1
|
Norheim KL, Ben Ezra M, Heckenbach I, Andreasson LM, Eriksen LL, Dyhre-Petersen N, Damgaard MV, Berglind M, Pricolo L, Sampson D, Dellinger RW, Sverrild A, Treebak JT, Ditlev SB, Porsbjerg C, Scheibye-Knudsen M. Effect of nicotinamide riboside on airway inflammation in COPD: a randomized, placebo-controlled trial. NATURE AGING 2024; 4:1772-1781. [PMID: 39548320 PMCID: PMC11645284 DOI: 10.1038/s43587-024-00758-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive, incurable disease associated with smoking and advanced age, ranking as the third leading cause of death worldwide. DNA damage and loss of the central metabolite nicotinamide adenine dinucleotide (NAD+) may contribute to both aging and COPD, presenting a potential avenue for interventions. In this randomized, double-blind, placebo-controlled clinical trial, we treated patients with stable COPD (n = 40) with the NAD+ precursor nicotinamide riboside (NR) for 6 weeks and followed-up 12 weeks later. The primary outcome was change in sputum interleukin-8 (IL-8) from baseline to week 6. The estimated treatment difference between NR and placebo in IL-8 after 6 weeks was -52.6% (95% confidence interval (CI): -75.7% to -7.6%; P = 0.030). This effect persisted until the follow-up 12 weeks after the end of treatment (-63.7%: 95% CI -85.7% to -7.8%; P = 0.034). For secondary outcomes, NR treatment increased NAD+ levels by more than twofold in whole blood, whereas IL-6 levels in plasma remained unchanged. In exploratory analyses, treatment with NR showed indications of upregulated gene pathways related to genomic integrity in the airways and reduced epigenetic aging, possibly through a reduction in cellular senescence. These exploratory analyses need to be confirmed in future trials. ClinicalTrials.gov identifier: NCT04990869 .
Collapse
Affiliation(s)
- Kristoffer L Norheim
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Michael Ben Ezra
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | | | - Louise Munkholm Andreasson
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Lise Lotte Eriksen
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Nanna Dyhre-Petersen
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Mads Vargas Damgaard
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Magnus Berglind
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Luca Pricolo
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Asger Sverrild
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sisse Bolm Ditlev
- Copenhagen Center for Translational Research, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Celeste Porsbjerg
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Morten Scheibye-Knudsen
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Kim JT, Jeon DH, Lee HJ. Molecular mechanism of skeletal muscle loss and its prevention by natural resources. Food Sci Biotechnol 2024; 33:3387-3400. [PMID: 39493391 PMCID: PMC11525361 DOI: 10.1007/s10068-024-01678-x] [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: 05/15/2024] [Revised: 07/23/2024] [Accepted: 08/04/2024] [Indexed: 11/05/2024] Open
Abstract
A skeletal muscle disorder has drawn attention due to the global aging issues. The loss of skeletal muscle mass has been suggested to be from the reduced muscle regeneration by dysfunction of muscle satellite cell/fibro-adipogenic progenitor cells and the muscle atrophy by dysfunction of mitochondria, ubiquitin-proteasome system, and autophagy. In this review, we highlighted the underlying mechanisms of skeletal muscle mass loss including Notch signaling, Wnt/β-catenin signaling, Hedgehog signaling, AMP-activated protein kinase (AMPK) signaling, and mammalian target of rapamycin (mTOR) signaling. In addition, we summarized accumulated studies of natural resources investigating their roles in ameliorating the loss of skeletal muscle mass and demonstrating the underlying mechanisms in vitro and in vivo. In conclusion, following the studies of natural resources exerting the preventive activity in muscle mass loss, the signaling-based approaches may accelerate the development of functional foods for sarcopenia prevention.
