1
|
Xing S, Kang X, Wang R, Wang C, Wang Y, Bao X, Zhao J. Microbial Production of Nicotinamide Mononucleotide: Key Enzymes Discovery, Host Cells Selection, and Pathways Design and Optimization. ACS Synth Biol 2025; 14:1352-1366. [PMID: 40237164 DOI: 10.1021/acssynbio.5c00038] [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] [Indexed: 04/18/2025]
Abstract
As an important bioactive substance in cells, nicotinamide mononucleotide (NMN) has been proven to play an important role in antiaging, treatment of neurodegenerative diseases, and cardioprotection. It presents a high potential for application in the research fields of functional foods, cosmetics, healthcare products, and active pharmaceuticals. With the increased demand, whether NMN can achieve large-scale industrial production has been a wide concern. The chemical synthesis method of NMN mainly faces the problems of separation, purification, and complex process control; in contrast, biosynthesis methods such as microbial fermentation and enzyme catalysis are considered to be the mainstream of the future industrial production of NMN due to the advantages of environmental friendliness, high efficiency, and simple separation. This review first describes the physiological functions of NMN and the related areas of its applications. Subsequently, it focuses on the research progress on different synthetic pathways of NMN in biosynthetic approaches, mining and modification of key enzymes, chassis cell design and optimization, and whole-cell catalysis. Meanwhile, the regulatory strategies, methods, and process control of the microbial synthesis of NMN are also elaborated, and the synthesis efficiencies of different chassis cells are systematically compared. Finally, this review summarizes the existing problems and challenges of microbial synthesis of NMN and proposes future strategies and directions to address these issues. This work provides technical references and a theoretical basis for researching efficient NMN microbial synthesis and application.
Collapse
Affiliation(s)
- Shuyi Xing
- State Key Laboratory of Green Papermaking and Resource Recycling, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Xiulong Kang
- State Key Laboratory of Green Papermaking and Resource Recycling, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Rui Wang
- State Key Laboratory of Green Papermaking and Resource Recycling, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Chengqiang Wang
- College of Life Sciences, National Engineering Research Center for Efficient Utilization of Soil and Fertilizer Resources, Shandong Key Laboratory of Agricultural Microbiology, Shandong Agricultural University, Taian 271018, China
| | - Yanjun Wang
- National Key Laboratory of Efficient Utilization of Nutrient Resources, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Xiaoming Bao
- State Key Laboratory of Green Papermaking and Resource Recycling, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Jianzhi Zhao
- State Key Laboratory of Green Papermaking and Resource Recycling, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| |
Collapse
|
2
|
Lin Y, Wu H, Wang J, He W, Hou J, Martin VT, Zhu C, Chen Y, Zhong J, Yu B, Lu A, Guan D, Qin G, Chen W. Nicotinamide Adenine Dinucleotide-Loaded Lubricated Hydrogel Microspheres with a Three-Pronged Approach Alleviate Age-Related Osteoarthritis. ACS NANO 2025; 19:17606-17626. [PMID: 40315404 PMCID: PMC12080321 DOI: 10.1021/acsnano.5c01184] [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] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/04/2025]
Abstract
Chondrocyte senescence, synovitis, and decreased level of lubrication play pivotal roles in the pathogenesis of age-related osteoarthritis (AROA). However, there are currently no effective therapeutic interventions capable of altering the progression of OA until it reaches advanced stages, necessitating joint replacement. In this study, lubricious and drug-loaded hydrogel microspheres were designed and fabricated by utilizing microfluidic technology for radical polymerization of chondroitin sulfate methacrylate and incorporating nicotinamide adenine dinucleotide (NAD)-loaded liposomes modified with lactoferrin that are positively charged. Mechanical, tribological, and drug release analyses demonstrated enhanced lubrication properties and an extended drug dissemination time for the NAD@NPs@HM microspheres. In vitro assays unveiled the ability of NAD@NPs@HM to counteract chondrocyte senescence. RNA sequencing analysis, untargeted metabolomics analysis, and in vitro experiments on macrophages revealed that NAD@NPs@HM can regulate the metabolic reprogramming of synovial macrophages, promoting their repolarization from the M1 to M2 phenotype, thereby alleviating synovitis. Intra-articular injection of NAD@NPs@HM in aged mice reduced the mechanisms associated with AROA. These results suggest that NAD@NPs@HM may provide extended drug release, improved joint lubrication leading to better gait, and attenuation of AROA pathogenic processes, indicating its potential as a therapeutic approach for AROA.
Collapse
Affiliation(s)
- Yanpeng Lin
- Department
of Radiology, Nanfang Hospital, Southern
Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Hangtian Wu
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Jun Wang
- School
of Animal Science and Technology, Foshan
University, Foshan, Guangdong 528231, People’s Republic of China
| | - Wanling He
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
- Guangdong
Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou 510515, P. R. China
| | - Jiahui Hou
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Vidmi Taolam Martin
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Chencheng Zhu
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Yupeng Chen
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
- Guangdong
Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou 510515, P. R. China
| | - Junyuan Zhong
- Department
of Medical Imaging, Ganzhou People’s
Hospital, Ganzhou, Jiangxi 341000, P. R. China
| | - Bin Yu
- Division
of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang
Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Aiping Lu
- Institute
of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong 999077, P. R. China
- Guangdong-Hong
Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510515, P. R. China
| | - Daogang Guan
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P. R. China
- Guangdong
Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou 510515, P. R. China
| | - Genggeng Qin
- Department
of Radiology, Nanfang Hospital, Southern
Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Weiguo Chen
- Department
of Radiology, Nanfang Hospital, Southern
Medical University, Guangzhou, Guangdong 510515, P. R. China
| |
Collapse
|
3
|
Song WS, Shen X, Du K, Ramirez CB, Park SH, Cao Y, Le J, Bae H, Kim J, Chun Y, Khong NJ, Kim M, Jung S, Choi W, Lopez ML, Said Z, Song Z, Lee SG, Nicholas D, Sasaki Y, Milbrandt J, Imagawa DK, Skowronska-Krawczyk D, Chen D, Lee G, Jang C, Yang Q. Nicotinic acid riboside maintains NAD + homeostasis and ameliorates aging-associated NAD + decline. Cell Metab 2025:S1550-4131(25)00217-7. [PMID: 40315855 DOI: 10.1016/j.cmet.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 02/05/2025] [Accepted: 04/14/2025] [Indexed: 05/04/2025]
Abstract
Liver-derived circulating nicotinamide from nicotinamide adenine dinucleotide (NAD+) catabolism primarily feeds systemic organs for NAD+ synthesis. We surprisingly found that, despite blunted hepatic NAD+ and nicotinamide production in liver-specific nicotinamide nucleotide adenylyltransferase 1 (NMNAT1) deletion mice (liver-specific knockout [LKO]), circulating nicotinamide and extra-hepatic organs' NAD+ are unaffected. Metabolomics reveals a massive accumulation of a novel molecule in the LKO liver, which we identify as nicotinic acid riboside (NaR). We further demonstrate cytosolic 5'-nucleotidase II (NT5C2) as the NaR-producing enzyme. The liver releases NaR to the bloodstream, and kidneys take up NaR to synthesize NAD+ through nicotinamide riboside kinase 1 (NRK1) and replenish circulating nicotinamide. Serum NaR levels decline with aging, whereas oral NaR supplementation in aged mice boosts serum nicotinamide and multi-organ NAD+, including kidneys, and reduces kidney inflammation and albuminuria. Thus, the liver-kidney axis maintains systemic NAD+ homeostasis via circulating NaR, and NaR supplement ameliorates aging-associated NAD+ decline and kidney dysfunction.
Collapse
Affiliation(s)
- Won-Suk Song
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Xiyu Shen
- Department of Medicine, Physiology and Biophysics, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Kang Du
- Department of Medicine, Physiology and Biophysics, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Cuauhtemoc B Ramirez
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA; Department of Microbiology and Molecular Genetics, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Sang Hee Park
- Department of Medicine, Physiology and Biophysics, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Yang Cao
- Department of Medicine, Physiology and Biophysics, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Johnny Le
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Hosung Bae
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Joohwan Kim
- Department of Microbiology and Molecular Genetics, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Yujin Chun
- Department of Microbiology and Molecular Genetics, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Nikki Joyce Khong
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Marie Kim
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Sunhee Jung
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Wonsuk Choi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Miranda L Lopez
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Zaid Said
- Department of Medicine, Physiology and Biophysics, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Zehan Song
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sang-Guk Lee
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA; Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Dequina Nicholas
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, School of Biological Sciences, Irvine, CA 92697, USA
| | - Yo Sasaki
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - David K Imagawa
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California, Irvine, Orange, CA 92868, USA
| | - Dorota Skowronska-Krawczyk
- Department of Physiology and Biophysics, Department of Ophthalmology, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Danica Chen
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gina Lee
- Department of Microbiology and Molecular Genetics, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA.
| | - Qin Yang
- Department of Medicine, Physiology and Biophysics, University of California, Irvine, School of Medicine, Irvine, CA 92697, USA.
| |
Collapse
|
4
|
Li H, Hu Q, Zhu D, Wu D. The Role of NAD + Metabolism in Cardiovascular Diseases: Mechanisms and Prospects. Am J Cardiovasc Drugs 2025; 25:307-327. [PMID: 39707143 DOI: 10.1007/s40256-024-00711-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 12/23/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a promising anti-aging molecule that plays a role in cellular energy metabolism and maintains redox homeostasis. Additionally, NAD+ is involved in regulating deacetylases, DNA repair enzymes, inflammation, and epigenetics, making it indispensable in maintaining the basic functions of cells. Research on NAD+ has become a hotspot, particularly regarding its potential in cardiovascular disease (CVD). Many studies have demonstrated that NAD+ plays a crucial role in the occurrence and development of CVD. This review summarizes the biosynthesis and consumption of NAD+, along with its precursors and their effects on raising NAD+ levels. We also discuss new mechanisms of NAD+ regulation in cardiovascular risk factors and its effects of NAD+ on atherosclerosis, aortic aneurysm, heart failure, hypertension, myocardial ischemia-reperfusion injury, diabetic cardiomyopathy, and dilated cardiomyopathy, elucidating different mechanisms and potential treatments. NAD+-centered therapy holds promising advantages and prospects in the field of CVD.
Collapse
Affiliation(s)
- Huimin Li
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Qingxun Hu
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Deqiu Zhu
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Dan Wu
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| |
Collapse
|
5
|
Ma X, Wang Q, Chen K, Shen Y, Guan J, Xu M, Rao Z, Zhang X. Protein Engineering and Dual-Module Optimization for Efficient NMN Production in E. coli. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9174-9186. [PMID: 40172130 DOI: 10.1021/acs.jafc.5c00043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Nicotinamide mononucleotide (NMN) has received widespread attention as a supplement of NAD+ in cells. In this study, a dual-module reaction system was constructed to synthesize NR using uridine and nicotinamide, and further to efficiently synthesize NMN. First, module 1 was constructed, which catalyzed the synthesis of NMN from NR using an efficient NRK and ATP regeneration system. Then module 2 was constructed by introducing pyrimidine nucleoside phosphorylase (PyNP) to synthesize NMN from uridine and NAM under the synergistic catalysis of NRK. Based on the fact that NRK has both phosphorylation and group transfer functions in the dual-module system, the mutant KlmNRKM4 with nearly 4-fold increased stability was obtained through predicted structure and evolutionary conservation analysis. At the same time, the pncC, deoD, ushA, nadR and deoB genes encoding endogenous degradative enzymes in Escherichia coli affect substrate and intermediate conversion were knocked out. Finally, by optimizing the reaction conditions of the dual-module recombination system, a high NMN conversion rate of 81.1% was achieved using 300 mM uridine and nicotinamide as substrates. This study provides a novel and efficient pathway for the biosynthesis of NMN.
Collapse
Affiliation(s)
- Xu Ma
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Qiang Wang
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Kewei Chen
- Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Yang Shen
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Jingyi Guan
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Meijuan Xu
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Zhiming Rao
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Xian Zhang
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
6
|
Yaku K, Palikhe S, Iqbal T, Hayat F, Watanabe Y, Fujisaka S, Izumi H, Yoshida T, Karim M, Uchida H, Nawaz A, Tobe K, Mori H, Migaud ME, Nakagawa T. Nicotinamide riboside and nicotinamide mononucleotide facilitate NAD + synthesis via enterohepatic circulation. SCIENCE ADVANCES 2025; 11:eadr1538. [PMID: 40117359 PMCID: PMC11927621 DOI: 10.1126/sciadv.adr1538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 02/18/2025] [Indexed: 03/23/2025]
Abstract
Decreased nicotinamide adenine dinucleotide (oxidized form) (NAD+) levels are reportedly associated with several aging-related disorders. Thus, supplementation with NAD+ precursors, such as nicotinamide mononucleotide (NMN) and nicotinamide riboside (NR), exhibits beneficial effects against these disorders. However, the in vivo metabolic pathways of NMN and NR remain to be elucidated. In this study, we comprehensively analyzed the fate of orally and intravenously administered NMN and NR in mice using NAD+ metabolomics. We found that only a small portion of orally administered NMN and NR was directly absorbed from the small intestine and that most of them underwent gut microbiota-mediated deamidation and conversion to nicotinic acid (NA). Moreover, intravenously administered NMN and NR were rapidly degraded into nicotinamide and secreted to bile followed by deamidation to NA by gut microbiota. Thus, enterohepatic circulated NA is preferentially used in the liver. These findings showed that NMN and NR are indirectly converted to NAD+ via unexpected metabolic pathways.
