USC Stem Cell, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
24
|
Abstract
NAD+ is a pivotal metabolite involved in cellular bioenergetics, genomic stability, mitochondrial homeostasis, adaptive stress responses, and cell survival. Multiple NAD+-dependent enzymes are involved in synaptic plasticity and neuronal stress resistance. Here, we review emerging findings that reveal key roles for NAD+ and related metabolites in the adaptation of neurons to a wide range of physiological stressors and in counteracting processes in neurodegenerative diseases, such as those occurring in Alzheimer's, Parkinson's, and Huntington diseases, and amyotrophic lateral sclerosis. Advances in understanding the molecular and cellular mechanisms of NAD+-based neuronal resilience will lead to novel approaches for facilitating healthy brain aging and for the treatment of a range of neurological disorders.
Collapse
Affiliation(s)
- Sofie Lautrup
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - David A Sinclair
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Evandro F Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway; The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway.
| |
Collapse
|
25
|
The DNA-damage response and nuclear events as regulators of nonapoptotic forms of cell death. Oncogene 2019; 39:1-16. [PMID: 31462710 DOI: 10.1038/s41388-019-0980-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 08/05/2019] [Accepted: 08/09/2019] [Indexed: 12/20/2022]
Abstract
The maintenance of genome stability is essential for the cell as the integrity of genomic information guaranties reproduction of a whole organism. DNA damage occurring in response to different natural and nonnatural stimuli (errors in DNA replication, UV radiation, chemical agents, etc.) is normally detected by special cellular machinery that induces DNA repair. However, further accumulation of genetic lesions drives the activation of cell death to eliminate cells with defective genome. This particular feature is used for targeting fast-proliferating tumor cells during chemo-, radio-, and immunotherapy. Among different cell death modalities induced by DNA damage, apoptosis is the best studied. Nevertheless, nonapoptotic cell death and adaptive stress responses are also activated following genotoxic stress and play a crucial role in the outcome of anticancer therapy. Here, we provide an overview of nonapoptotic cell death pathways induced by DNA damage and discuss their interplay with cellular senescence, mitotic catastrophe, and autophagy.
Collapse
|
26
|
Hopp AK, Grüter P, Hottiger MO. Regulation of Glucose Metabolism by NAD + and ADP-Ribosylation. Cells 2019; 8:cells8080890. [PMID: 31412683 PMCID: PMC6721828 DOI: 10.3390/cells8080890] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/09/2019] [Accepted: 08/11/2019] [Indexed: 12/28/2022] Open
Abstract
Cells constantly adapt their metabolic pathways to meet their energy needs and respond to nutrient availability. During the last two decades, it has become increasingly clear that NAD+, a coenzyme in redox reactions, also mediates several ubiquitous cell signaling processes. Protein ADP-ribosylation is a post-translational modification that uses NAD+ as a substrate and is best known as part of the genotoxic stress response. However, there is increasing evidence that NAD+-dependent ADP-ribosylation regulates other cellular processes, including metabolic pathways. In this review, we will describe the compartmentalized regulation of NAD+ biosynthesis, consumption, and regeneration with a particular focus on the role of ADP-ribosylation in the regulation of glucose metabolism in different cellular compartments.
Collapse
Affiliation(s)
- Ann-Katrin Hopp
- Department of Molecular Mechanisms of Disease (DMMD), University of Zurich, CH-8057 Zurich, Switzerland
- Molecular Life Science Ph.D. Program, Life Science Zurich Graduate School, CH-8057 Zurich, Switzerland
| | - Patrick Grüter
- Department of Molecular Mechanisms of Disease (DMMD), University of Zurich, CH-8057 Zurich, Switzerland
| | - Michael O Hottiger
- Department of Molecular Mechanisms of Disease (DMMD), University of Zurich, CH-8057 Zurich, Switzerland.
| |
Collapse
|
27
|
Zhu Y, Liu J, Park J, Rai P, Zhai RG. Subcellular compartmentalization of NAD + and its role in cancer: A sereNADe of metabolic melodies. Pharmacol Ther 2019; 200:27-41. [PMID: 30974124 PMCID: PMC7010080 DOI: 10.1016/j.pharmthera.2019.04.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/02/2019] [Indexed: 02/07/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential biomolecule involved in many critical processes. Its role as both a driver of energy production and a signaling molecule underscores its importance in health and disease. NAD+ signaling impacts multiple processes that are dysregulated in cancer, including DNA repair, cell proliferation, differentiation, redox regulation, and oxidative stress. Distribution of NAD+ is highly compartmentalized, with each subcellular NAD+ pool differentially regulated and preferentially involved in distinct NAD+-dependent signaling or metabolic events. Emerging evidence suggests that targeting NAD+ metabolism is likely to repress many specific mechanisms underlying tumor development and progression, including proliferation, survival, metabolic adaptations, invasive capabilities, heterotypic interactions with the tumor microenvironment, and stress response including notably DNA maintenance and repair. Here we provide a comprehensive overview of how compartmentalized NAD+ metabolism in mitochondria, nucleus, cytosol, and extracellular space impacts cancer formation and progression, along with a discussion of the therapeutic potential of NAD+-targeting drugs in cancer.