Collapse
Affiliation(s)
- Jin Tae Kim
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 17546 South Korea
- GreenTech-Based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, 17546 South Korea
| | - Dong Hyeon Jeon
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 17546 South Korea
- GreenTech-Based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, 17546 South Korea
| | - Hong Jin Lee
- Department of Food Science and Biotechnology, Chung-Ang University, Anseong, 17546 South Korea
- GreenTech-Based Food Safety Research Group, BK21 Four, Chung-Ang University, Anseong, 17546 South Korea
| |
Collapse
|
3
|
Madsen HB, Navarro C, Gasparini E, Park JH, Li Z, Croteau DL, Bohr VA. Urolithin A and nicotinamide riboside differentially regulate innate immune defenses and metabolism in human microglial cells. Front Aging Neurosci 2024; 16:1503336. [PMID: 39665042 PMCID: PMC11631940 DOI: 10.3389/fnagi.2024.1503336] [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: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction During aging, many cellular processes, such as autophagic clearance, DNA repair, mitochondrial health, metabolism, nicotinamide adenine dinucleotide (NAD+) levels, and immunological responses, become compromised. Urolithin A (UA) and Nicotinamide Riboside (NR) are two naturally occurring compounds known for their anti-inflammatory and mitochondrial protective properties, yet the effects of these natural substances on microglia cells have not been thoroughly investigated. As both UA and NR are considered safe dietary supplements, it is equally important to understand their function in normal cells and in disease states. Methods This study investigates the effects of UA and NR on immune signaling, mitochondrial function, and microglial activity in a human microglial cell line (HMC3). Results Both UA and NR were shown to reduce DNA damage-induced cellular senescence. However, they differentially regulated gene expression related to neuroinflammation, with UA enhancing cGAS-STING pathway activation and NR displaying broader anti-inflammatory effects. Furthermore, UA and NR differently influenced mitochondrial dynamics, with both compounds improving mitochondrial respiration but exhibiting distinct effects on production of reactive oxygen species and glycolytic function. Discussion These findings underscore the potential of UA and NR as therapeutic agents in managing neuroinflammation and mitochondrial dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Helena Borland Madsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Claudia Navarro
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Emilie Gasparini
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jae-Hyeon Park
- Section on DNA Repair, National Institute on Aging, Baltimore, MD, United States
| | - Zhiquan Li
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Deborah L. Croteau
- Section on DNA Repair, National Institute on Aging, Baltimore, MD, United States
- Computational Biology and Genomics Core, Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, MD, United States
| | - Vilhelm A. Bohr
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Section on DNA Repair, National Institute on Aging, Baltimore, MD, United States
| |
Collapse
|
4
|
Martinović A, Mantovani M, Trpchevska N, Novak E, Milev NB, Bode L, Ewald CY, Bischof E, Reichmuth T, Lapides R, Navarini A, Saravi B, Roider E. Climbing the longevity pyramid: overview of evidence-driven healthcare prevention strategies for human longevity. FRONTIERS IN AGING 2024; 5:1495029. [PMID: 39659760 PMCID: PMC11628525 DOI: 10.3389/fragi.2024.1495029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024]
Abstract
Longevity medicine is an emerging and iterative healthcare discipline focusing on early detection, preventive measures, and personalized approaches that aim to extend healthy lifespan and promote healthy aging. This comprehensive review introduces the innovative concept of the "Longevity Pyramid." This conceptual framework delineates progressive intervention levels, providing a structured approach to understanding the diverse strategies available in longevity medicine. At the base of the Longevity Pyramid lies the level of prevention, emphasizing early detection strategies and advanced diagnostics or timely identification of potential health issues. Moving upwards, the next step involves lifestyle modifications, health-promoting behaviors, and proactive measures to delay the onset of age-related conditions. The Longevity Pyramid further explores the vast range of personalized interventions, highlighting the importance of tailoring medical approaches based on genetic predispositions, lifestyle factors, and unique health profiles, thereby optimizing interventions for maximal efficacy. These interventions aim to extend lifespan and reduce the impact and severity of age-related conditions, ensuring that additional years are characterized by vitality and wellbeing. By outlining these progressive levels of intervention, this review offers valuable insights into the evolving field of longevity medicine. This structured framework guides researchers and practitioners toward a nuanced strategic approach to advancing the science and practice of healthy aging.