Collapse
Affiliation(s)
- Keisuke Yaku
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Sailesh Palikhe
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Tooba Iqbal
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Faisal Hayat
- Mitchell Cancer Institute, Department of Pharmacology, University of South Alabama, 1660 Springhill Avenue, Mobile, AL 36693, USA
| | - Yoshiyuki Watanabe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Shiho Fujisaka
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Hironori Izumi
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
- Research Center for Idling Brain Science (RCIBS), University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Tomoyuki Yoshida
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
- Research Center for Idling Brain Science (RCIBS), University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Mariam Karim
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Hitoshi Uchida
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Allah Nawaz
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
- Research Center for Pre-Disease Science, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
- Research Center for Idling Brain Science (RCIBS), University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| | - Marie E. Migaud
- Mitchell Cancer Institute, Department of Pharmacology, University of South Alabama, 1660 Springhill Avenue, Mobile, AL 36693, USA
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
- Research Center for Pre-Disease Science, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan
| |
Collapse
|
7
|
Madawala R, Banks JL, Hancock SE, Quek LE, Turner N, Wu LE. CD38 mediates nicotinamide mononucleotide base exchange to yield nicotinic acid mononucleotide. J Biol Chem 2025; 301:108248. [PMID: 39894219 PMCID: PMC11903787 DOI: 10.1016/j.jbc.2025.108248] [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: 10/15/2024] [Revised: 01/19/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025] Open
Abstract
Nicotinamide mononucleotide (NMN) is a widely investigated metabolic precursor to the prominent enzyme cofactor NAD+, where it is assumed that delivery of this compound results in its direct incorporation into NAD+via the canonical salvage/recycling pathway. Surprisingly, treatment with this salvage pathway intermediate leads to increases in nicotinic acid mononucleotide (NaMN) and nicotinic acid adenine dinucleotide, two members of the Preiss-Handler/de novo pathways. In mammals, these pathways are not known to intersect prior to the production of NAD+. Here, we show that the cell surface enzyme CD38 can mediate a base-exchange reaction on NMN, whereby the nicotinamide ring is exchanged with a free nicotinic acid to yield the Preiss-Handler/de novo pathway intermediate NaMN, with in vivo small molecule inhibition of CD38 abolishing the NMN-induced increase in NaMN and nicotinic acid adenine dinucleotide. Together, these data demonstrate a new mechanism by which the salvage pathway and Preiss-Handler/de novo pathways can exchange intermediates in mammalian NAD+ biosynthesis.
Collapse
Affiliation(s)
- Romanthi Madawala
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW, Australia
| | - Jasmine L Banks
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia; School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Sarah E Hancock
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW, Australia; Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Lake-Ee Quek
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, Australia
| | - Nigel Turner
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW, Australia; Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.
| | - Lindsay E Wu
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW, Australia.
| |
Collapse
|
8
|
Cai C, Yang P, Shi Y, Wang X, Chen G, Zhang Q, Cheng G, Kong W, Xu Z. Transcriptomic and metabolomic analysis revealed potential mechanisms of growth and disease resistance dimorphism in male and female common carp (Cyprinus carpio). FISH & SHELLFISH IMMUNOLOGY 2025; 158:110150. [PMID: 39842680 DOI: 10.1016/j.fsi.2025.110150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
Sexual dimorphism is well-documented in aquaculture, particularly regarding growth differences, wherein one sex often grows faster than the other. However, despite the phenomenon being so widely documented, its underlying molecular mechanisms remain poorly understood. As an important digestive and immune organ, the gut plays key roles in the regulation of fish growth. In this study, we conducted RNA-seq and metabolomic analysis on the gut of female and male common carp. We discovered that growth-related pathways, such as "Glycolysis/Gluconeogenesis" and "Riboflavin metabolism" are significantly enriched in the gut of female carp. Conversely, pathways linked to disease resistance, such as "Th17 cell differentiation" and "Autophagy-animal" are predominantly enriched in male carp. Following intraperitoneal injection of spring viraemia of carp virus (SVCV) into both male and female carp, quantitative reverse transcription polymerase chain reaction (RT-qPCR) analysis and histopathological staining confirmed that male carp exhibit greater disease resistance compared to females. This study identified the disease resistance dimorphism in common carp and specific mechanisms underlying growth differences. Our findings offer valuable insights for the application of growth dimorphism and disease-resistant breeding in fish.
Collapse
Affiliation(s)
- Chang Cai
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Peng Yang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Yong Shi
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Xinyou Wang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Guanghui Chen
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Qianqian Zhang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Gaofeng Cheng
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Weiguang Kong
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Zhen Xu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.
| |
Collapse
|
9
|
Zhang M, Jia H, Zhuang L, Xu Y, Zhang T, Gu J, He S, Li D. Ultrathin high-entropy hydrotalcites-based injectable hydrogel with programmed bactericidal and anti-inflammatory effects to accelerate drug-resistant bacterial infected wound healing. Colloids Surf B Biointerfaces 2025; 247:114450. [PMID: 39671734 DOI: 10.1016/j.colsurfb.2024.114450] [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: 10/17/2024] [Revised: 11/29/2024] [Accepted: 12/08/2024] [Indexed: 12/15/2024]
Abstract
Drug-resistant bacteria infected wounds often bring high risks of delayed healing process and even death. Sonodynamic therapy (SDT) can efficiently kill drug-resistant bacteria. However, superabundant reactive oxygen species (ROS) generated during SDT inevitably trigger significant inflammatory responses, hindering tissue remodeling. Herein, we develop intelligent ultrathin high-entropy hydrotalcites (UHE-HTs)-based injectable thermal-responsive hydrogel loaded with nicotinamide mononucleotide (UHE-HTs/PFN), aiming to achieve programmed antibacterial and anti-inflammatory effects. In the early infection stage, sonosensitive UHE-HTs/PFN hydrogel simultaneously can trigger rapid production of singlet oxygen (1O2) under ultrasound and efficient MDR bacterial sterilization. After halting ultrasonic irradiation, oxidoreductase-mimicking catalysis and nicotinamide mononucleotide release of UHE-HTs/PFN hydrogel effectively reduce ROS levels at wound sites, dampening the NF-κB inflammatory pathway. Such inhibited NF-κB expression can not only reduce the production of pro-inflammatory cytokines and inflammatory responses, but also significantly down-regulate the pyroptosis pathways (NLRP3/ASC/Casp-1) and inhibit pyroptosis that leads to inflammation. Moreover, significantly reduced ROS levels and synergistic release of Mg2+ reverse pro-inflammatory immune microenvironment. Both in vitro and in vivo assays demonstrate that UHE-HTs/PFN hydrogel can transform the adverse infected wound environment into a regenerative one by eradicating drug-resistant bacteria, scavenging ROS, and synergistic anti-inflammation. Therefore, this work develop an intelligent UHE-HTs/PFN hydrogel act as a "lever" that effectively achieve a balance between ROS generation and annihilation, rebuilding harmonious bactericidal and anti-inflammatory effects to remedy drug-resistant bacteria infected wound.
Collapse
Affiliation(s)
- Mingming Zhang
- The Ninth Medical Center of Chinese PLA General Hospital, 9 Anxiang Beili, Chaoyang District, Beijing 100101, China
| | - Huaping Jia
- The Ninth Medical Center of Chinese PLA General Hospital, 9 Anxiang Beili, Chaoyang District, Beijing 100101, China
| | - Liang Zhuang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, 11 Fucheng Road, Haidian District, Beijing 100048, China
| | - Yongjie Xu
- The Ninth Medical Center of Chinese PLA General Hospital, 9 Anxiang Beili, Chaoyang District, Beijing 100101, China
| | - Ting Zhang
- The Ninth Medical Center of Chinese PLA General Hospital, 9 Anxiang Beili, Chaoyang District, Beijing 100101, China
| | - Jianwen Gu
- The Ninth Medical Center of Chinese PLA General Hospital, 9 Anxiang Beili, Chaoyang District, Beijing 100101, China.
| | - Shan He
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, 11 Fucheng Road, Haidian District, Beijing 100048, China.
| | - Dawei Li
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing 100091, China.
| |
Collapse
|
10
|
Chen M, Yuan L, Chen B, Chang H, Luo J, Zhang H, Chen Z, Kong J, Yi Y, Bai M, Dong M, Zhou H, Jiang H. SLC29A1 and SLC29A2 are human nicotinamide cell membrane transporters. Nat Commun 2025; 16:1181. [PMID: 39885119 PMCID: PMC11782521 DOI: 10.1038/s41467-025-56402-y] [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: 11/23/2023] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
Nicotinamide (NAM), a main precursor of NAD+, is essential for cellular fuel respiration, energy production, and other cellular processes. Transporters for other precursors of NAD+ such as nicotinic acid and nicotinamide mononucleotide (NMN) have been identified, but the cellular transporter of nicotinamide has not been elucidated. Here, we demonstrate that equilibrative nucleoside transporter 1 and 2 (ENT1 and 2, encoded by SLC29A1 and 2) drive cellular nicotinamide uptake and establish nicotinamide metabolism homeostasis. In addition, ENT1/2 exhibits a strong capacity to change the cellular metabolite composition and the transcript, especially those related to nicotinamide. We further observe that ENT1/2 regulates cellular respiration and senescence, contributing by altering the NAD+ pool level and mitochondrial status. Changes to cellular respiration, mitochondrial status and senescence by ENT1/2 knockdown are reversed by NMN supplementation. Together, ENT1 and ENT2 act as both cellular nicotinamide-level keepers and nicotinamide biological regulators through their NAM transport functions.
Collapse
Affiliation(s)
- Mingyang Chen
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Luexiang Yuan
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Binxin Chen
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University, Hangzhou, China
| | - Hui Chang
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Luo
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University, Hangzhou, China
| | - Hengbin Zhang
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Zhongjian Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, China
| | - Jiao Kong
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Yaodong Yi
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Mengru Bai
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Westlake University, Hangzhou, China
| | - Minlei Dong
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Hui Zhou
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
| | - Huidi Jiang
- College of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
| |
Collapse
|
11
|
Imai SI. NAD World 3.0: the importance of the NMN transporter and eNAMPT in mammalian aging and longevity control. NPJ AGING 2025; 11:4. [PMID: 39870672 PMCID: PMC11772665 DOI: 10.1038/s41514-025-00192-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 01/07/2025] [Indexed: 01/29/2025]
Abstract
Over the past five years, systemic NAD+ (nicotinamide adenine dinucleotide) decline has been accepted to be a key driving force of aging in the field of aging research. The original version of the NAD World concept was proposed in 2009, providing an integrated view of the NAD+-centric, systemic regulatory network for mammalian aging and longevity control. The reformulated version of the concept, the NAD World 2.0, was then proposed in 2016, emphasizing the importance of the inter-tissue communications between the hypothalamus and peripheral tissues including adipose tissue and skeletal muscle. There has been significant progress in our understanding of the importance of nicotinamide mononucleotide (NMN), a key NAD+ intermediate, and nicotinamide phosphoribosyltransferase (NAMPT), particularly extracellular NAMPT (eNAMPT). With these exciting developments, the further reformulated version of the concept, the NAD World 3.0, is now proposed, featuring multi-layered feedback loops mediated by NMN and eNAMPT for mammalian aging and longevity control.
Collapse
Affiliation(s)
- Shin-Ichiro Imai
- Department of Developmental Biology, Department of Medicine (Joint), Washington University School of Medicine, St. Louis, Missouri, USA.
- Institute for Research on Productive Aging (IRPA), Tokyo, Japan.
| |
Collapse
|
12
|
Hashimoto S, Gamage U, Inoue Y, Iwata H, Morimoto Y. Nicotinamide mononucleotide boosts the development of bovine oocyte by enhancing mitochondrial function and reducing chromosome lagging. Sci Rep 2025; 15:310. [PMID: 39747142 PMCID: PMC11696260 DOI: 10.1038/s41598-024-81393-z] [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: 08/13/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025] Open
Abstract
Nicotinamide adenine dinucleotide (NAD(H)) and its metabolites function as crucial regulators of physiological processes, allowing cells to adapt to environmental changes such as nutritional deficiencies, genotoxic factors, disruptions in circadian rhythms, infections, inflammation, and exogenous substances. Here, we investigated whether elevated NAD(H) levels in oocytes enhance their quality and improve developmental competence following in vitro fertilization (IVF). Bovine cumulus-oocyte complexes (COCs) were matured in a culture medium supplemented with 0-100 μM nicotinamide mononucleotide (NMN), a precursor of NAD(H). The addition of NMN caused an increase in intracellular NAD(H) and nicotinamide adenine dinucleotide phosphate levels, leading to enhanced competence for development to the blastocyst stage after IVF. The increase in intracellular NAD(H) levels led to changes in the expression of mitochondria function-related genes. As a result, NMN supplementation increased the ratio of MitoTracker Orange fluorescence to nonyl acridine orange fluorescence, as well as adenosine triphosphate levels, while decreasing reactive oxygen species levels in the oocytes. NMN also lowered chromosome lagging during anaphase. These results suggest that increased NAD(H) levels in oocytes following NMN treatment enhances post-fertilization developmental competence through improved mitochondrial function.