Collapse
Affiliation(s)
- Yi Zhu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, China; Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jiaqi Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, China
| | - Joun Park
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Priyamvada Rai
- Department of Medicine/Medical Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rong G Zhai
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, China.
| |
Collapse
|
28
|
Osuagwu N, Dölle C, Tzoulis C. Poly-ADP-ribose assisted protein localization resolves that DJ-1, but not LRRK2 or α-synuclein, is localized to the mitochondrial matrix. PLoS One 2019; 14:e0219909. [PMID: 31323073 PMCID: PMC6641658 DOI: 10.1371/journal.pone.0219909] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 07/04/2019] [Indexed: 12/21/2022] Open
Abstract
Several proteins linked to familial Parkinson disease have been associated with mitochondrial (dys-)function and have been described to reside within mitochondria. The putative mitochondrial and sub-mitochondrial localization of these proteins remains disputed, however, potentially due to conflicting results obtained by diverging technical approaches. Using the high-resolution poly-ADP-ribose assisted protein localization assay that also allows for detection of low level and even partial mitochondrial matrix localization, we demonstrate here that DJ-1, but not LRRK2 or α-synuclein, resides in the mitochondrial matrix. The localization of the proteins was not changed in cellular stress models of Parkinson disease and, in case of α-synuclein, not affected by pathological mutations. Our results verify the ability of DJ-1 to carry out its role also from within mitochondria and suggest that LRRK2 and α-synuclein may interact with and affect mitochondria from outside the mitochondrial matrix.
Collapse
Affiliation(s)
- Nelson Osuagwu
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Neuro-SysMed Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital and University of Bergen, Bergen, Norway
| | - Christian Dölle
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Neuro-SysMed Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital and University of Bergen, Bergen, Norway
- * E-mail: (CD); (CT)
| | - Charalampos Tzoulis
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Neuro-SysMed Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital and University of Bergen, Bergen, Norway
- * E-mail: (CD); (CT)
| |
Collapse
|
29
|
Bugaj O, Zieliński J, Kusy K, Kantanista A, Wieliński D, Guzik P. The Effect of Exercise on the Skin Content of the Reduced Form of NAD and Its Response to Transient Ischemia and Reperfusion in Highly Trained Athletes. Front Physiol 2019; 10:600. [PMID: 31156467 PMCID: PMC6529559 DOI: 10.3389/fphys.2019.00600] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/26/2019] [Indexed: 12/22/2022] Open
Abstract
Reduced nicotinamide adenine dinucleotide (NADH) is synthesized in the cellular nucleus, cytoplasm and mitochondria but oxidized into NAD+ almost exclusively in mitochondria. Activation of human skin by the 340 nm ultraviolet light triggers natural fluorescence at the light length of 460 nm, which intensity is proportional to the skin NADH content. This phenomenon is used by the Flow Mediated Skin Fluorescence (FMSF) which measures changes in the skin NADH content during transient ischemia and reperfusion. We examined the effects of exercise to exhaustion on the skin changes of NADH in response to 200 s forearm ischemia and reperfusion in 121 highly trained athletes (94 men and 27 women, long-distance running, triathlon, taekwondo, rowing, futsal, sprint running, fencing, and tennis). We found that exercise until exhaustion changes the skin content of NADH, modifies NADH turnover at rest, during ischemia and reperfusion in the most superficial living skin cells. Compared to the pre-exercise, there were significant increases in: mean fluorescence recorded during rest as the baseline value (B mean) (p < 0.001), the maximal fluorescence that increased above the baseline during controlled forearm ischemia (FImax) (p < 0.001, only in men), the minimal fluorescence after decreasing below the baseline during reperfusion (FRmin) (p < 0.001 men; p < 0.01 women) and the difference between B mean and FRmin (R min) (p < 0.01), and reductions in the difference between FImax and B mean (I max) (p < 0.001) and I max/IRampl ratio (CImax) (p < 0.001) after the incremental exercise test. There was no statistical difference between pre- and post-exercise the maximal range of the fluorescence change during ischemia and reperfusion (IRampl). In conclusion, exercise to exhaustion modifies the skin NADH content at rest, during ischemia and reperfusion as well as the magnitude of changes in the NADH caused by ischemia and reperfusion. Our findings suggest that metabolic changes in the skin NADH accompanying exercise extend beyond muscles and affect other cells and organs.
Collapse
Affiliation(s)
- Olga Bugaj
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Jacek Zieliński
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Krzysztof Kusy
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Adam Kantanista
- Department of Sport Kinesiology, Poznań University of Physical Education, Poznań, Poland
| | - Dariusz Wieliński
- Department of Anthropology and Biometry, Poznań University of Physical Education, Poznań, Poland
| | - Przemysław Guzik
- Department of Cardiology-Intensive Therapy, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
30
|
ADP-ribosylation and intracellular traffic: an emerging role for PARP enzymes. Biochem Soc Trans 2019; 47:357-370. [DOI: 10.1042/bst20180416] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/02/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022]
Abstract
AbstractADP-ribosylation is an ancient and reversible post-translational modification (PTM) of proteins, in which the ADP-ribose moiety is transferred from NAD+ to target proteins by members of poly-ADP-ribosyl polymerase (PARP) family. The 17 members of this family have been involved in a variety of cellular functions, where their regulatory roles are exerted through the modification of specific substrates, whose identification is crucial to fully define the contribution of this PTM. Evidence of the role of the PARPs is now available both in the context of physiological processes and of cell responses to stress or starvation. An emerging role of the PARPs is their control of intracellular transport, as it is the case for tankyrases/PARP5 and PARP12. Here, we discuss the evidence pointing at this novel aspect of PARPs-dependent cell regulation.