Collapse
Affiliation(s)
- Anđela Martinović
- Maximon AG, Zug, Switzerland
- Department of Food Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | | | | | | | | | | | - Collin Y. Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Evelyne Bischof
- Shanghai University of Medicine and Health Sciences, Shanghai, China
- Sheba Longevity Center, Sheba Medical Center Tel Aviv, Ramat Gan, Israel
| | | | - Rebecca Lapides
- The Robert Larner, M.D., College of Medicine at the University of Vermont, Burlington, VT, United States
| | - Alexander Navarini
- Department of Dermatology, University Hospital Basel, Basel, Switzerland
| | - Babak Saravi
- Department of Orthopedics and Trauma Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Elisabeth Roider
- Maximon AG, Zug, Switzerland
- Department of Dermatology, University Hospital of Basel, Basel, Switzerland
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
5
|
Gherardi G, Weiser A, Bermont F, Migliavacca E, Brinon B, Jacot GE, Hermant A, Sturlese M, Nogara L, Vascon F, De Mario A, Mattarei A, Garratt E, Burton M, Lillycrop K, Godfrey KM, Cendron L, Barron D, Moro S, Blaauw B, Rizzuto R, Feige JN, Mammucari C, De Marchi U. Mitochondrial calcium uptake declines during aging and is directly activated by oleuropein to boost energy metabolism and skeletal muscle performance. Cell Metab 2024:S1550-4131(24)00417-0. [PMID: 39603237 DOI: 10.1016/j.cmet.2024.10.021] [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: 04/07/2023] [Revised: 07/24/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024]
Abstract
Mitochondrial calcium (mtCa2+) uptake via the mitochondrial calcium uniporter (MCU) couples calcium homeostasis and energy metabolism. mtCa2+ uptake via MCU is rate-limiting for mitochondrial activation during muscle contraction, but its pathophysiological role and therapeutic application remain largely uncharacterized. By profiling human muscle biopsies, patient-derived myotubes, and preclinical models, we discovered a conserved downregulation of mitochondrial calcium uniporter regulator 1 (MCUR1) during skeletal muscle aging that associates with human sarcopenia and impairs mtCa2+ uptake and mitochondrial respiration. Through a screen of 5,000 bioactive molecules, we identify the natural polyphenol oleuropein as a specific MCU activator that stimulates mitochondrial respiration via mitochondrial calcium uptake 1 (MICU1) binding. Oleuropein activates mtCa2+ uptake and energy metabolism to enhance endurance and reduce fatigue in young and aged mice but not in muscle-specific MCU knockout (KO) mice. Our work demonstrates that impaired mtCa2+ uptake contributes to mitochondrial dysfunction during aging and establishes oleuropein as a novel food-derived molecule that specifically targets MCU to stimulate mitochondrial bioenergetics and muscle performance.
Collapse
Affiliation(s)
- Gaia Gherardi
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Anna Weiser
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produit Nestlé S.A., EPFL Innovation Park, 1015 Lausanne, Switzerland; Molecular Nutritional Medicine, Else Kröner Fresenius Center for Nutritional Medicine, Technische Universität München, 85354 Freising, Germany
| | - Flavien Bermont
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produit Nestlé S.A., EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Eugenia Migliavacca
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produit Nestlé S.A., EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Benjamin Brinon
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produit Nestlé S.A., EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Guillaume E Jacot
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produit Nestlé S.A., EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Aurélie Hermant
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produit Nestlé S.A., EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Mattia Sturlese
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, 35131 Padova, Italy
| | - Leonardo Nogara
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, 35131 Padova, Italy
| | - Filippo Vascon
- Department of Biology, University of Padova, 35131 Padova, Italy
| | - Agnese De Mario
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Andrea Mattarei
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, 35131 Padova, Italy
| | - Emma Garratt
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Mark Burton
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Karen Lillycrop
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK; Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Keith M Godfrey
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK; NIHR Southampton Biomedical Research Centre, University of Southampton & University Hospital Southampton NHS Foundation Trust, Southampton, UK; Medical Research Council Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Laura Cendron
- Department of Biology, University of Padova, 35131 Padova, Italy
| | - Denis Barron
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produit Nestlé S.A., EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Stefano Moro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, 35131 Padova, Italy
| | - Bert Blaauw
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, 35129 Padova, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; Myology Center (CIR-Myo), University of Padova, 35131 Padova, Italy.
| | - Jerome N Feige
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produit Nestlé S.A., EPFL Innovation Park, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Cristina Mammucari
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; Myology Center (CIR-Myo), University of Padova, 35131 Padova, Italy.
| | - Umberto De Marchi
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produit Nestlé S.A., EPFL Innovation Park, 1015 Lausanne, Switzerland.