Collapse
Affiliation(s)
- Shu Hashimoto
- Graduate School of Medicine, Osaka Metropolitan University, Osaka, 545-8585, Japan.
| | | | - Yuki Inoue
- Department of Animal Science, Tokyo University of Agriculture, Kanagawa, 243-0034, Japan
| | - Hisataka Iwata
- Department of Animal Science, Tokyo University of Agriculture, Kanagawa, 243-0034, Japan
| | | |
Collapse
|
13
|
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
|
14
|
Cabulong RB, Kafle SR, Singh A, Sharma M, Kim BS. Biological production of nicotinamide mononucleotide: a review. Crit Rev Biotechnol 2024:1-18. [PMID: 39675885 DOI: 10.1080/07388551.2024.2433993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 07/30/2024] [Accepted: 10/27/2024] [Indexed: 12/17/2024]
Abstract
Nicotinamide mononucleotide (NMN) presents significant therapeutic potential against aging-related conditions, such as Alzheimer's disease, due to its consistent and strong pharmacological effects. Aside from its anti-aging effect, NMN is also an emerging noncanonical cofactor for orthogonal metabolic pathways in the field of biomanufacturing. This has significant advantages in the field of metabolic engineering, allowing cells to produce unnatural chemicals without disrupting the natural cellular processes. NMN is produced through both the chemical and biological methods, with the latter being more environmentally sustainable. The primary biological production pathway centers on the enzyme nicotinamide phosphoribosyltransferase, which transforms nicotinamide and phosphoribosyl pyrophosphate to NMN. Efforts to increase NMN production have been explored in microorganisms, such as: Escherichia coli, Bacillus subtilis, and yeast, serving as biocatalysts, by rewiring their metabolic processes. Although most researchers are focusing on genetically and metabolically manipulating microorganisms to act as biocatalysts, a growing number of studies on cell-free synthesis are emerging as a promising strategy for producing NMN. This review explores the different biological production techniques of NMN employing microorganisms. This article, in particular, is essential to those who are working on NMN production using microbial strain engineering and cell-free systems.
Collapse
Affiliation(s)
- Rhudith B Cabulong
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Saroj Raj Kafle
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Anju Singh
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Mukesh Sharma
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Beom Soo Kim
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
15
|
Kong L, Li X, Liu T, Yao Q, Qin J. Harnessing lactic acid bacteria for nicotinamide mononucleotide biosynthesis: a review of strategies and future directions. Front Microbiol 2024; 15:1492179. [PMID: 39735184 PMCID: PMC11681623 DOI: 10.3389/fmicb.2024.1492179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/22/2024] [Indexed: 12/31/2024] Open
Abstract
Nicotinamide mononucleotide (NMN), one of the crucial precursors of nicotinamide adenine dinucleotide, has garnered considerable interest for its pharmacological and anti-aging effects, conferring potential health and economic benefits for humans. Lactic acid bacteria (LAB) are one of the most important probiotics, which is commonly used in the dairy industry. Due to its probiotic properties, it presents an attractive platform for food-grade NMN production. LAB have also been extensively utilized to enhance the functional properties of pharmaceuticals and cosmetics, making them promising candidates for large-scale up synthesis of NMN. This review provides an in-depth analysis of various metabolic engineering strategies, including enzyme optimization, pathway rewiring, and fermentation process enhancements, to increase NMN yields in LAB. It explores both CRISPR/Cas9 and traditional methods to manipulate key biosynthetic pathways. In particular, this study discussed future research directions, emphasizing the application of synthetic biology, systems biology, and AI-driven optimization to further enhance NMN production. It provides invaluable insights into developing scalable and industrially relevant processes for NMN production to meet the growing market demand.
Collapse
Affiliation(s)
- Linghui Kong
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Xinyu Li
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Taiyu Liu
- Lab of Biorefinery, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Qingshou Yao
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Jiayang Qin
- School of Pharmacy, Binzhou Medical University, Yantai, China
| |
Collapse
|
16
|
Li B, Meng X, Liu W. An overview of engineering microbial production of nicotinamide mononucleotide. J Biotechnol 2024; 396:80-88. [PMID: 39491727 DOI: 10.1016/j.jbiotec.2024.10.014] [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: 08/29/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
As the human body gradually ages, the cellular level of NAD+ will decline, which has been found to be related to a variety of age-related diseases. As a precursor of NAD+, NMN is able to effectively promote the synthesis of NAD+ with no significant side effects. Microbial production of NMN holds the potential to lower the production cost and facilitate its wide application. In this review, based on the metabolic pathway of NAD+, we summarize recent advances of metabolic engineering strategies for NMN biosynthesis. An outlook for future optimization to improve NMN production is also discussed.
Collapse
Affiliation(s)
- Boting Li
- State Key Laboratory of Microbial Technology, Microbiology Technology Institute, Shandong University, No. 72 Binhai Road, Qingdao 266237, PR China
| | - Xiangfeng Meng
- State Key Laboratory of Microbial Technology, Microbiology Technology Institute, Shandong University, No. 72 Binhai Road, Qingdao 266237, PR China
| | - Weifeng Liu
- State Key Laboratory of Microbial Technology, Microbiology Technology Institute, Shandong University, No. 72 Binhai Road, Qingdao 266237, PR China.
| |
Collapse
|
17
|
Kolotyeva NA, Groshkov AA, Rozanova NA, Berdnikov AK, Novikova SV, Komleva YK, Salmina AB, Illarioshkin SN, Piradov MA. Pathobiochemistry of Aging and Neurodegeneration: Deregulation of NAD+ Metabolism in Brain Cells. Biomolecules 2024; 14:1556. [PMID: 39766263 PMCID: PMC11673498 DOI: 10.3390/biom14121556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
NAD+ plays a pivotal role in energy metabolism and adaptation to external stimuli and stressful conditions. A significant reduction in intracellular NAD+ levels is associated with aging and contributes to the development of chronic cardiovascular, neurodegenerative, and metabolic diseases. It is of particular importance to maintain optimal levels of NAD+ in cells with high energy consumption, particularly in the brain. Maintaining the tissue level of NAD+ with pharmacological tools has the potential to slow down the aging process, to prevent the development of age-related diseases. This review covers key aspects of NAD+ metabolism in terms of brain metabolic plasticity, including NAD+ biosynthesis and degradation in different types of brain cells, as well as its contribution to the development of neurodegeneration and aging, and highlights up-to-date approaches to modulate NAD+ levels in brain cells.
Collapse
|
18
|
Nanga RPR, Wiers CE, Elliott MA, Wilson NE, Liu F, Cao Q, Swago S, Jacobs PS, Armbruster R, Reddy D, Baur JA, Witschey WR, Detre JA, Reddy R. Acute nicotinamide riboside supplementation increases human cerebral NAD + levels in vivo. Magn Reson Med 2024; 92:2284-2293. [PMID: 39044608 PMCID: PMC11436296 DOI: 10.1002/mrm.30227] [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: 03/22/2024] [Revised: 05/30/2024] [Accepted: 07/01/2024] [Indexed: 07/25/2024]
Abstract
PURPOSE The purpose of this study was to determine the effect of acute nicotinamide riboside (NR) supplementation on cerebral nicotinamide adenine dinucleotide (NAD+) levels in the human brain in vivo by means of downfield proton MRS (DF 1H MRS). METHODS DF 1H MRS was performed on 10 healthy volunteers in a 7.0 T MRI scanner with spectrally selective excitation and spatially selective localization to determine cerebral NAD+ levels on two back-to-back days: once after an overnight fast (baseline) and once 4 h after oral ingestion of nicotinamide riboside (900 mg). Additionally, two more baseline scans were performed following the same paradigm to assess test-retest reliability of the NAD+ levels in the absence of NR. RESULTS NR supplementation increased mean NAD+ concentration compared to the baseline (0.458 ± 0.053 vs. 0.392 ± 0.058 mM; p < 0.001). The additional two baseline scans demonstrated no differences in mean NAD+ concentrations (0.425 ± 0.118 vs. 0.405 ± 0.082 mM; p = 0.45), and no difference from the first baseline scan (F(2, 16) = 0.907; p = 0.424). CONCLUSION These preliminary results confirm that acute NR supplementation increases cerebral NAD+ levels in healthy human volunteers and shows the promise of DF 1H MRS utility for robust detection of NAD+ in humans in vivo.
Collapse
Affiliation(s)
- Ravi Prakash Reddy Nanga
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA
| | - Corinde E. Wiers
- Department of Psychiatry, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA
| | - Mark A. Elliott
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA
| | - Neil E. Wilson
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA
| | - Fang Liu
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA
| | - Quy Cao
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA
| | - Sophie Swago
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA
| | - Paul S. Jacobs
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA
| | - Ryan Armbruster
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA
| | - Damodara Reddy
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA
| | - Joseph A. Baur
- Department of Physiology, Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA
| | - Walter R. Witschey
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA
| | - John A. Detre
- Department of Neurology, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA
| | - Ravinder Reddy
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
19
|
Fan Y, Wei Z, Zhang Y, Duan X. Enhancing L-asparagine Production Through In Vivo ATP Regeneration System Utilizing Glucose Metabolism of Escherichia coli. Appl Biochem Biotechnol 2024; 196:8685-8699. [PMID: 38900400 DOI: 10.1007/s12010-024-04982-8] [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] [Accepted: 06/05/2024] [Indexed: 06/21/2024]
Abstract
L-asparaginase synthetase, an ATP-dependent enzyme, necessitates ATP for its catalytic activity. However, the integration of L-asparaginase synthetase into industrial processes is curtailed by the prohibitive cost of ATP. To address this limitation, this study explores the construction of an efficient ATP regeneration system using the glucose metabolism of Escherichia coli, synergistically coupled with L-asparaginase synthetase catalysis. The optimal conditions for L-asparagine yield were determined in shake flasks. A total of 2.7 g/L was the highest yield achieved under specific parameters, including 0.1 mol/L of substrate, 0.2 mol/L glucose, 0.01 mol/L MgCl2 at pH 7.5, a temperature of 37 °C, and agitation at 300 r/min over 12 h. The process was then scaled to a 3-L fermenter, optimizing the addition rates of the substrate and magnesium chloride, and employing a constant glucose feed of 10 g/L/h. The scale-up process led to a significant enhancement in the production of L-asparagine. The yield of L-asparagine was increased to 38.49 g/L after 20 h of conversion, and the molar conversion rate reached 29.16%. This strategy has proven to be effective in improving the efficiency of L-asparagine production. When compared to in vitro ATP regeneration methods, this in vivo approach showcased superior efficiency and reduced costs. These findings furnish pivotal insights that may propel the enzymatic synthesis of L-asparagine toward viable industrial application.
Collapse
Affiliation(s)
- Yucheng Fan
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, 210037, Jiangsu, China
| | - Zijia Wei
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, 210037, Jiangsu, China
| | - Yuhua Zhang
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, 210037, Jiangsu, China
| | - Xuguo Duan
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, 210037, Jiangsu, China.
| |
Collapse
|
20
|
Ahmad F, Qaisar R. Nicotinamide riboside kinase 2: A unique target for skeletal muscle and cardiometabolic diseases. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167487. [PMID: 39216649 DOI: 10.1016/j.bbadis.2024.167487] [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: 06/05/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Myopathy leads to skeletal and cardiac muscle degeneration which is a major cause of physical disability and heart failure. Despite the therapeutic advancement the prevalence of particularly cardiac diseases is rising at an alarming rate and novel therapeutic targets are required. Nicotinamide riboside kinase-2 (NRK-2 or NMRK2) is a muscle-specific β1-integrin binding protein abundantly expressed in the skeletal muscle while only a trace amount is detected in the healthy cardiac muscle. The level in cardiac tissue is profoundly upregulated under pathogenic conditions such as ischemia and hypertension. NRK-2 was initially identified to regulate myoblast differentiation and to enhance the levels of NAD+, an important coenzyme that potentiates cellular energy production and stress resilience. Recent advancement has shown that NRK-2 critically regulates numerous cellular and molecular processes under pathogenic conditions to modulate the disease severity. Therefore, given its restricted expression in the cardiac and skeletal muscle, NRK-2 may serve as a unique therapeutic target. In this review, we provided a comprehensive overview of the diverse roles of NRK-2 played in different cardiac and muscular diseases and discussed the underlying molecular mechanisms in detail. Moreover, this review precisely examined how NRK-2 regulates metabolism in cardiac muscle, and how dysfunctional NRK-2 is associated with energetic deficit and impaired muscle function, manifesting various cardiac and skeletal muscle disease conditions.