Collapse
|
31
|
Keeping the balance in NAD metabolism. Biochem Soc Trans 2019; 47:119-130. [PMID: 30626706 DOI: 10.1042/bst20180417] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 12/02/2018] [Accepted: 12/05/2018] [Indexed: 12/30/2022]
Abstract
Research over the last few decades has extended our understanding of nicotinamide adenine dinucleotide (NAD) from a vital redox carrier to an important signalling molecule that is involved in the regulation of a multitude of fundamental cellular processes. This includes DNA repair, cell cycle regulation, gene expression and calcium signalling, in which NAD is a substrate for several families of regulatory proteins, such as sirtuins and ADP-ribosyltransferases. At the molecular level, NAD-dependent signalling events differ from hydride transfer by cleavage of the dinucleotide into an ADP-ribosyl moiety and nicotinamide. Therefore, non-redox functions of NAD require continuous biosynthesis of the dinucleotide. Maintenance of cellular NAD levels is mainly achieved by nicotinamide salvage, yet a variety of other precursors can be used to sustain cellular NAD levels via different biosynthetic routes. Biosynthesis and consumption of NAD are compartmentalised at the subcellular level, and currently little is known about the generation and role of some of these subcellular NAD pools. Impaired biosynthesis or increased NAD consumption is deleterious and associated with ageing and several pathologies. Insults to neurons lead to depletion of axonal NAD and rapid degeneration, partial rescue can be achieved pharmacologically by administration of specific NAD precursors. Restoring NAD levels by stimulating biosynthesis or through supplementation with precursors also produces beneficial therapeutic effects in several disease models. In this review, we will briefly discuss the most recent achievements and the challenges ahead in this diverse research field.
Collapse
|
32
|
Orlandi I, Stamerra G, Vai M. Altered Expression of Mitochondrial NAD + Carriers Influences Yeast Chronological Lifespan by Modulating Cytosolic and Mitochondrial Metabolism. Front Genet 2018; 9:676. [PMID: 30619489 PMCID: PMC6305841 DOI: 10.3389/fgene.2018.00676] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 12/04/2018] [Indexed: 01/07/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) represents an essential cofactor in sustaining cellular bioenergetics and maintaining cellular fitness, and has emerged as a therapeutic target to counteract aging and age-related diseases. Besides NAD+ involvement in multiple redox reactions, it is also required as co-substrate for the activity of Sirtuins, a family of evolutionary conserved NAD+-dependent deacetylases that regulate both metabolism and aging. The founding member of this family is Sir2 of Saccharomyces cerevisiae, a well-established model system for studying aging of post-mitotic mammalian cells. In this context, it refers to chronological aging, in which the chronological lifespan (CLS) is measured. In this paper, we investigated the effects of changes in the cellular content of NAD+ on CLS by altering the expression of mitochondrial NAD+ carriers, namely Ndt1 and Ndt2. We found that the deletion or overexpression of these carriers alters the intracellular levels of NAD+ with opposite outcomes on CLS. In particular, lack of both carriers decreases NAD+ content and extends CLS, whereas NDT1 overexpression increases NAD+ content and reduces CLS. This correlates with opposite cytosolic and mitochondrial metabolic assets shown by the two types of mutants. In the former, an increase in the efficiency of oxidative phosphorylation is observed together with an enhancement of a pro-longevity anabolic metabolism toward gluconeogenesis and trehalose storage. On the contrary, NDT1 overexpression brings about on the one hand, a decrease in the respiratory efficiency generating harmful superoxide anions, and on the other, a decrease in gluconeogenesis and trehalose stores: all this is reflected into a time-dependent loss of mitochondrial functionality during chronological aging.
Collapse
Affiliation(s)
- Ivan Orlandi
- SYSBIO Centre for Systems Biology, Milan, Italy.,Dipartimento di Biotecnologie e Bioscienze, Università di Milano-Bicocca, Milan, Italy
| | - Giulia Stamerra
- Dipartimento di Biotecnologie e Bioscienze, Università di Milano-Bicocca, Milan, Italy
| | - Marina Vai
- Dipartimento di Biotecnologie e Bioscienze, Università di Milano-Bicocca, Milan, Italy
| |
Collapse
|
33
|
Eller JM, Stewart ML, Slepian AJ, Markwardt S, Wiedrick J, Cohen MS, Goodman RH, Cambronne XA. Flow Cytometry Analysis of Free Intracellular NAD + Using a Targeted Biosensor. ACTA ACUST UNITED AC 2018; 88:e54. [PMID: 30556645 DOI: 10.1002/cpcy.54] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Flow cytometry approaches combined with a genetically encoded targeted fluorescent biosensor are used to determine the subcellular compartmental availability of the oxidized form of nicotinamide adenine dinucleotide (NAD+ ). The availability of free NAD+ can affect the activities of NAD+ -consuming enzymes such as sirtuin, PARP/ARTD, and cyclic ADPR-hydrolase family members. Many methods for measuring the NAD+ available to these enzymes are limited because they cannot determine free NAD+ as it exists in various subcellular compartments distinctly from bound NAD+ or NADH. Here, an approach to express the sensor in mammalian cells, monitor NAD+ -dependent fluorescence intensity changes using flow cytometry approaches, and analyze data obtained is described. The benefit of flow cytometry approaches with the NAD+ sensor is the ability to monitor compartmentalized free NAD+ fluctuations simultaneously within many cells, which greatly facilitates analyses and calibration. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Jared M Eller
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas
| | - Melissa L Stewart
- Vollum Institute, Oregon Health & Science University, Portland, Oregon
| | | | - Sheila Markwardt
- Biostatistics and Design Program, Oregon Health & Science University, Portland, Oregon
| | - Jack Wiedrick
- Biostatistics and Design Program, Oregon Health & Science University, Portland, Oregon
| | - Michael S Cohen
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon
| | - Richard H Goodman
- Vollum Institute, Oregon Health & Science University, Portland, Oregon
| | - Xiaolu A Cambronne
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas
| |
Collapse
|
34
|
Kulikova VA, Gromyko DV, Nikiforov AA. The Regulatory Role of NAD in Human and Animal Cells. BIOCHEMISTRY (MOSCOW) 2018; 83:800-812. [PMID: 30200865 DOI: 10.1134/s0006297918070040] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD) and its phosphorylated form NADP are the major coenzymes in the redox reactions of various essential metabolic pathways. NAD+ also serves as a substrate for several families of regulatory proteins, such as protein deacetylases (sirtuins), ADP-ribosyltransferases, and poly(ADP-ribose) polymerases, that control vital cell processes including gene expression, DNA repair, apoptosis, mitochondrial biogenesis, unfolded protein response, and many others. NAD+ is also a precursor for calcium-mobilizing secondary messengers. Proper regulation of these NAD-dependent metabolic and signaling pathways depends on how efficiently cells can maintain their NAD levels. Generally, mammalian cells regulate their NAD supply through biosynthesis from the precursors delivered with the diet: nicotinamide and nicotinic acid (vitamin B3), as well as nicotinamide riboside and nicotinic acid riboside. Administration of NAD precursors has been demonstrated to restore NAD levels in tissues (i.e., to produce beneficial therapeutic effects) in preclinical models of various diseases, such as neurodegenerative disorders, obesity, diabetes, and metabolic syndrome.