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Xiao Y, Pang N, Ma S, Gao M, Yang L. Effect of Nicotinamide Riboside Against the Exhaustion of CD8 + T Cells via Alleviating Mitochondrial Dysfunction. Nutrients 2024; 16:3577. [PMID: 39519411 PMCID: PMC11547570 DOI: 10.3390/nu16213577] [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: 08/31/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Targeting mitochondria and protecting the mitochondrial function of CD8+ T cells are crucial for enhancing the clinical efficacy of cancer immunotherapy. Objectives: In this study, our objective was to investigate the potential of nicotinamide riboside (NR) in preserving the mitochondrial function of CD8+ T cells and mitigating their exhaustion. Methods: We established two in vitro models to induce CD8+ T cell exhaustion either by tumor cell-conditioned medium (TCM) or by continuous stimulation with OVA(257-264) peptide. CD8+ T cells were treated in the absence/presence of NR. Results: Our findings demonstrated that NR supplementation effectively inhibited CD8+ T cell exhaustion and preserved mitochondrial function in both models. Moreover, apoptosis of CD8+ T cells was reduced after NR treatment. Western blot data indicated that NR treatment upregulated Silent information regulator 1 (SirT1) expression. Further inhibition of Sirt1 activity using EX527 uncovered that the inhibitory effect of NR on CD8+ T cell exhaustion and its protective effect on mitochondria were attenuated. Conclusions: In conclusion, our results indicate that NR supplementation attenuates CD8+ T cell exhaustion, and its underlying mechanism is associated with increased mitochondrial function regulated by the SirT1 pathway. Our research provides evidence that NR may assist in enhancing the clinical efficacy of immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Lili Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
9
|
Aaltio J, Euro L, Tynninen O, Vu HS, Ni M, DeBerardinis RJ, Suomalainen A, Isohanni P. Niacin supplementation in a child with novel MTTN variant m.5670A>G causing early onset mitochondrial myopathy and NAD + deficiency. Neuromuscul Disord 2024; 43:14-19. [PMID: 39173541 DOI: 10.1016/j.nmd.2024.07.005] [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: 03/14/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 08/24/2024]
Abstract
Myopathy is a common manifestation in mitochondrial disorders, but the pathomechanisms are still insufficiently studied in children. Here, we report a severe, progressive mitochondrial myopathy in a four-year-old child, who died at eight years. He developed progressive loss of muscle strength with nocturnal hypoventilation and dilated cardiomyopathy. Skeletal muscle showed ragged red fibers and severe combined respiratory chain deficiency. Mitochondrial DNA sequencing revealed a novel m.5670A>G mutation in mitochondrial tRNAAsn (MTTN) with 88 % heteroplasmy in muscle. The proband also had systemic NAD+ deficiency but rescuing this with the NAD+ precursor niacin did not stop disease progression. Targeted metabolomics revealed an overall shift of metabolism towards controls after niacin supplementation, with normalized tryptophan metabolites and lipid-metabolic markers, but most amino acids did not respond to niacin therapy. To conclude, we report a new MTTN mutation, secondary NAD+ deficiency in childhood-onset mitochondrial myopathy with metabolic but meager clinical response to niacin supplementation.