Collapse
Affiliation(s)
- Firdos Ahmad
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Space Medicine Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Rizwan Qaisar
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Space Medicine Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
21
|
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
|
22
|
Kao G, Zhang XN, Nasertorabi F, Katz BB, Li Z, Dai Z, Zhang Z, Zhang L, Louie SG, Cherezov V, Zhang Y. Nicotinamide Riboside and CD38: Covalent Inhibition and Live-Cell Labeling. JACS AU 2024; 4:4345-4360. [PMID: 39610739 PMCID: PMC11600175 DOI: 10.1021/jacsau.4c00695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/03/2024] [Accepted: 10/21/2024] [Indexed: 11/30/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is required for a myriad of metabolic, signaling, and post-translational events in cells. Its levels in tissues and organs are closely associated with health conditions. The homeostasis of NAD+ is regulated by biosynthetic pathways and consuming enzymes. As a membrane-bound protein with robust NAD+ hydrolase activity, cluster of differentiation 38 (CD38) is a major degrader of NAD+. Deficiency or inhibition of CD38 enhances NAD+ levels in vivo, resulting in various therapeutic benefits. As a metabolic precursor of NAD+, nicotinamide mononucleotide can be rapidly hydrolyzed by CD38, whereas nicotinamide riboside (NR) lacks CD38 substrate activity. Given their structural similarities, we explored the inhibition potential of NR. To our surprise, NR exhibits marked inhibitory activity against CD38 by forming a stable ribosyl-ester bond with the glutamate residue 226 at the active site. Inspired by this discovery, we designed and synthesized a clickable NR featuring an azido substitution at the 5'-OH position. This cell-permeable NR analogue enables covalent labeling and imaging of both extracellular and intracellular CD38 in live cells. Our work discovers an unrecognized molecular function of NR and generates a covalent probe for health-related CD38. These findings offer new insights into the role of NR in modulating NAD+ metabolism and CD38-mediated signaling as well as an innovative tool for in-depth studies of CD38 in physiology and pathophysiology.
Collapse
Affiliation(s)
- Guoyun Kao
- Department
of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School
of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States
| | - Xiao-Nan Zhang
- Department
of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School
of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States
| | - Fariborz Nasertorabi
- Departments
of Biological Sciences and Chemistry, Bridge Institute, Michelson
Center for Convergent Bioscience, USC Structure Biology Center, University of Southern California, Los Angeles, California 90089, United States
| | - Benjamin B. Katz
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Zeyang Li
- Titus
Family Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy
and Pharmaceutical Sciences, University
of Southern California, Los Angeles, California 90089, United States
| | - Zhefu Dai
- Department
of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School
of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States
| | - Zeyu Zhang
- Department
of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School
of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States
| | - Lei Zhang
- Department
of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School
of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States
| | - Stan G. Louie
- Titus
Family Department of Clinical Pharmacy, Alfred E. Mann School of Pharmacy
and Pharmaceutical Sciences, University
of Southern California, Los Angeles, California 90089, United States
| | - Vadim Cherezov
- Bridge
Institute, University of Southern California, Los Angeles, California 90089, United States
- Department
of Chemistry, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California 90089, United States
| | - Yong Zhang
- Department
of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School
of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States
- Department
of Chemistry, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California 90089, United States
- Norris
Comprehensive Cancer Center, University
of Southern California, Los Angeles, California 90089, United States
- Research
Center for Liver Diseases, University of
Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
23
|
Chen F, Zhou D, Kong APS, Yim NT, Dai S, Chen YN, Hui LL. Effects of Nicotinamide Mononucleotide on Glucose and Lipid Metabolism in Adults: A Systematic Review and Meta-analysis of Randomised Controlled Trials. Curr Diab Rep 2024; 25:4. [PMID: 39531138 PMCID: PMC11557618 DOI: 10.1007/s11892-024-01557-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW Supplementation of nicotinamide mononucleotides (NMN) has been claimed to improve metabolic function. We reviewed human randomised controlled trials (RCTs) of NMN to evaluate its effect on markers of glucose and lipid metabolism. RECENT FINDINGS Eight RCTs on NMN (dosage ranged 250-2000 mg/d for a duration of 14 days to 12 weeks) involving a total of 342 middle-age/older adults (49% females, mainly non-diabetic) reporting at least one outcome on glucose control or lipid profile published in 2021-2023 were reviewed. The random-effects meta-analyses indicated no significant benefit of NMN on fasting glucose, fasting insulin, glycated hemoglobin, homeostatic model assessment for insulin resistance and lipid profile. Based on the small number of RCTs involving mainly relatively healthy adults, short-term supplementation of NMN of 250-2000 mg/d did not show significantly positive impacts on glucose control and lipid profile.
Collapse
Affiliation(s)
- Feng Chen
- School of Nursing, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Disheng Zhou
- School of Nursing, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Alice Pik-Shan Kong
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Nga Ting Yim
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Siyu Dai
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- School of Clinical Medicine, Hangzhou Normal University, Hangzhou, China.
- The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China.
| | - Yu Nan Chen
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Lai Ling Hui
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, China.
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
24
|
Yin L, Tong Y, Xie R, Zhang Z, Islam ZH, Zhang K, Burger J, Hoyt N, Kent EW, Marcum WA, Johnston C, Kanchetty R, Tetz Z, Stanisic S, Huang Y, Guo LW, Gong S, Wang B. Targeted NAD + repletion via biomimetic nanoparticle enables simultaneous management of intimal hyperplasia and accelerated re-endothelialization: A proof-of-concept study toward next-generation of endothelium-protective, anti-restenotic therapy. J Control Release 2024; 376:806-815. [PMID: 39461367 DOI: 10.1016/j.jconrel.2024.10.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 09/24/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
Endovascular interventions often fail due to restenosis, primarily caused by smooth muscle cell (SMC) proliferation, leading to intimal hyperplasia (IH). Current strategies to prevent restenosis are far from perfect and impose significant collateral damage on the fragile endothelial cell (EC), causing profound thrombotic risks. Nicotinamide adenine dinucleotide (NAD+) is a co-enzyme and signaling substrate implicated in redox and metabolic homeostasis, with a pleiotropic role in protecting against cardiovascular diseases. However, a functional link between NAD+ repletion and the delicate duo of IH and EC regeneration has yet to be established. NAD+ repletion has been historically challenging due to its poor cellular uptake and low bioavailability. We have recently invented the first nanocarrier that enables direct intracellular delivery of NAD+ in vivo. Combining the merits of this prototypic NAD+-loaded calcium phosphate (CaP) nanoparticle (NP) and biomimetic surface functionalization, we created a biomimetic P-NAD+-NP with platelet membrane coating, which enabled an injectable modality that targets IH with excellent biocompatibility. Using human cell primary culture, we demonstrated the benefits of NP-assisted NAD+ repletion in selectively inhibiting the excessive proliferation of aortic SMC, while differentially protecting aortic EC from apoptosis. Moreover, in a rat balloon angioplasty model, a single-dose treatment with intravenously injected P-NAD+-NP immediately post angioplasty not only mitigated IH, but also accelerated the regeneration of EC (re-endothelialization) in vivo in comparison to control groups (i.e., saline, free NAD+ solution, empty CaP-NP). Collectively, our current study provides proof-of-concept evidence supporting the role of targeted NAD+ repletion nanotherapy in managing restenosis and improving reendothelialization.
Collapse
Affiliation(s)
- Li Yin
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60603, USA; Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Department of Vascular Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang 310058, China
| | - Yao Tong
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Ruosen Xie
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Zhanpeng Zhang
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Zain Husain Islam
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Kaijie Zhang
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60603, USA; Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Department of Vascular Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang 310058, China
| | - Jacobus Burger
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Nicholas Hoyt
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Eric William Kent
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - William Aaron Marcum
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Campbell Johnston
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Rohan Kanchetty
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Zoe Tetz
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Sophia Stanisic
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Yitao Huang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Lian-Wang Guo
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Shaoqin Gong
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53715, USA.
| | - Bowen Wang
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60603, USA; Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
25
|
Pérez‐Rodríguez M, García‐Verdugo A, Sánchez‐Mendoza LM, Muñoz‐Martín A, Bolaños N, Pérez‐Sánchez C, Moreno JA, Burón MI, de Cabo R, González‐Reyes JA, Villalba JM. Cytochrome b 5 reductase 3 overexpression and dietary nicotinamide riboside supplementation promote distinctive mitochondrial alterations in distal convoluted tubules of mouse kidneys during aging. Aging Cell 2024; 23:e14273. [PMID: 39001573 PMCID: PMC11561664 DOI: 10.1111/acel.14273] [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: 02/05/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 11/15/2024] Open
Abstract
The kidney undergoes structural and physiological changes with age, predominantly studied in glomeruli and proximal tubules. However, limited knowledge is available about the impact of aging and anti-aging interventions on distal tubules. In this study, we investigated the effects of cytochrome b5 reductase 3 (CYB5R3) overexpression and/or dietary nicotinamide riboside (NR) supplementation on distal tubule mitochondria. Initially, transcriptomic data were analyzed to evaluate key genes related with distal tubules, CYB5R3, and NAD+ metabolism, showing significant differences between males and females in adult and old mice. Subsequently, our emphasis focused on assessing how these interventions, that have demonstrated the anti-aging potential, influenced structural parameters of distal tubule mitochondria, such as morphology and mass, as well as abundance, distance, and length of mitochondria-endoplasmic reticulum contact sites, employing an electron microscopy approach. Our findings indicate that both interventions have differential effects depending on the age and sex of the mice. Aging resulted in an increase in mitochondrial size and a decrease in mitochondrial abundance in males, while a reduction in abundance, size, and mitochondrial mass was observed in old females when compared with their adult counterparts. Combining both the interventions, CYB5R3 overexpression and dietary NR supplementation mitigated age-related changes; however, these effects were mainly accounted by NR in males and by transgenesis in females. In conclusion, the influence of CYB5R3 overexpression and dietary NR supplementation on distal tubule mitochondria depends on sex, genotype, and diet. This underscores the importance of incorporating these variables in subsequent studies to comprehensively address the multifaceted aspects of aging.
Collapse
Affiliation(s)
- M. Pérez‐Rodríguez
- Departamento de Biología Celular, Fisiología e InmunologíaUniversidad de Córdoba, Campus de Excelencia Internacional AgroalimentarioCórdobaSpain
| | - A. García‐Verdugo
- Departamento de Biología Celular, Fisiología e InmunologíaUniversidad de Córdoba, Campus de Excelencia Internacional AgroalimentarioCórdobaSpain
- Experimental Gerontology Section, Translational Gerontology BranchNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - L. M. Sánchez‐Mendoza
- Departamento de Biología Celular, Fisiología e InmunologíaUniversidad de Córdoba, Campus de Excelencia Internacional AgroalimentarioCórdobaSpain
| | - A. Muñoz‐Martín
- Departamento de Biología Celular, Fisiología e InmunologíaUniversidad de Córdoba, Campus de Excelencia Internacional AgroalimentarioCórdobaSpain
| | - N. Bolaños
- Departamento de Biología Celular, Fisiología e InmunologíaUniversidad de Córdoba, Campus de Excelencia Internacional AgroalimentarioCórdobaSpain
| | - C. Pérez‐Sánchez
- Departamento de Biología Celular, Fisiología e InmunologíaUniversidad de Córdoba, Campus de Excelencia Internacional AgroalimentarioCórdobaSpain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina SofíaCórdobaSpain
| | - J. A. Moreno
- Departamento de Biología Celular, Fisiología e InmunologíaUniversidad de Córdoba, Campus de Excelencia Internacional AgroalimentarioCórdobaSpain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina SofíaCórdobaSpain
| | - M. I. Burón
- Departamento de Biología Celular, Fisiología e InmunologíaUniversidad de Córdoba, Campus de Excelencia Internacional AgroalimentarioCórdobaSpain
| | - R. de Cabo
- Experimental Gerontology Section, Translational Gerontology BranchNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - J. A. González‐Reyes
- Departamento de Biología Celular, Fisiología e InmunologíaUniversidad de Córdoba, Campus de Excelencia Internacional AgroalimentarioCórdobaSpain
| | - J. M. Villalba
- Departamento de Biología Celular, Fisiología e InmunologíaUniversidad de Córdoba, Campus de Excelencia Internacional AgroalimentarioCórdobaSpain
| |
Collapse
|
26
|
Migaud ME, Ziegler M, Baur JA. Regulation of and challenges in targeting NAD + metabolism. Nat Rev Mol Cell Biol 2024; 25:822-840. [PMID: 39026037 DOI: 10.1038/s41580-024-00752-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2024] [Indexed: 07/20/2024]
Abstract
Nicotinamide adenine dinucleotide, in its oxidized (NAD+) and reduced (NADH) forms, is a reduction-oxidation (redox) co-factor and substrate for signalling enzymes that have essential roles in metabolism. The recognition that NAD+ levels fall in response to stress and can be readily replenished through supplementation has fostered great interest in the potential benefits of increasing or restoring NAD+ levels in humans to prevent or delay diseases and degenerative processes. However, much about the biology of NAD+ and related molecules remains poorly understood. In this Review, we discuss the current knowledge of NAD+ metabolism, including limitations of, assumptions about and unappreciated factors that might influence the success or contribute to risks of NAD+ supplementation. We highlight several ongoing controversies in the field, and discuss the role of the microbiome in modulating the availability of NAD+ precursors such as nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN), the presence of multiple cellular compartments that have distinct pools of NAD+ and NADH, and non-canonical NAD+ and NADH degradation pathways. We conclude that a substantial investment in understanding the fundamental biology of NAD+, its detection and its metabolites in specific cells and cellular compartments is needed to support current translational efforts to safely boost NAD+ levels in humans.