Collapse
Affiliation(s)
- V A Kulikova
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia.,Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 195251, Russia
| | - D V Gromyko
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| | - A A Nikiforov
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia. .,Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 195251, Russia
| |
Collapse
|
35
|
VanLinden MR, Niere M, Nikiforov AA, Ziegler M, Dölle C. Compartment-Specific Poly-ADP-Ribose Formation as a Biosensor for Subcellular NAD Pools. Methods Mol Biol 2018; 1608:45-56. [PMID: 28695502 DOI: 10.1007/978-1-4939-6993-7_4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD) is vital to many cellular processes and is distributed between distinct subcellular pools in the compartmentalized eukaryotic cell. The detection and relative quantification of these individual pools is difficult because of the methods usually applied, which require cell disruption and fractionation.Here, we describe an immunochemical method to visualize and relatively quantify subcellular NAD+ pools, which relies on the NAD+-consuming activity of poly-ADP-ribose polymerase 1 (PARP1). We demonstrate that this system can be readily applied to detect changes in the mitochondrial, Golgi, endoplasmic reticulum, and peroxisomal NAD+ pools.
Collapse
Affiliation(s)
- Magali R VanLinden
- Department of Molecular Biology, University of Bergen, Thormøhlensgate 55, 5008, Bergen, Norway
| | - Marc Niere
- Department of Molecular Biology, University of Bergen, Thormøhlensgate 55, 5008, Bergen, Norway
| | - Andrey A Nikiforov
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia.,Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Mathias Ziegler
- Department of Molecular Biology, University of Bergen, Thormøhlensgate 55, 5008, Bergen, Norway.
| | - Christian Dölle
- Department of Neurology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
36
|
Abstract
Nicotinamide adenine dinucleotide (NAD), the cell's hydrogen carrier for redox enzymes, is well known for its role in redox reactions. More recently, it has emerged as a signaling molecule. By modulating NAD+-sensing enzymes, NAD+ controls hundreds of key processes from energy metabolism to cell survival, rising and falling depending on food intake, exercise, and the time of day. NAD+ levels steadily decline with age, resulting in altered metabolism and increased disease susceptibility. Restoration of NAD+ levels in old or diseased animals can promote health and extend lifespan, prompting a search for safe and efficacious NAD-boosting molecules that hold the promise of increasing the body's resilience, not just to one disease, but to many, thereby extending healthy human lifespan.
Collapse
Affiliation(s)
- Luis Rajman
- Paul F. Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Karolina Chwalek
- Paul F. Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - David A Sinclair
- Paul F. Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Laboratory for Ageing Research, Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
37
|
Cohen MS, Stewart ML, Goodman RH, Cambronne XA. Methods for Using a Genetically Encoded Fluorescent Biosensor to Monitor Nuclear NAD<sup/>. Methods Mol Biol 2018; 1813:391-414. [PMID: 30097882 PMCID: PMC6378224 DOI: 10.1007/978-1-4939-8588-3_26] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Free nicotinamide adenine dinucleotide (NAD+) serves as substrate for NAD+-consuming enzymes. As such, the local concentration of free NAD+ can influence enzymatic activities. Here we describe methods for using a fluorescent, genetically-encoded sensor to measure subcellular NAD+ concentrations. We also include a discussion of the limitations and potential applications for the current sensor. Presented in this chapter are (1) guidelines for calibrating instrumentation and experimental setups using a bead-based method, (2) instructions for incorporating required controls and properly performing ratiometric measurements in cells, and (3) descriptions of how to evaluate relative and quantitative fluctuations using appropriate statistical methods for ratio-of-ratio measurements.
Collapse
Affiliation(s)
- Michael S Cohen
- Department of Physiology and Pharmacology, Program in Chemical Biology, Oregon Health and Science University, Portland, OR, USA
| | - Melissa L Stewart
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Richard H Goodman
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Xiaolu A Cambronne
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA.
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
38
|
Mesquita I, Vergnes B, Silvestre R. Alterations on Cellular Redox States upon Infection and Implications for Host Cell Homeostasis. EXPERIENTIA SUPPLEMENTUM (2012) 2018; 109:197-220. [PMID: 30535600 DOI: 10.1007/978-3-319-74932-7_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The cofactors nicotinamide adenine dinucleotide (NAD+) and its phosphate form, NADP+, are crucial molecules present in all living cells. The delicate balance between the oxidized and reduced forms of these molecules is tightly regulated by intracellular metabolism assuring the maintenance of homeostatic conditions, which are essential for cell survival and proliferation. A recent cluster of data has highlighted the importance of the intracellular NAD+/NADH and NADP+/NADPH ratios during host-pathogen interactions, as fluctuations in the levels of these cofactors and in precursors' bioavailability may condition host response and, therefore, pathogen persistence or elimination. Furthermore, an increasing interest has been given towards how pathogens are capable of hijacking host cell proteins in their own advantage and, consequently, alter cellular redox states and immune function. Here, we review the basic principles behind biosynthesis and subcellular compartmentalization of NAD+ and NADP+, as well as the importance of these cofactors during infection, with a special emphasis on pathogen-driven modulation of host NAD+/NADP+ levels and contribution to the associated immune response.
Collapse
Affiliation(s)
- Inês Mesquita
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Baptiste Vergnes
- MIVEGEC (IRD 224-CNRS 5290-Université Montpellier), Institut de Recherche pour le Développement (IRD), Montpellier, France
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
39
|
Harlan BA, Pehar M, Sharma DR, Beeson G, Beeson CC, Vargas MR. Enhancing NAD+ Salvage Pathway Reverts the Toxicity of Primary Astrocytes Expressing Amyotrophic Lateral Sclerosis-linked Mutant Superoxide Dismutase 1 (SOD1). J Biol Chem 2016; 291:10836-46. [PMID: 27002158 DOI: 10.1074/jbc.m115.698779] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Indexed: 01/08/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD(+)) participates in redox reactions and NAD(+)-dependent signaling pathways. Although the redox reactions are critical for efficient mitochondrial metabolism, they are not accompanied by any net consumption of the nucleotide. On the contrary, NAD(+)-dependent signaling processes lead to its degradation. Three distinct families of enzymes consume NAD(+) as substrate: poly(ADP-ribose) polymerases, ADP-ribosyl cyclases (CD38 and CD157), and sirtuins (SIRT1-7). Because all of the above enzymes generate nicotinamide as a byproduct, mammalian cells have evolved an NAD(+) salvage pathway capable of resynthesizing NAD(+) from nicotinamide. Overexpression of the rate-limiting enzyme in this pathway, nicotinamide phosphoribosyltransferase, increases total and mitochondrial NAD(+) levels in astrocytes. Moreover, targeting nicotinamide phosphoribosyltransferase to the mitochondria also enhances NAD(+) salvage pathway in astrocytes. Supplementation with the NAD(+) precursors nicotinamide mononucleotide and nicotinamide riboside also increases NAD(+) levels in astrocytes. Amyotrophic lateral sclerosis (ALS) is caused by the progressive degeneration of motor neurons in the spinal cord, brain stem, and motor cortex. Superoxide dismutase 1 (SOD1) mutations account for up to 20% of familial ALS and 1-2% of apparently sporadic ALS cases. Primary astrocytes isolated from mutant human superoxide dismutase 1-overexpressing mice as well as human post-mortem ALS spinal cord-derived astrocytes induce motor neuron death in co-culture. Increasing total and mitochondrial NAD(+) content in ALS astrocytes increases oxidative stress resistance and reverts their toxicity toward co-cultured motor neurons. Taken together, our results suggest that enhancing the NAD(+) salvage pathway in astrocytes could be a potential therapeutic target to prevent astrocyte-mediated motor neuron death in ALS.
Collapse
Affiliation(s)
- Benjamin A Harlan
- From the Department of Cell and Molecular Pharmacology and Experimental Therapeutics and
| | - Mariana Pehar
- From the Department of Cell and Molecular Pharmacology and Experimental Therapeutics and
| | - Deep R Sharma
- From the Department of Cell and Molecular Pharmacology and Experimental Therapeutics and
| | - Gyda Beeson
- South Carolina College of Pharmacy Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Craig C Beeson
- South Carolina College of Pharmacy Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Marcelo R Vargas
- From the Department of Cell and Molecular Pharmacology and Experimental Therapeutics and
| |
Collapse
|
40
|
Mesquita I, Varela P, Belinha A, Gaifem J, Laforge M, Vergnes B, Estaquier J, Silvestre R. Exploring NAD+ metabolism in host-pathogen interactions. Cell Mol Life Sci 2016; 73:1225-36. [PMID: 26718485 PMCID: PMC11108276 DOI: 10.1007/s00018-015-2119-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 11/27/2015] [Accepted: 12/14/2015] [Indexed: 01/01/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD(+)) is a vital molecule found in all living cells. NAD(+) intracellular levels are dictated by its synthesis, using the de novo and/or salvage pathway, and through its catabolic use as co-enzyme or co-substrate. The regulation of NAD(+) metabolism has proven to be an adequate drug target for several diseases, including cancer, neurodegenerative or inflammatory diseases. Increasing interest has been given to NAD(+) metabolism during innate and adaptive immune responses suggesting that its modulation could also be relevant during host-pathogen interactions. While the maintenance of NAD(+) homeostatic levels assures an adequate environment for host cell survival and proliferation, fluctuations in NAD(+) or biosynthetic precursors bioavailability have been described during host-pathogen interactions, which will interfere with pathogen persistence or clearance. Here, we review the double-edged sword of NAD(+) metabolism during host-pathogen interactions emphasizing its potential for treatment of infectious diseases.
Collapse
Affiliation(s)
- Inês Mesquita
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Varela
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Belinha
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Gaifem
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | - Baptiste Vergnes
- MIVEGEC (IRD 224-CNRS 5290-Université Montpellier), Institut de Recherche pour le Développement (IRD), Montpellier, France
| | - Jérôme Estaquier
- CNRS FR 3636, Université Paris Descartes, 75006, Paris, France.