Collapse
Affiliation(s)
- Marie E Migaud
- Mitchell Cancer Institute, Department of Pharmacology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, USA.
| | - Mathias Ziegler
- Department of Biomedicine, University of Bergen, Bergen, Norway.
| | - Joseph A Baur
- Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
27
|
Feng H, Cao S, Fu S, Liu J, Gao Y, Dong Z, Cai T, Wen L, Xiong Z, Li S, Zhang X, Ma X, Li X. NMRK2 is an efficient diagnostic indicator for Xp11.2 translocation renal cell carcinoma. J Pathol 2024; 264:228-240. [PMID: 39092712 DOI: 10.1002/path.6340] [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: 01/15/2024] [Revised: 06/24/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024]
Abstract
Xp11.2 translocation renal cell carcinomas (tRCC) are a rare and highly malignant type of renal cancer, lacking efficient diagnostic indicators and therapeutic targets. Through the analysis of public databases and our cohort, we identified NMRK2 as a potential diagnostic marker for distinguishing Xp11.2 tRCC from kidney renal clear cell carcinoma (KIRC) and kidney renal papillary cell carcinoma (KIRP) due to its specific upregulation in Xp11.2 tRCC tissues. Mechanistically, we discovered that TFE3 fusion protein binds to the promoter of the NMRK2 gene, leading to its upregulation. Importantly, we established RNA- and protein-based diagnostic methods for identifying Xp11.2 tRCC based on NMRK2 expression levels, and the diagnostic performance of our methods was comparable to a dual-color break-apart fluorescence in situ hybridization assay. Moreover, we successfully identified fresh Xp11.2 tRCC tissues after surgical excision using our diagnostic methods and established an immortalized Xp11.2 tRCC cell line for further research purposes. Functional studies revealed that NMRK2 promotes the progression of Xp11.2 tRCC by upregulating the NAD+/NADH ratio, and supplementation with β-nicotinamide mononucleotide (NMN) or nicotinamide riboside chloride (NR), effectively rescued the phenotypes induced by the knockdown of NMRK2 in Xp11.2 tRCC. Taken together, these data introduce a new diagnostic indicator capable of accurately distinguishing Xp11.2 tRCC and highlight the possibility of developing novel targeted therapeutics. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/diagnosis
- Kidney Neoplasms/genetics
- Kidney Neoplasms/diagnosis
- Kidney Neoplasms/pathology
- Translocation, Genetic
- Chromosomes, Human, X/genetics
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Male
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Cell Line, Tumor
Collapse
Affiliation(s)
- Huayi Feng
- Chinese PLA Medical School, Beijing, PR China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Shouqing Cao
- College of Graduate, Hebei North University, Zhangjiakou, PR China
| | - Shihui Fu
- Department of Cardiology, Hainan Hospital of Chinese PLA General Hospital, Sanya, PR China
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, PR China
| | - Junxiao Liu
- Chinese PLA Medical School, Beijing, PR China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Yu Gao
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Zhouhuan Dong
- Department of Pathology, The First Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Tianwei Cai
- Chinese PLA Medical School, Beijing, PR China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Lequan Wen
- Chinese PLA Medical School, Beijing, PR China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Zhuang Xiong
- Chinese PLA Medical School, Beijing, PR China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Shangwei Li
- Chinese PLA Medical School, Beijing, PR China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Xu Zhang
- Chinese PLA Medical School, Beijing, PR China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Xin Ma
- Chinese PLA Medical School, Beijing, PR China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Xiubin Li
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| |
Collapse
|
28
|
Myong S, Nguyen AQ, Challa S. Biological Functions and Therapeutic Potential of NAD + Metabolism in Gynecological Cancers. Cancers (Basel) 2024; 16:3085. [PMID: 39272943 PMCID: PMC11394644 DOI: 10.3390/cancers16173085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/31/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an important cofactor for both metabolic and signaling pathways, with the dysregulation of NAD+ levels acting as a driver for diseases such as neurodegeneration, cancers, and metabolic diseases. NAD+ plays an essential role in regulating the growth and progression of cancers by controlling important cellular processes including metabolism, transcription, and translation. NAD+ regulates several metabolic pathways such as glycolysis, the citric acid (TCA) cycle, oxidative phosphorylation, and fatty acid oxidation by acting as a cofactor for redox reactions. Additionally, NAD+ acts as a cofactor for ADP-ribosyl transferases and sirtuins, as well as regulating cellular ADP-ribosylation and deacetylation levels, respectively. The cleavage of NAD+ by CD38-an NAD+ hydrolase expressed on immune cells-produces the immunosuppressive metabolite adenosine. As a result, metabolizing and maintaining NAD+ levels remain crucial for the function of various cells found in the tumor microenvironment, hence its critical role in tissue homeostasis. The NAD+ levels in cells are maintained by a balance between NAD+ biosynthesis and consumption, with synthesis being controlled by the Preiss-Handler, de novo, and NAD+ salvage pathways. The primary source of NAD+ synthesis in a variety of cell types is directed by the expression of the enzymes central to the three biosynthesis pathways. In this review, we describe the role of NAD+ metabolism and its synthesizing and consuming enzymes' control of cancer cell growth and immune responses in gynecologic cancers. Additionally, we review the ongoing efforts to therapeutically target the enzymes critical for NAD+ homeostasis in gynecologic cancers.
Collapse
Affiliation(s)
- Subin Myong
- The University of Chicago Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Anh Quynh Nguyen
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL 60637, USA
| | - Sridevi Challa
- The University of Chicago Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
29
|
Deng H, Ding D, Ma Y, Zhang H, Wang N, Zhang C, Yang G. Nicotinamide Mononucleotide: Research Process in Cardiovascular Diseases. Int J Mol Sci 2024; 25:9526. [PMID: 39273473 PMCID: PMC11394709 DOI: 10.3390/ijms25179526] [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: 07/29/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential metabolite that plays a crucial role in diverse biological processes, including energy metabolism, gene expression, DNA repair, and mitochondrial function. An aberrant NAD+ level mediates the development of cardiovascular dysfunction and diseases. Both in vivo and in vitro studies have demonstrated that nicotinamide mononucleotide (NMN), as a NAD+ precursor, alleviates the development of cardiovascular diseases such as heart failure, atherosclerosis, and myocardial ischemia/reperfusion injury. Importantly, NMN has suggested pharmacological activities mostly through its involvement in NAD+ biosynthesis. Several clinical studies have been conducted to investigate the efficacy and safety of NMN supplementation, indicating its potential role in cardiovascular protection without significant adverse effects. In this review, we systematically summarize the impact of NMN as a nutraceutical and potential therapeutic drug on cardiovascular diseases and emphasize the correlation between NMN supplementation and cardiovascular protection.
Collapse
Affiliation(s)
- Haoyuan Deng
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Ding Ding
- School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Yu Ma
- Department of Health Toxicology, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Hao Zhang
- School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Ningning Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Cong Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Guang Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
30
|
Shi C, Wen Z, Yang Y, Shi L, Liu D. NAD+ metabolism and therapeutic strategies in cardiovascular diseases. ATHEROSCLEROSIS PLUS 2024; 57:1-12. [PMID: 38974325 PMCID: PMC11223091 DOI: 10.1016/j.athplu.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/25/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a central and pleiotropic metabolite involved in cellular energy metabolism, cell signaling, DNA repair, and protein modifications. Cardiovascular diseases (CVDs) are the leading cause of death worldwide. Metabolic stress and aging directly affect the cardiovascular system. Compelling data suggest that NAD + levels decrease with age, obesity, and hypertension, which are all notable risk factors for CVD. In addition, the therapeutic elevation of NAD + levels reduces chronic low-grade inflammation, reactivates autophagy and mitochondrial biogenesis, and enhances oxidative metabolism in vascular cells of humans and rodents with vascular disorders. In preclinical models, NAD + boosting can also expand the health span, prevent metabolic syndrome, and decrease blood pressure. Moreover, NAD + storage by genetic, pharmacological, or natural dietary NAD + -increasing strategies has recently been shown to be effective in improving the pathophysiology of cardiac and vascular health in different animal models, and human health. Here, we review and discuss NAD + -related mechanisms pivotal for vascular health and summarize recent experimental evidence in NAD + research directly related to vascular disease, including atherosclerosis, and coronary artery disease. Finally, we comparatively assess distinct NAD + precursors for their clinical efficacy and the efficiency of NAD + elevation in the treatment of major CVD. These findings may provide ideas for new therapeutic strategies to prevent and treat CVD in the clinic.
Collapse
Affiliation(s)
- Chongxu Shi
- Nantong Laboratory of Development and Diseases, School of Life Science, Nantong University, Nantong, China
| | - Zhaozhi Wen
- Nantong Laboratory of Development and Diseases, School of Life Science, Nantong University, Nantong, China
| | - Yihang Yang
- Nantong Laboratory of Development and Diseases, School of Life Science, Nantong University, Nantong, China
| | - Linsheng Shi
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Dong Liu
- Nantong Laboratory of Development and Diseases, School of Life Science, Nantong University, Nantong, China
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
- Co-Innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, China
| |
Collapse
|
31
|
He WM, Yang JY, Zhao ZY, Xiao W, Li WH, Zhao YJ. The Fluorinated NAD Precursors Enhance FK866 Cytotoxicity by Activating SARM1 in Glioblastoma Cells. BIOLOGY 2024; 13:649. [PMID: 39336077 PMCID: PMC11429243 DOI: 10.3390/biology13090649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024]
Abstract
Glioblastoma, a formidable brain tumor characterized by dysregulated NAD metabolism, poses a significant therapeutic challenge. The NAMPT inhibitor FK866, which induces NAD depletion, has shown promise in controlling tumor proliferation and modifying the tumor microenvironment. However, the clinical efficacy of FK866 as a single drug therapy for glioma is limited. In this study, we aim to disrupt NAD metabolism using fluorinated NAD precursors and explore their synergistic effect with FK866 in inducing cytotoxicity in glioblastoma cells. The synthesized analogue of nicotinamide riboside (NR), ara-F nicotinamide riboside (F-NR), inhibits nicotinamide ribose kinase (NRK) activity in vitro, reduces cellular NAD levels, and enhances FK866's cytotoxicity in U251 glioblastoma cells, indicating a collaborative impact on cell death. Metabolic analyses reveal that F-NR undergoes conversion to fluorinated nicotinamide mononucleotide (F-NMN) and other metabolites, highlighting the intact NAD metabolic pathway in glioma cells. The activation of SARM1 by F-NMN, a potent NAD-consuming enzyme, is supported by the synergistic effect of CZ-48, a cell-permeable SARM1 activator. Temporal analysis underscores the sequential nature of events, establishing NAD depletion as a precursor to ATP depletion and eventual massive cell death. This study not only elucidates the molecular intricacies of glioblastoma cell death but also proposes a promising strategy to enhance FK866 efficacy through fluorinated NAD precursors, offering potential avenues for innovative therapeutic interventions in the challenging landscape of glioblastoma treatment.
Collapse
Affiliation(s)
- Wei Ming He
- State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China; (W.M.H.); (Z.Y.Z.)
| | - Jian Yuan Yang
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China;
| | - Zhi Ying Zhao
- State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China; (W.M.H.); (Z.Y.Z.)
| | - Weimin Xiao
- Shenzhen Academy of Metrology and Quality Inspection, Shenzhen 518110, China;
| | - Wan Hua Li
- State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China; (W.M.H.); (Z.Y.Z.)
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China;
| | - Yong Juan Zhao
- State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China; (W.M.H.); (Z.Y.Z.)
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China;
| |
Collapse
|
32
|
Cordeiro EF, Marzola EL, Maekawa RS, Santos MR, Assunção LG, Massafera MP, Oliveira JD, Batista TG, Sales MCOPD, Maria-Engler SS, Di Mascio P, Medeiros MHD, Ronsein GE, Loureiro APDM. Nicotinamide riboside Induced Energy Stress and Metabolic Reprogramming in BEAS-2B Cells. Chem Res Toxicol 2024; 37:1246-1268. [PMID: 38990804 PMCID: PMC11337214 DOI: 10.1021/acs.chemrestox.3c00312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
Nicotinamide riboside (NR), a NAD+ precursor, has received attention due to several health benefits it has induced in experimental models. Studies in cultured cells, animals, and humans consistently show increased NAD+ availability after NR supplementation, which is considered the only mode of NR action that leads to health benefits. In the present study, we show that a persistently low NR concentration (1 μM) in the growth medium of BEAS-2B human cells, grown in a monolayer, induces energy stress, which precedes a cellular NAD+ increase after 192 h. NR concentrations greater than 1 μM under the specified conditions were cytotoxic in the 2D cell culture model, while all concentrations tested in the 3D cell culture model (BEAS-2B cell spheroids exposed to 1, 5, 10, and 50 μM NR) induced apoptosis. Shotgun proteomics revealed that NR modulated the abundance of proteins, agreeing with the observed effects on cellular energy metabolism and cell growth or survival. Energy stress may activate pathways that lead to health benefits such as cancer prevention. Accordingly, the premalignant 1198 cell line was more sensitive to NR cytotoxicity than the phenotypically normal parent BEAS-2B cell line. The role of a mild energy stress induced by low concentrations of NR in its beneficial effects deserves further investigation. On the other hand, strategies to increase the bioavailability of NR require attention to toxic effects that may arise.