- Centre de Recherche du CHU de Québec, Université Laval, Quebec, G1V 4G2, Canada.
| | - Ricardo Silvestre
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
41
|
Rack JGM, Perina D, Ahel I. Macrodomains: Structure, Function, Evolution, and Catalytic Activities. Annu Rev Biochem 2016; 85:431-54. [PMID: 26844395 DOI: 10.1146/annurev-biochem-060815-014935] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
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.
Collapse
Affiliation(s)
| | - Dragutin Perina
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb 10002, Croatia;
| | - Ivan Ahel
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom; ,
| |
Collapse
|
42
|
VanLinden MR, Dölle C, Pettersen IKN, Kulikova VA, Niere M, Agrimi G, Dyrstad SE, Palmieri F, Nikiforov AA, Tronstad KJ, Ziegler M. Subcellular Distribution of NAD+ between Cytosol and Mitochondria Determines the Metabolic Profile of Human Cells. J Biol Chem 2015; 290:27644-59. [PMID: 26432643 DOI: 10.1074/jbc.m115.654129] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Indexed: 12/21/2022] Open
Abstract
The mitochondrial NAD pool is particularly important for the maintenance of vital cellular functions. Although at least in some fungi and plants, mitochondrial NAD is imported from the cytosol by carrier proteins, in mammals, the mechanism of how this organellar pool is generated has remained obscure. A transporter mediating NAD import into mammalian mitochondria has not been identified. In contrast, human recombinant NMNAT3 localizes to the mitochondrial matrix and is able to catalyze NAD(+) biosynthesis in vitro. However, whether the endogenous NMNAT3 protein is functionally effective at generating NAD(+) in mitochondria of intact human cells still remains to be demonstrated. To modulate mitochondrial NAD(+) content, we have expressed plant and yeast mitochondrial NAD(+) carriers in human cells and observed a profound increase in mitochondrial NAD(+). None of the closest human homologs of these carriers had any detectable effect on mitochondrial NAD(+) content. Surprisingly, constitutive redistribution of NAD(+) from the cytosol to the mitochondria by stable expression of the Arabidopsis thaliana mitochondrial NAD(+) transporter NDT2 in HEK293 cells resulted in dramatic growth retardation and a metabolic shift from oxidative phosphorylation to glycolysis, despite the elevated mitochondrial NAD(+) levels. These results suggest that a mitochondrial NAD(+) transporter, similar to the known one from A. thaliana, is likely absent and could even be harmful in human cells. We provide further support for the alternative possibility, namely intramitochondrial NAD(+) synthesis, by demonstrating the presence of endogenous NMNAT3 in the mitochondria of human cells.
Collapse
Affiliation(s)
| | | | | | - Veronika A Kulikova
- the Institute of Nanobiotechnologies, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
| | - Marc Niere
- From the Departments of Molecular Biology and
| | - Gennaro Agrimi
- the Department of Biosciences, Biotechnologies and Biopharmaceutics and
| | | | - Ferdinando Palmieri
- the Department of Biosciences, Biotechnologies and Biopharmaceutics and the Center of Excellence in Comparative Genomics, University of Bari, 70125 Bari, Italy, and
| | - Andrey A Nikiforov
- the Institute of Nanobiotechnologies, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia, the Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | | | | |
Collapse
|
43
|
Abstract
Impaired mitochondrial structure and function are common features of neurodegenerative disorders, ultimately characterized by the death of neural cells promoted by still unknown signals. Among the possible modulators of neurodegeneration, the activation of poly(ADP-ribosylation), a post-translational modification of proteins, has been considered, being the product of the reaction, poly(ADP-ribose), a signaling molecule for different cell death paradigms. The basic properties of poly(ADP-ribosylation) are here described, focusing on the mitochondrial events; cell death paradigms such as apoptosis, parthanatos, necroptosis and mitophagy are illustrated. Finally, the promising use of poly(ADP-ribosylation) inhibitors to rescue neurodegeneration is addressed.
Collapse
Affiliation(s)
| | - Anna Ivana Scovassi
- Istituto di Genetica Molecolare CNR, Via Abbiategrasso 207, 27100 Pavia, Italy.
| |
Collapse
|
44
|
Regulation of vascular endothelial growth factor by metabolic context of the cell. Glycoconj J 2015; 31:427-34. [PMID: 25214198 DOI: 10.1007/s10719-014-9547-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Expression of vascular endothelial growth factor, major endothelial specific glycoprotein growth factor that promotes angiogenesis is regulated at transcriptional, post transcriptional and posttranslational levels. One of the key posttranslational modifications involved in regulating the angiogenic potential of VEGF is covalent modification involving polyADP ribosylation. Major factors contributing to the regulation of VEGF include factors relating to hypoxia, growth factors and cytokines and hormones. Apart from these, the metabolite status of the cell as sensed by various metabolite regulators can influence the angiogenic potential. Changes in the metabolite status of the cell occur during different conditions associated with excessive or insufficient angiogenesis contributing to pathology. Effect of metabolites, as exemplified by certain metabolites such as lactate, citrate, sarcosine, metabolites of arachidonic acid on angiogenesis through the regulation of expression of VEGF as well as its angiogenic potential through polyADP ribosylation is discussed.
Collapse
|
45
|
Nikiforov A, Kulikova V, Ziegler M. The human NAD metabolome: Functions, metabolism and compartmentalization. Crit Rev Biochem Mol Biol 2015; 50:284-97. [PMID: 25837229 PMCID: PMC4673589 DOI: 10.3109/10409238.2015.1028612] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The metabolism of NAD has emerged as a key regulator of cellular and organismal homeostasis. Being a major component of both bioenergetic and signaling pathways, the molecule is ideally suited to regulate metabolism and major cellular events. In humans, NAD is synthesized from vitamin B3 precursors, most prominently from nicotinamide, which is the degradation product of all NAD-dependent signaling reactions. The scope of NAD-mediated regulatory processes is wide including enzyme regulation, control of gene expression and health span, DNA repair, cell cycle regulation and calcium signaling. In these processes, nicotinamide is cleaved from NAD+ and the remaining ADP-ribosyl moiety used to modify proteins (deacetylation by sirtuins or ADP-ribosylation) or to generate calcium-mobilizing agents such as cyclic ADP-ribose. This review will also emphasize the role of the intermediates in the NAD metabolome, their intra- and extra-cellular conversions and potential contributions to subcellular compartmentalization of NAD pools.
Collapse
Affiliation(s)
- Andrey Nikiforov
- a Institute of Nanobiotechnologies, St. Petersburg State Polytechnical University , St. Petersburg , Russia .,b Institute of Cytology, Russian Academy of Sciences , St. Petersburg , Russia , and
| | - Veronika Kulikova
- a Institute of Nanobiotechnologies, St. Petersburg State Polytechnical University , St. Petersburg , Russia
| | - Mathias Ziegler
- c Department of Molecular Biology , University of Bergen , Bergen , Norway
| |
Collapse
|
46
|
Fabrizio G, Di Paola S, Stilla A, Giannotta M, Ruggiero C, Menzel S, Koch-Nolte F, Sallese M, Di Girolamo M. ARTC1-mediated ADP-ribosylation of GRP78/BiP: a new player in endoplasmic-reticulum stress responses. Cell Mol Life Sci 2015; 72:1209-25. [PMID: 25292337 PMCID: PMC11113179 DOI: 10.1007/s00018-014-1745-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 09/23/2014] [Accepted: 09/25/2014] [Indexed: 12/12/2022]
Abstract
Protein mono-ADP-ribosylation is a reversible post-translational modification of cellular proteins. This scheme of amino-acid modification is used not only by bacterial toxins to attack host cells, but also by endogenous ADP-ribosyltransferases (ARTs) in mammalian cells. These latter ARTs include members of three different families of proteins: the well characterised arginine-specific ecto-enzymes (ARTCs), two sirtuins, and some members of the poly(ADP-ribose) polymerase (PARP/ARTD) family. In the present study, we demonstrate that human ARTC1 is localised to the endoplasmic reticulum (ER), in contrast to the previously characterised ARTC proteins, which are typical GPI-anchored ecto-enzymes. Moreover, using the "macro domain" cognitive binding module to identify ADP-ribosylated proteins, we show here that the ER luminal chaperone GRP78/BiP (glucose-regulated protein of 78 kDa/immunoglobulin heavy-chain-binding protein) is a cellular target of human ARTC1 and hamster ARTC2. We further developed a procedure to visualise ADP-ribosylated proteins using immunofluorescence. With this approach, in cells overexpressing ARTC1, we detected staining of the ER that co-localises with GRP78/BiP, thus confirming that this modification occurs in living cells. In line with the key role of GRP78/BiP in the ER stress response system, we provide evidence here that ARTC1 is activated during the ER stress response, which results in acute ADP-ribosylation of GRP78/BiP paralleling translational inhibition. Thus, this identification of ARTC1 as a regulator of GRP78/BiP defines a novel, previously unsuspected, player in GRP78-mediated ER stress responses.
Collapse
Affiliation(s)
- Gaia Fabrizio
- Laboratory of G-Protein-mediated Signalling, Department of Cellular and Translational Pharmacology, Mario Negri Sud Foundation, Via Nazionale 8/A, 66030 Santa Maria Imbaro, CH Italy
| | - Simone Di Paola
- Laboratory of G-Protein-mediated Signalling, Department of Cellular and Translational Pharmacology, Mario Negri Sud Foundation, Via Nazionale 8/A, 66030 Santa Maria Imbaro, CH Italy
- Present Address: Telethon Institute of Genetics and Medicine, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Annalisa Stilla
- Laboratory of G-Protein-mediated Signalling, Department of Cellular and Translational Pharmacology, Mario Negri Sud Foundation, Via Nazionale 8/A, 66030 Santa Maria Imbaro, CH Italy
| | - Monica Giannotta
- Genomic Approaches to Membrane Traffic Unit, Department of Cellular and Translational Pharmacology, Mario Negri Sud Foundation, Via Nazionale, 8/A, 66030 Santa Maria Imbaro, CH Italy
- Present Address: Unit of Vascular Biology, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Carmen Ruggiero
- Genomic Approaches to Membrane Traffic Unit, Department of Cellular and Translational Pharmacology, Mario Negri Sud Foundation, Via Nazionale, 8/A, 66030 Santa Maria Imbaro, CH Italy
- Present Address: Associated International Laboratory (LIA) NEOGENEX CNRS, University of Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, 660 route des Lucioles, Sophia Antipolis, 06560 Valbonne, France
| | - Stephan Menzel
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, Martinist 52, 20246 Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, Martinist 52, 20246 Hamburg, Germany
| | - Michele Sallese
- Genomic Approaches to Membrane Traffic Unit, Department of Cellular and Translational Pharmacology, Mario Negri Sud Foundation, Via Nazionale, 8/A, 66030 Santa Maria Imbaro, CH Italy
| | - Maria Di Girolamo
- Laboratory of G-Protein-mediated Signalling, Department of Cellular and Translational Pharmacology, Mario Negri Sud Foundation, Via Nazionale 8/A, 66030 Santa Maria Imbaro, CH Italy
| |
Collapse
|
47
|
Aredia F, Scovassi AI. Poly(ADP-ribose): a signaling molecule in different paradigms of cell death. Biochem Pharmacol 2014; 92:157-63. [PMID: 24976506 DOI: 10.1016/j.bcp.2014.06.021] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 06/17/2014] [Accepted: 06/17/2014] [Indexed: 12/11/2022]
Abstract
Poly(ADP-ribosylation) results from the conversion of NAD(+) into ADP-ribose and the following addition of ADP-ribose units to form polymers, further bound to acceptor proteins; once post-translationally ADP-ribosylated, proteins could change their function in basic processes. Poly(ADP-ribosylation) is activated under critical situations represented by DNA damage and cellular stress, and modulated in different paradigms of cell death. The hallmarks of the main death processes, i.e. apoptosis, parthanatos, necroptosis and autophagy, will be described, focusing on the role of poly(ADP-ribose) as a signaling molecule.