Collapse
Affiliation(s)
- Everson
Willian Fialho Cordeiro
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade
de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes 580, CEP 05508-000 São Paulo, Brazil
| | - Elisabete Leide Marzola
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade
de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes 580, CEP 05508-000 São Paulo, Brazil
| | - Ricardo Soei Maekawa
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade
de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes 580, CEP 05508-000 São Paulo, Brazil
| | - Matheus Relvas
dos Santos
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade
de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes 580, CEP 05508-000 São Paulo, Brazil
| | - Lucas Gade Assunção
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade
de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes 580, CEP 05508-000 São Paulo, Brazil
| | - Mariana Pereira Massafera
- Departamento
de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, CEP 05508-000 São Paulo, Brazil
| | - Joseana de Oliveira
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade
de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes 580, CEP 05508-000 São Paulo, Brazil
| | - Thainá Gomes
Cury Batista
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade
de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes 580, CEP 05508-000 São Paulo, Brazil
| | - Maria Cármen Oliveira Pinho de Sales
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade
de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes 580, CEP 05508-000 São Paulo, Brazil
| | - Silvya Stuchi Maria-Engler
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade
de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes 580, CEP 05508-000 São Paulo, Brazil
| | - Paolo Di Mascio
- Departamento
de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, CEP 05508-000 São Paulo, Brazil
| | - Marisa Helena
Gennari de Medeiros
- Departamento
de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, CEP 05508-000 São Paulo, Brazil
| | - Graziella Eliza Ronsein
- Departamento
de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, CEP 05508-000 São Paulo, Brazil
| | - Ana Paula de Melo Loureiro
- Departamento
de Análises Clínicas e Toxicológicas, Faculdade
de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes 580, CEP 05508-000 São Paulo, Brazil
| |
Collapse
|
33
|
Zhang J, Poon ETC, Wong SHS. Efficacy of oral nicotinamide mononucleotide supplementation on glucose and lipid metabolism for adults: a systematic review with meta-analysis on randomized controlled trials. Crit Rev Food Sci Nutr 2024:1-19. [PMID: 39116016 DOI: 10.1080/10408398.2024.2387324] [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: 08/10/2024]
Abstract
A surge of public interest in NMN supplementation has been observed in recent years. However, whether NMN supplements are effective in improving metabolic health remains unclear. The objective of the review was to assess the effects of NMN supplementation on fasting glucose, triglycerides, total cholesterol, LDL-C, and HDL-C in adults. Studies were located by searching four databases (PubMed, Embase, Cochrane, and Web of Science). Two reviewers independently conducted title/abstract and full-text screening, data extraction, and risk-of-bias assessment. Of the 4049 records reviewed, 12 studies with a total of 513 participants met the criteria for analysis. Random-effects meta-analyses found an overall significant effect of NMN supplementation in elevating blood NAD levels. However, most of the clinically relevant outcomes were not significantly different between NMN supplementation and control group. Risk-of-bias assessment using RoB2 showed some concerns in seven studies and high risk of bias in the other five studies. Together, our findings suggest that an exaggeration of the benefits of NMN supplementation may exist in the field. Although the limited number of eligible studies was sufficiently powered to detect changes in the abovementioned primary outcomes, more studies are needed to conclude about the exact effects of NMN supplementation.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Department of Sports Science & Physical Education, The Chinese University of Hong Kong, Shatin, New Territory, Hong Kong
| | - Eric Tsz-Chun Poon
- Department of Sports Science & Physical Education, The Chinese University of Hong Kong, Shatin, New Territory, Hong Kong
| | - Stephen Heung-Sang Wong
- Department of Sports Science & Physical Education, The Chinese University of Hong Kong, Shatin, New Territory, Hong Kong
| |
Collapse
|
34
|
Yang Y, Huang J, Xie L, Wang Y, Guo S, Wang M, Shao X, Liu W, Wang Y, Li Q, Wu X, Zhang Z, Zeng F, Gong W. Nicotinamide protects against diabetic kidney disease through regulation of Sirt1. Endocrine 2024; 85:638-648. [PMID: 38446387 PMCID: PMC11291543 DOI: 10.1007/s12020-024-03721-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/30/2024] [Indexed: 03/07/2024]
Abstract
PURPOSE To investigate the effect of nicotinamide (Nam) on diabetic kidney disease (DKD) in mice and explore its mechanism. METHODS Thirty DBA/2 J mice were randomly assigned to three groups. After 8 weeks of hyperglycemia induced by streptozocin (STZ), Nam and saline were administrated to STZ + Nam and STZ + NS mice, respectively, for 8 weeks. Non-diabetic mice (NDM) were used as control group. Twenty In2-/- Akita mice were randomly divided into two groups. After 8 weeks of hyperglycemia, Nam and saline were administered to Akita + Nam and Akita + NS mice, respectively, for 6 weeks. Wild-type littermates were used as control group. Markers of renal injury were analyzed, and the molecular mechanisms were explored in human proximal tubular HK2 cells. RESULTS Urinary albumin-to-creatinine ratio (UACR) and kidney injury molecule 1 (KIM-1) decreased in the STZ + Nam and Akita + Nam groups. Pathological analysis showed that Nam improved the structure of glomerular basement membrane, ameliorated glomerular sclerosis, and decreased the accumulation of extracellular matrix and collagen. Compared to the diabetic control group, renal fibrosis, inflammation, and oxidative stress were reduced in the Nam-treated mice. The expression of sirtuin 1 (Sirt1) in human proximal tubular HK2 cells was inhibited by high glucose and Nam treatment enhanced its expression. However, in HK2 cells with Sirt1 knockdown, the protective effect of Nam was abolished, indicating that the beneficial effect of Nam was partially dependent on Sirt1. CONCLUSIONS Nam has a renoprotective effect against renal injury caused by hyperglycemia and may be a potential target for the treatment of DKD.
Collapse
Affiliation(s)
- Yeping Yang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jinya Huang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Lijie Xie
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yilin Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shizhe Guo
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Meng Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xiaoqing Shao
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Wenjuan Liu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yi Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qin Li
- Division of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200011, China
| | - Xia Wu
- Department of Endocrinology and Metabolism, Jing'an District Center Hospital of Shanghai, Shanghai, 200040, China
| | - Zhaoyun Zhang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai, 200040, China
| | - Fangfang Zeng
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Wei Gong
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
35
|
Zhan J, Huang L, Niu L, Lu W, Sun C, Liu S, Ding Z, Li E. Regulation of CD73 on NAD metabolism: Unravelling the interplay between tumour immunity and tumour metabolism. Cell Commun Signal 2024; 22:387. [PMID: 39090604 PMCID: PMC11292923 DOI: 10.1186/s12964-024-01755-y] [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: 06/05/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024] Open
Abstract
CD73, a cell surface-bound nucleotidase, serves as a crucial metabolic and immune checkpoint. Several studies have shown that CD73 is widely expressed on immune cells and plays a critical role in immune escape, cell adhesion and migration as a costimulatory molecule for T cells and a factor in adenosine production. However, recent studies have revealed that the protumour effects of CD73 are not limited to merely inhibiting the antitumour immune response. Nicotinamide adenine dinucleotide (NAD+) is a vital bioactive molecule in organisms that plays essential regulatory roles in diverse biological processes within tumours. Accumulating evidence has demonstrated that CD73 is involved in the transport and metabolism of NAD, thereby regulating tumour biological processes to promote growth and proliferation. This review provides a holistic view of CD73-regulated NAD + metabolism as a complex network and further highlights the emerging roles of CD73 as a novel target for cancer therapies.
Collapse
Affiliation(s)
- Jianhao Zhan
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- HuanKui Academy, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Le Huang
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China
- HuanKui Academy, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Liyan Niu
- HuanKui Academy, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Wenhui Lu
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Chengpeng Sun
- HuanKui Academy, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Shanshan Liu
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi province, China
| | - Zijun Ding
- School of Ophthalmology and Optometry, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Enliang Li
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, 330006, China.
| |
Collapse
|
36
|
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
|
37
|
Zheng C, Li Y, Wu X, Gao L, Chen X. Advances in the Synthesis and Physiological Metabolic Regulation of Nicotinamide Mononucleotide. Nutrients 2024; 16:2354. [PMID: 39064797 PMCID: PMC11279976 DOI: 10.3390/nu16142354] [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: 06/25/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Nicotinamide mononucleotide (NMN), the direct precursor of nicotinamide adenine dinucleotide (NAD+), is involved in the regulation of many physiological and metabolic reactions in the body. NMN can indirectly affect cellular metabolic pathways, DNA repair, and senescence, while also being essential for maintaining tissues and dynamic metabolic equilibria, promoting healthy aging. Therefore, NMN has found many applications in the food, pharmaceutical, and cosmetics industries. At present, NMN synthesis strategies mainly include chemical synthesis and biosynthesis. Despite its potential benefits, the commercial production of NMN by organic chemistry approaches faces environmental and safety problems. With the rapid development of synthetic biology, it has become possible to construct microbial cell factories to produce NMN in a cost-effective way. In this review, we summarize the chemical and biosynthetic strategies of NMN, offering an overview of the recent research progress on host selection, chassis cell optimization, mining of key enzymes, metabolic engineering, and adaptive fermentation strategies. In addition, we also review the advances in the role of NMN in aging, metabolic diseases, and neural function. This review provides comprehensive technical guidance for the efficient biosynthesis of NMN as well as a theoretical basis for its application in the fields of food, medicine, and cosmetics.
Collapse
Affiliation(s)
- Chuxiong Zheng
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China;
| | - Yumeng Li
- National Technology Innovation Center for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, No. 32, Xiqi Road, Tianjin Airport Economic Park, Tianjin 300308, China; (Y.L.); (X.W.)
| | - Xin Wu
- National Technology Innovation Center for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, No. 32, Xiqi Road, Tianjin Airport Economic Park, Tianjin 300308, China; (Y.L.); (X.W.)
| | - Le Gao
- National Technology Innovation Center for Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, No. 32, Xiqi Road, Tianjin Airport Economic Park, Tianjin 300308, China; (Y.L.); (X.W.)
| | - Xiaoyi Chen
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China;
| |
Collapse
|
38
|
Devereaux J, Robinson AM, Stavely R, Davidson M, Dargahi N, Ephraim R, Kiatos D, Apostolopoulos V, Nurgali K. Alterations in tryptophan metabolism and de novo NAD + biosynthesis within the microbiota-gut-brain axis in chronic intestinal inflammation. Front Med (Lausanne) 2024; 11:1379335. [PMID: 39015786 PMCID: PMC11250461 DOI: 10.3389/fmed.2024.1379335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Background Inflammatory bowel disease is an incurable and idiopathic disease characterized by recurrent gastrointestinal tract inflammation. Tryptophan metabolism in mammalian cells and some gut microbes comprise intricate chemical networks facilitated by catalytic enzymes that affect the downstream metabolic pathways of de novo nicotinamide adenine dinucleotide (NAD+) synthesis. It is hypothesized that a correlation exists between tryptophan de novo NAD+ synthesis and chronic intestinal inflammation. Methods Transcriptome analysis was performed using high-throughput sequencing of mRNA extracted from the distal colon and brain tissue of Winnie mice with spontaneous chronic colitis and C57BL/6 littermates. Metabolites were assessed using ultra-fast liquid chromatography to determine differences in concentrations of tryptophan metabolites. To evaluate the relative abundance of gut microbial genera involved in tryptophan and nicotinamide metabolism, we performed 16S rRNA gene amplicon sequencing of fecal samples from C57BL/6 and Winnie mice. Results Tryptophan and nicotinamide metabolism-associated gene expression was altered in distal colons and brains of Winnie mice with chronic intestinal inflammation. Changes in these metabolic pathways were reflected by increases in colon tryptophan metabolites and decreases in brain tryptophan metabolites in Winnie mice. Furthermore, dysbiosis of gut microbiota involved in tryptophan and nicotinamide metabolism was evident in fecal samples from Winnie mice. Our findings shed light on the physiological alterations in tryptophan metabolism, specifically, its diversion from the serotonergic pathway toward the kynurenine pathway and consequential effects on de novo NAD+ synthesis in chronic intestinal inflammation. Conclusion The results of this study reveal differential expression of tryptophan and nicotinamide metabolism-associated genes in the distal colon and brain in Winnie mice with chronic intestinal inflammation. These data provide evidence supporting the role of tryptophan metabolism and de novo NAD+ synthesis in IBD pathophysiology.