Collapse
Affiliation(s)
- Francesca Aredia
- Istituto di Genetica Molecolare CNR, Via Abbiategrasso 207, 27100 Pavia, Italy
| | - Anna Ivana Scovassi
- Istituto di Genetica Molecolare CNR, Via Abbiategrasso 207, 27100 Pavia, Italy.
| |
Collapse
|
48
|
Bereiter-Hahn J. Do we age because we have mitochondria? PROTOPLASMA 2014; 251:3-23. [PMID: 23794102 DOI: 10.1007/s00709-013-0515-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 05/21/2013] [Indexed: 06/02/2023]
Abstract
The process of aging remains a great riddle. Production of reactive oxygen species (ROS) by mitochondria is an inevitable by-product of respiration, which has led to a hypothesis proposing the oxidative impairment of mitochondrial components (e.g., mtDNA, proteins, lipids) that initiates a vicious cycle of dysfunctional respiratory complexes producing more ROS, which again impairs function. This does not exclude other processes acting in parallel or targets for ROS action in other organelles than mitochondria. Given that aging is defined as the process leading to death, the role of mitochondria-based impairments in those organ systems responsible for human death (e.g., the cardiovascular system, cerebral dysfunction, and cancer) is described within the context of "garbage" accumulation and increasing insulin resistance, type 2 diabetes, and glycation of proteins. Mitochondrial mass, fusion, and fission are important factors in coping with impaired function. Both biogenesis of mitochondria and their degradation are important regulatory mechanisms stimulated by physical exercise and contribute to healthy aging. The hypothesis of mitochondria-related aging should be revised to account for the limitations of the degradative capacity of the lysosomal system. The processes involved in mitochondria-based impairments are very similar across a large range of organisms. Therefore, studies on model organisms from yeast, fungi, nematodes, flies to vertebrates, and from cells to organisms also add considerably to the understanding of human aging.
Collapse
Affiliation(s)
- Jürgen Bereiter-Hahn
- Institut für Zellbiologie und Neurowissenschaften, Goethe Universität Frankfurt am Main, Max-von-Lauestrasse 13, 60438, Frankfurt am Main, Germany,
| |
Collapse
|
49
|
Felici R, Lapucci A, Ramazzotti M, Chiarugi A. Insight into molecular and functional properties of NMNAT3 reveals new hints of NAD homeostasis within human mitochondria. PLoS One 2013; 8:e76938. [PMID: 24155910 PMCID: PMC3796565 DOI: 10.1371/journal.pone.0076938] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/30/2013] [Indexed: 11/23/2022] Open
Abstract
Among the enzymes involved in NAD homeostasis, nicotinamide mononucleotide adenylyltransferases (NMNAT1-3) are central to intracellular NAD formation. Although NMNAT3 is postulated to be a mitochondrial enzyme contributing to NAD-dependent organelle functioning, information on endogenous proteins is lacking. We report that in human cells a single gene nmnat3 localized on chromosome 3 codes for two mRNA splice variants NMNATv1 and FKSG76, whereas the previously reported NMNAT3v2 transcript is not present. However, NMNAT3v1 and FKSG76 proteins are not detectable, consistent with the finding that an upstream ORF in their mRNAs negatively regulates translation. NMNAT3v1 transfection demonstrates that the protein is cytosolic and inactive, whereas FKSG76 is mitochondrial but operates NAD cleavage rather than synthesis. In keeping with the lack of NMNAT3, we show that extracellular NAD, but not its metabolic precursors, sustains mitochondrial NAD pool in an ATP-independent manner. Data of the present study modify the scenario of the origin of mitochondrial NAD by showing that, in human cells, NMNAT3 is absent in mitochondria, and, akin to plants and yeast, cytosolic NAD maintains the mitochondrial NAD pool.
Collapse
Affiliation(s)
- Roberta Felici
- Department of Health Sciences, University of Florence, Florence, Italy
- * E-mail:
| | - Andrea Lapucci
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Matteo Ramazzotti
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Alberto Chiarugi
- Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
50
|
Satori CP, Henderson MM, Krautkramer EA, Kostal V, Distefano MM, Arriaga EA. Bioanalysis of eukaryotic organelles. Chem Rev 2013; 113:2733-811. [PMID: 23570618 PMCID: PMC3676536 DOI: 10.1021/cr300354g] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Chad P. Satori
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN, USA, 55455
| | - Michelle M. Henderson
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN, USA, 55455
| | - Elyse A. Krautkramer
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN, USA, 55455
| | - Vratislav Kostal
- Tescan, Libusina trida 21, Brno, 623 00, Czech Republic
- Institute of Analytical Chemistry ASCR, Veveri 97, Brno, 602 00, Czech Republic
| | - Mark M. Distefano
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN, USA, 55455
| | - Edgar A. Arriaga
- Department of Chemistry, University of Minnesota, Twin Cities, Minneapolis, MN, USA, 55455
| |
Collapse
|