Collapse
Affiliation(s)
- Jeannie Devereaux
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Ainsley M. Robinson
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- School of Rural Health, La Trobe University, Melbourne, VIC, Australia
- Department of Medicine, Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Rhian Stavely
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Majid Davidson
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Narges Dargahi
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Ramya Ephraim
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Dimitros Kiatos
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Department of Medicine, Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
- Immunology Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Department of Medicine, Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
- Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia
| |
Collapse
|
39
|
Benjamin C, Crews R. Nicotinamide Mononucleotide Supplementation: Understanding Metabolic Variability and Clinical Implications. Metabolites 2024; 14:341. [PMID: 38921475 PMCID: PMC11205942 DOI: 10.3390/metabo14060341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Recent years have seen a surge in research focused on NAD+ decline and potential interventions, and despite significant progress, new discoveries continue to highlight the complexity of NAD+ biology. Nicotinamide mononucleotide (NMN), a well-established NAD+ precursor, has garnered considerable interest due to its capacity to elevate NAD+ levels and induce promising health benefits in preclinical models. Clinical trials investigating NMN supplementation have yielded variable outcomes while shedding light on the intricacies of NMN metabolism and revealing the critical roles played by gut microbiota and specific cellular uptake pathways. Individual variability in factors such as lifestyle, health conditions, genetics, and gut microbiome composition likely contributes to the observed discrepancies in clinical trial results. Preliminary evidence suggests that NMN's effects may be context-dependent, varying based on a person's physiological state. Understanding these nuances is critical for definitively assessing the impact of manipulating NAD+ levels through NMN supplementation. Here, we review NMN metabolism, focusing on current knowledge, pinpointing key areas where further research is needed, and outlining future directions to advance our understanding of its potential clinical significance.
Collapse
|
40
|
Rahman SU, Qadeer A, Wu Z. Role and Potential Mechanisms of Nicotinamide Mononucleotide in Aging. Aging Dis 2024; 15:565-583. [PMID: 37548938 PMCID: PMC10917541 DOI: 10.14336/ad.2023.0519-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/19/2023] [Indexed: 08/08/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) has recently attracted much attention due to its role in aging and lifespan extension. NAD+ directly and indirectly affects many cellular processes, including metabolic pathways, DNA repair, and immune cell activities. These mechanisms are critical for maintaining cellular homeostasis. However, the decline in NAD+ levels with aging impairs tissue function, which has been associated with several age-related diseases. In fact, the aging population has been steadily increasing worldwide, and it is important to restore NAD+ levels and reverse or delay these age-related disorders. Therefore, there is an increasing demand for healthy products that can mitigate aging, extend lifespan, and halt age-related consequences. In this case, several studies in humans and animals have targeted NAD+ metabolism with NAD+ intermediates. Among them, nicotinamide mononucleotide (NMN), a precursor in the biosynthesis of NAD+, has recently received much attention from the scientific community for its anti-aging properties. In model organisms, ingestion of NMN has been shown to improve age-related diseases and probably delay death. Here, we review aspects of NMN biosynthesis and the mechanism of its absorption, as well as potential anti-aging mechanisms of NMN, including recent preclinical and clinical tests, adverse effects, limitations, and perceived challenges.
Collapse
Affiliation(s)
- Sajid Ur Rahman
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Abdul Qadeer
- Institute for Infectious Diseases and Vaccine Development, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Ziyun Wu
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
41
|
Qiu S, Shao S, Zhang Y, Zhang Y, Yin J, Hong Y, Yang J, Tan X, Di C. Comparison of protective effects of nicotinamide mononucleotide and nicotinamide riboside on DNA damage induced by cisplatin in HeLa cells. Biochem Biophys Rep 2024; 37:101655. [PMID: 38333051 PMCID: PMC10851195 DOI: 10.1016/j.bbrep.2024.101655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/15/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Background Previous studies have shown that the nicotinamide adenine dinucleotide (NAD+) precursors, nicotinamide mononucleotide (NMN) and nicotinamide riboside (NR), protect against endogenously or exogenously induced DNA damage. However, whether the two compounds have the same or different efficacy against DNA damage is not clear. In the current study, we systematically compared the effects of NMN and NR on cisplatin-induced DNA damage in HeLa cells. Methods To evaluate the protective effects of NMN or NR, HeLa cells were pretreated with different doses of NMN or NR followed with 10 μM of cisplatin treatment. Cell viability was examined by Trypan blue staining assay. For observing the DNA damage repair process, HeLa cells were treated with 10 μM of cisplatin for 12 h, followed with 10 mM NMN or NR treatment for another 8, 16, 24, or 32 h, DNA damage levels were assessed for each time point by immunofluorescent staining against phosphor-H2AX (γH2AX) and alkaline comet assay. The effects of NMN and NR on intracellular NAD+ and reactive oxygen species (ROS) levels were also determined. Results NMN and NR treatment alone did not have any significant effects on cell viability, however, both can protect HeLa cells from cisplatin-induced decrease of cell viability with similar efficacy in a dose-dependent manner. On the other hand, while both can reduce the DNA damage levels in cisplatin-treated cells, NR exhibited better protective effect. However, both appeared to boost the DNA damage repair process with similar efficacy. NMN or NR treatment alone could increase cellular NAD+ levels, and both can reverse cisplatin-induced decrease of NAD+ levels. Finally, while neither NMN nor NR affected cellular ROS levels, both inhibited cisplatin-induced increase of ROS with no significant difference between them. Conclusion NR have a better protective effect against cisplatin-induced DNA damage than NMN.
Collapse
Affiliation(s)
- Shuting Qiu
- Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Shihan Shao
- Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Yunheng Zhang
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Yingying Zhang
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Jie Yin
- Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
| | - Yu Hong
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Jun Yang
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Xiaohua Tan
- Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Chunhong Di
- Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
- School of Public Health, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
42
|
Yan G, Han Z, Kwon Y, Jousma J, Nukala SB, Prosser BL, Du X, Pinho S, Ong SB, Lee WH, Ong SG. Integrated Stress Response Potentiates Ponatinib-Induced Cardiotoxicity. Circ Res 2024; 134:482-501. [PMID: 38323474 PMCID: PMC10940206 DOI: 10.1161/circresaha.123.323683] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/22/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Mitochondrial dysfunction is a primary driver of cardiac contractile failure; yet, the cross talk between mitochondrial energetics and signaling regulation remains obscure. Ponatinib, a tyrosine kinase inhibitor used to treat chronic myeloid leukemia, is among the most cardiotoxic tyrosine kinase inhibitors and causes mitochondrial dysfunction. Whether ponatinib-induced mitochondrial dysfunction triggers the integrated stress response (ISR) to induce ponatinib-induced cardiotoxicity remains to be determined. METHODS Using human induced pluripotent stem cells-derived cardiomyocytes and a recently developed mouse model of ponatinib-induced cardiotoxicity, we performed proteomic analysis, molecular and biochemical assays to investigate the relationship between ponatinib-induced mitochondrial stress and ISR and their role in promoting ponatinib-induced cardiotoxicity. RESULTS Proteomic analysis revealed that ponatinib activated the ISR in cardiac cells. We identified GCN2 (general control nonderepressible 2) as the eIF2α (eukaryotic translation initiation factor 2α) kinase responsible for relaying mitochondrial stress signals to trigger the primary ISR effector-ATF4 (activating transcription factor 4), upon ponatinib exposure. Mechanistically, ponatinib treatment exerted inhibitory effects on ATP synthase activity and reduced its expression levels resulting in ATP deficits. Perturbed mitochondrial function resulting in ATP deficits then acts as a trigger of GCN2-mediated ISR activation, effects that were negated by nicotinamide mononucleotide, an NAD+ precursor, supplementation. Genetic inhibition of ATP synthase also activated GCN2. Interestingly, we showed that the decreased abundance of ATP also facilitated direct binding of ponatinib to GCN2, unexpectedly causing its activation most likely because of a conformational change in its structure. Importantly, administering an ISR inhibitor protected human induced pluripotent stem cell-derived cardiomyocytes against ponatinib. Ponatinib-treated mice also exhibited reduced cardiac function, effects that were attenuated upon systemic ISRIB administration. Importantly, ISRIB does not affect the antitumor effects of ponatinib in vitro. CONCLUSIONS Neutralizing ISR hyperactivation could prevent or reverse ponatinib-induced cardiotoxicity. The findings that compromised ATP production potentiates GCN2-mediated ISR activation have broad implications across various cardiac diseases. Our results also highlight an unanticipated role of ponatinib in causing direct activation of a kinase target despite its role as an ATP-competitive kinase inhibitor.
Collapse
Affiliation(s)
- Gege Yan
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Zhenbo Han
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Youjeong Kwon
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Jordan Jousma
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Sarath Babu Nukala
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Benjamin L Prosser
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xiaoping Du
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Sandra Pinho
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Sang-Bing Ong
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Centre for Cardiovascular Genomics and Medicine (CCGM), Lui Che Woo Institute of Innovative Medicine, CUHK, Hong Kong SAR, China
- Hong Kong Hub of Pediatric Excellence (HK HOPE), Hong Kong Children’s Hospital (HKCH), Kowloon Bay, Hong Kong SAR, China
- Kunming Institute of Zoology – The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Neural, Vascular, and Metabolic Biology Thematic Research Program, School of Biomedical Sciences (SBS), Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Won Hee Lee
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, USA
| | - Sang-Ging Ong
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, USA
| |
Collapse
|
43
|
Norambuena-Soto I, Deng Y, Brenner C, Lavandero S, Wang ZV. NAD in pathological cardiac remodeling: Metabolic regulation and beyond. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167038. [PMID: 38281710 PMCID: PMC10922927 DOI: 10.1016/j.bbadis.2024.167038] [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: 08/12/2023] [Revised: 01/05/2024] [Accepted: 01/19/2024] [Indexed: 01/30/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD) coenzymes are carriers of high energy electrons in metabolism and also play critical roles in numerous signaling pathways. NAD metabolism is decreased in various cardiovascular diseases. Importantly, stimulation of NAD biosynthesis protects against heart disease under different pathological conditions. In this review, we describe pathways for both generation and catabolism of NAD coenzymes and the respective changes of these pathways in the heart under cardiac diseases, including pressure overload, myocardial infarction, cardiometabolic disease, cancer treatment cardiotoxicity, and heart failure. We next provide an update on the strategies and treatments to increase NAD levels, such as supplementation of NAD precursors, in the heart that prevent or reverse cardiomyopathy. We also introduce the approaches to manipulate NAD consumption enzymes to ameliorate cardiac disease. Finally, we discuss the mechanisms associated with improvements in cardiac function by NAD coenzymes, differentiating between mitochondria-dependent effects and those independent of mitochondrial metabolism.
Collapse
Affiliation(s)
- Ignacio Norambuena-Soto
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Químicas y Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago 8380494, Chile
| | - Yingfeng Deng
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Charles Brenner
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Químicas y Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago 8380494, Chile; Cardiology Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8573, USA.
| | - Zhao V Wang
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
44
|
Cheng F, Li KX, Wu SS, Liu HY, Li H, Shen Q, Xue YP, Zheng YG. Biosynthesis of Nicotinamide Mononucleotide: Synthesis Method, Enzyme, and Biocatalytic System. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:3302-3313. [PMID: 38330904 DOI: 10.1021/acs.jafc.3c09217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Nicotinamide mononucleotide (NMN) has garnered substantial interest as a functional food product. Industrial NMN production relies on chemical methods, facing challenges in separation, purification, and regulatory complexities, leading to elevated prices. In contrast, NMN biosynthesis through fermentation or enzyme catalysis offers notable benefits like eco-friendliness, recyclability, and efficiency, positioning it as a primary avenue for future NMN synthesis. Enzymatic NMN synthesis encompasses the nicotinamide-initial route and nicotinamide ribose-initial routes. Key among these is nicotinamide riboside kinase (NRK), pivotal in the latter route. The NRK-mediated biosynthesis is emerging as a prominent trend due to its streamlined route, simplicity, and precise specificity. The essential aspect is to obtain an engineered NRK that exhibits elevated activity and robust stability. This review comprehensively assesses diverse NMN synthesis methods, offering valuable insights into efficient, sustainable, and economical production routes. It spotlights the emerging NRK-mediated biosynthesis pathway and its significance. The establishment of an adenosine triphosphate (ATP) regeneration system plays a pivotal role in enhancing NMN synthesis efficiency through NRK-catalyzed routes. The review aims to be a reference for researchers developing green and sustainable NMN synthesis, as well as those optimizing NMN production.
Collapse
Affiliation(s)
- Feng Cheng
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou 310014, P. R. China
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Engineering Research Center of Bioconversion and Biopurification of the Ministry of Education, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Ke-Xin Li
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou 310014, P. R. China
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Engineering Research Center of Bioconversion and Biopurification of the Ministry of Education, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Shan-Shan Wu
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou 310014, P. R. China
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Engineering Research Center of Bioconversion and Biopurification of the Ministry of Education, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Hai-Yun Liu
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou 310014, P. R. China
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Engineering Research Center of Bioconversion and Biopurification of the Ministry of Education, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Huan Li
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou 310014, P. R. China
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Engineering Research Center of Bioconversion and Biopurification of the Ministry of Education, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Qi Shen
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou 310014, P. R. China
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Engineering Research Center of Bioconversion and Biopurification of the Ministry of Education, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Ya-Ping Xue
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou 310014, P. R. China
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Engineering Research Center of Bioconversion and Biopurification of the Ministry of Education, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| | - Yu-Guo Zheng
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, 18 Chaowang Road, Hangzhou 310014, P. R. China
- The National and Local Joint Engineering Research Center for Biomanufacturing of Chiral Chemicals, Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Engineering Research Center of Bioconversion and Biopurification of the Ministry of Education, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, P. R. China
| |
Collapse
|
45
|
Szot JO, Cuny H, Martin EM, Sheng DZ, Iyer K, Portelli S, Nguyen V, Gereis JM, Alankarage D, Chitayat D, Chong K, Wentzensen IM, Vincent-Delormé C, Lermine A, Burkitt-Wright E, Ji W, Jeffries L, Pais LS, Tan TY, Pitt J, Wise CA, Wright H, Andrews ID, Pruniski B, Grebe TA, Corsten-Janssen N, Bouman K, Poulton C, Prakash S, Keren B, Brown NJ, Hunter MF, Heath O, Lakhani SA, McDermott JH, Ascher DB, Chapman G, Bozon K, Dunwoodie SL. A metabolic signature for NADSYN1-dependent congenital NAD deficiency disorder. J Clin Invest 2024; 134:e174824. [PMID: 38357931 PMCID: PMC10866660 DOI: 10.1172/jci174824] [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: 09/06/2023] [Accepted: 12/20/2023] [Indexed: 02/16/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is essential for embryonic development. To date, biallelic loss-of-function variants in 3 genes encoding nonredundant enzymes of the NAD de novo synthesis pathway - KYNU, HAAO, and NADSYN1 - have been identified in humans with congenital malformations defined as congenital NAD deficiency disorder (CNDD). Here, we identified 13 further individuals with biallelic NADSYN1 variants predicted to be damaging, and phenotypes ranging from multiple severe malformations to the complete absence of malformation. Enzymatic assessment of variant deleteriousness in vitro revealed protein domain-specific perturbation, complemented by protein structure modeling in silico. We reproduced NADSYN1-dependent CNDD in mice and assessed various maternal NAD precursor supplementation strategies to prevent adverse pregnancy outcomes. While for Nadsyn1+/- mothers, any B3 vitamer was suitable to raise NAD, preventing embryo loss and malformation, Nadsyn1-/- mothers required supplementation with amidated NAD precursors (nicotinamide or nicotinamide mononucleotide) bypassing their metabolic block. The circulatory NAD metabolome in mice and humans before and after NAD precursor supplementation revealed a consistent metabolic signature with utility for patient identification. Our data collectively improve clinical diagnostics of NADSYN1-dependent CNDD, provide guidance for the therapeutic prevention of CNDD, and suggest an ongoing need to maintain NAD levels via amidated NAD precursor supplementation after birth.
Collapse
Affiliation(s)
- Justin O. Szot
- Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
| | - Hartmut Cuny
- Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, Sydney, New South Wales, Australia
| | - Ella M.M.A. Martin
- Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
| | - Delicia Z. Sheng
- Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
| | - Kavitha Iyer
- Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
| | - Stephanie Portelli
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Vivien Nguyen
- Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
| | - Jessica M. Gereis
- Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
| | - Dimuthu Alankarage
- Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
| | - David Chitayat
- Department of Pediatrics, Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, and
- Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Karen Chong
- Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Alban Lermine
- Laboratoire de Biologie Médicale Multisites SeqOIA, FMG2025, Paris, France
| | - Emma Burkitt-Wright
- Manchester Centre for Genomic Medicine, St. Mary’s Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Weizhen Ji
- Yale University School of Medicine, Pediatric Genomics Discovery Program, New Haven, Connecticut, USA
| | - Lauren Jeffries
- Yale University School of Medicine, Pediatric Genomics Discovery Program, New Haven, Connecticut, USA
| | - Lynn S. Pais
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Tiong Y. Tan
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - James Pitt
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Metabolic Laboratory, Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
| | - Cheryl A. Wise
- Department of Diagnostic Genomics, PathWest Laboratory Medicine Western Australia, Nedlands, Perth, Western Australia, Australia
| | - Helen Wright
- General Paediatric Department, Perth Children’s Hospital, Perth, Western Australia, Australia
- Rural Clinical School, University of Western Australia, Perth, Western Australia, Australia
| | | | - Brianna Pruniski
- Division of Genetics and Metabolism, Phoenix Children’s Hospital, Phoenix, Arizona, USA
| | - Theresa A. Grebe
- Division of Genetics and Metabolism, Phoenix Children’s Hospital, Phoenix, Arizona, USA
| | - Nicole Corsten-Janssen
- Department of Genetics, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Katelijne Bouman
- Department of Genetics, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Cathryn Poulton
- Genetic Services of Western Australia, King Edward Memorial Hospital, Perth, Western Australia, Australia
| | - Supraja Prakash
- Division of Genetics and Metabolism, Phoenix Children’s Hospital, Phoenix, Arizona, USA
| | - Boris Keren
- Département de Génétique, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique – Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Natasha J. Brown
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Matthew F. Hunter
- Monash Genetics, Monash Health, Clayton, Victoria, Australia
- Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Oliver Heath
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
- Department of Metabolic Medicine, The Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Saquib A. Lakhani
- Yale University School of Medicine, Pediatric Genomics Discovery Program, New Haven, Connecticut, USA
| | - John H. McDermott
- Manchester Centre for Genomic Medicine, St. Mary’s Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - David B. Ascher
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Gavin Chapman
- Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, Sydney, New South Wales, Australia
| | - Kayleigh Bozon
- Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
| | - Sally L. Dunwoodie
- Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
46
|
Li Y, Zhang C, Li Z, Bai F, Jing Y, Ke H, Zhang S, Yan Y, Yu Y. Nicotinamide Riboside Regulates Chemotaxis to Decrease Inflammation and Ameliorate Functional Recovery Following Spinal Cord Injury in Mice. Curr Issues Mol Biol 2024; 46:1291-1307. [PMID: 38392200 PMCID: PMC10887503 DOI: 10.3390/cimb46020082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
Changes in intracellular nicotinamide adenine dinucleotide (NAD+) levels have been observed in various disease states. A decrease in NAD+ levels has been noted following spinal cord injury (SCI). Nicotinamide riboside (NR) serves as the precursor of NAD+. Previous research has demonstrated the anti-inflammatory and apoptosis-reducing effects of NR supplements. However, it remains unclear whether NR exerts a similar role in mice after SCI. The objective of this study was to investigate the impact of NR on these changes in a mouse model of SCI. Four groups were considered: (1) non-SCI without NR (Sham), (2) non-SCI with NR (Sham +NR), (3) SCI without NR (SCI), and (4) SCI with NR (SCI + NR). Female C57BL/6J mice aged 6-8 weeks were intraperitoneally administered with 500 mg/kg/day NR for a duration of one week. The supplementation of NR resulted in a significant elevation of NAD+ levels in the spinal cord tissue of mice after SCI. In comparison to the SCI group, NR supplementation exhibited regulatory effects on the chemotaxis/recruitment of leukocytes, leading to reduced levels of inflammatory factors such as IL-1β, TNF-α, and IL-22 in the injured area. Moreover, NR supplementation notably enhanced the survival of neurons and synapses within the injured area, ultimately resulting in improved motor functions after SCI. Therefore, our research findings demonstrated that NR supplementation had inhibitory effects on leukocyte chemotaxis, anti-inflammatory effects, and could significantly improve the immune micro-environment after SCI, thereby promoting neuronal survival and ultimately enhancing the recovery of motor functions after SCI. NR supplementation showed promise as a potential clinical treatment strategy for SCI.
Collapse
Affiliation(s)
- Yan Li
- Institute of Rehabilitation Medicine, China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100068, China
| | - Chunjia Zhang
- School of Rehabilitation, Capital Medical University, Beijing 100068, China
| | - Zihan Li
- Institute of Rehabilitation Medicine, China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100068, China
| | - Fan Bai
- Institute of Rehabilitation Medicine, China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100068, China
| | - Yingli Jing
- Institute of Rehabilitation Medicine, China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100068, China
| | - Han Ke
- School of Rehabilitation, Capital Medical University, Beijing 100068, China
| | - Shuangyue Zhang
- Institute of Rehabilitation Medicine, China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100068, China
| | - Yitong Yan
- Institute of Rehabilitation Medicine, China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100068, China
| | - Yan Yu
- Institute of Rehabilitation Medicine, China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100068, China
- School of Rehabilitation, Capital Medical University, Beijing 100068, China
| |
Collapse
|
47
|
Li F, Wu C, Wang G. Targeting NAD Metabolism for the Therapy of Age-Related Neurodegenerative Diseases. Neurosci Bull 2024; 40:218-240. [PMID: 37253984 PMCID: PMC10838897 DOI: 10.1007/s12264-023-01072-3] [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: 01/25/2023] [Accepted: 04/10/2023] [Indexed: 06/01/2023] Open
Abstract
As the aging population continues to grow rapidly, age-related diseases are becoming an increasing burden on the healthcare system and a major concern for the well-being of elderly individuals. While aging is an inevitable process for all humans, it can be slowed down and age-related diseases can be treated or alleviated. Nicotinamide adenine dinucleotide (NAD) is a critical coenzyme or cofactor that plays a central role in metabolism and is involved in various cellular processes including the maintenance of metabolic homeostasis, post-translational protein modifications, DNA repair, and immune responses. As individuals age, their NAD levels decline, and this decrease has been suggested to be a contributing factor to the development of numerous age-related diseases, such as cancer, diabetes, cardiovascular diseases, and neurodegenerative diseases. In pursuit of healthy aging, researchers have investigated approaches to boost or maintain NAD levels. Here, we provide an overview of NAD metabolism and the role of NAD in age-related diseases and summarize recent progress in the development of strategies that target NAD metabolism for the treatment of age-related diseases, particularly neurodegenerative diseases.
Collapse
Affiliation(s)
- Feifei Li
- School of Pharmaceutical Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Chou Wu
- School of Pharmaceutical Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Gelin Wang
- School of Pharmaceutical Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
48
|
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
|
49
|
Chini CCS, Cordeiro HS, Tran NLK, Chini EN. NAD metabolism: Role in senescence regulation and aging. Aging Cell 2024; 23:e13920. [PMID: 37424179 PMCID: PMC10776128 DOI: 10.1111/acel.13920] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/11/2023] Open
Abstract
The geroscience hypothesis proposes that addressing the biology of aging could directly prevent the onset or mitigate the severity of multiple chronic diseases. Understanding the interplay between key aspects of the biological hallmarks of aging is essential in delivering the promises of the geroscience hypothesis. Notably, the nucleotide nicotinamide adenine dinucleotide (NAD) interfaces with several biological hallmarks of aging, including cellular senescence, and changes in NAD metabolism have been shown to be involved in the aging process. The relationship between NAD metabolism and cellular senescence appears to be complex. On the one hand, the accumulation of DNA damage and mitochondrial dysfunction induced by low NAD+ can promote the development of senescence. On the other hand, the low NAD+ state that occurs during aging may inhibit SASP development as this secretory phenotype and the development of cellular senescence are both highly metabolically demanding. However, to date, the impact of NAD+ metabolism on the progression of the cellular senescence phenotype has not been fully characterized. Therefore, to explore the implications of NAD metabolism and NAD replacement therapies, it is essential to consider their interactions with other hallmarks of aging, including cellular senescence. We propose that a comprehensive understanding of the interplay between NAD boosting strategies and senolytic agents is necessary to advance the field.
Collapse
Affiliation(s)
- Claudia Christiano Silva Chini
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Heidi Soares Cordeiro
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| | - Ngan Le Kim Tran
- Center for Clinical and Translational Science and Mayo Clinic Graduate School of Biomedical SciencesMayo ClinicJacksonvilleFloridaUSA
| | - Eduardo Nunes Chini
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineRochesterMinnesotaUSA
- Metabolism and Molecular Nutrition Laboratory, Kogod Center on Aging, Department of Anesthesiology and Perioperative MedicineMayo Clinic College of MedicineJacksonvilleFloridaUSA
| |
Collapse
|
50
|
Alghamdi M, Braidy N. Supplementation with NAD+ Precursors for Treating Alzheimer's Disease: A Metabolic Approach. J Alzheimers Dis 2024; 101:S467-S477. [PMID: 39422945 DOI: 10.3233/jad-231277] [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] [Indexed: 10/19/2024]
Abstract
Background Alzheimer's disease (AD) is a progressive neurocognitive disorder. There is no cure for AD. Maintenance on intracellular levels of nicotinamide adenine dinucleotide (NAD+) has been reported to be a promising therapeutic strategy for the treatment of AD. NAD+ precursors that represent candidate targets include nicotinamide mononucleotide (NMN) and nicotinamide riboside (NR). Objective This systematic review provides insights into the potential therapeutic value of NAD+ precursors including NMN and NR, for the treatment of AD using preclinical and clinical studies published in the last 5 years. Methods The Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) protocol was followed to systematically search the literature using two databases. Results We found 3 studies that used NMN to treat AD in preclinical murine models. However, human clinical trials using NMN as a therapeutic intervention in AD was not available in the current literature. We also found 4 studies that investigated the potential benefits of NR for the treatment of AD in preclinical models. We also found 2 human clinical trials that showed marked improvements in plasma and neuroimaging biomarkers, and cognitive measures following supplementation with NR. Conclusions Results of preclinical and clinical studies confirm the potential benefits of NAD+ precursors for the treatment of AD. However, further clinical studies are required to confirm the increasingly important value of NAD+ precursors as effective pharmacological interventions in the clinic.
Collapse
Affiliation(s)
- Mohammed Alghamdi
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
- Department of Radiology and Medical Imaging, Faculty of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| |
Collapse
|