Collapse
Affiliation(s)
- Juho Aaltio
- Research Programs Unit, Stem Cells and Metabolism Research, University of Helsinki, Helsinki, Finland.
| | - Liliya Euro
- Research Programs Unit, Stem Cells and Metabolism Research, University of Helsinki, Helsinki, Finland
| | - Olli Tynninen
- Department of Pathology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Hieu S Vu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Min Ni
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Anu Suomalainen
- Research Programs Unit, Stem Cells and Metabolism Research, University of Helsinki, Helsinki, Finland; HUS Diagnostic Centre, Helsinki University Hospital, Helsinki, Finland; HiLife, University of Helsinki, Helsinki, Finland
| | - Pirjo Isohanni
- Research Programs Unit, Stem Cells and Metabolism Research, University of Helsinki, Helsinki, Finland; Child Neurology, Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
10
|
Madreiter-Sokolowski CT, Hiden U, Krstic J, Panzitt K, Wagner M, Enzinger C, Khalil M, Abdellatif M, Malle E, Madl T, Osto E, Schosserer M, Binder CJ, Olschewski A. Targeting organ-specific mitochondrial dysfunction to improve biological aging. Pharmacol Ther 2024; 262:108710. [PMID: 39179117 DOI: 10.1016/j.pharmthera.2024.108710] [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: 04/27/2024] [Revised: 08/09/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
In an aging society, unveiling new anti-aging strategies to prevent and combat aging-related diseases is of utmost importance. Mitochondria are the primary ATP production sites and key regulators of programmed cell death. Consequently, these highly dynamic organelles play a central role in maintaining tissue function, and mitochondrial dysfunction is a pivotal factor in the progressive age-related decline in cellular homeostasis and organ function. The current review examines recent advances in understanding the interplay between mitochondrial dysfunction and organ-specific aging. Thereby, we dissect molecular mechanisms underlying mitochondrial impairment associated with the deterioration of organ function, exploring the role of mitochondrial DNA, reactive oxygen species homeostasis, metabolic activity, damage-associated molecular patterns, biogenesis, turnover, and dynamics. We also highlight emerging therapeutic strategies in preclinical and clinical tests that are supposed to rejuvenate mitochondrial function, such as antioxidants, mitochondrial biogenesis stimulators, and modulators of mitochondrial turnover and dynamics. Furthermore, we discuss potential benefits and challenges associated with the use of these interventions, emphasizing the need for organ-specific approaches given the unique mitochondrial characteristics of different tissues. In conclusion, this review highlights the therapeutic potential of addressing mitochondrial dysfunction to mitigate organ-specific aging, focusing on the skin, liver, lung, brain, skeletal muscle, and lung, as well as on the reproductive, immune, and cardiovascular systems. Based on a comprehensive understanding of the multifaceted roles of mitochondria, innovative therapeutic strategies may be developed and optimized to combat biological aging and promote healthy aging across diverse organ systems.
Collapse
Affiliation(s)
| | - Ursula Hiden
- Department of Obstetrics and Gynecology, Research Unit of Early Life Determinants, Medical University of Graz, Austria
| | - Jelena Krstic
- Division of Cell Biology, Histology and Embryology, Medical University of Graz, BioTechMed-Graz, Austria
| | - Katrin Panzitt
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Austria
| | - Martin Wagner
- Division of Gastroenterology and Hepatology, Medical University of Graz, Austria
| | | | - Michael Khalil
- Department of Neurology, Medical University of Graz, Austria
| | - Mahmoud Abdellatif
- Division of Cardiology, Medical University of Graz, BioTechMed-Graz, Austria
| | - Ernst Malle
- Division of Molecular Biology and Biochemistry, Medical University of Graz, BioTechMed-Graz, Austria
| | - Tobias Madl
- Division of Medicinal Chemistry, Medical University of Graz, BioTechMed-Graz, Austria
| | - Elena Osto
- Division of Physiology and Pathophysiology, Medical University of Graz
| | - Markus Schosserer
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Austria; Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Andrea Olschewski
- Department of Anesthesiology and Intensive Care Medicine, LBI for Lung Vascular Research, Medical University of Graz, Austria.
| |
Collapse
|
11
|
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
|
12
|
Han H, Jia H, Wang YF, Song JP. Cardiovascular adaptations and pathological changes induced by spaceflight: from cellular mechanisms to organ-level impacts. Mil Med Res 2024; 11:68. [PMID: 39334239 PMCID: PMC11429428 DOI: 10.1186/s40779-024-00570-3] [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: 03/04/2024] [Accepted: 09/01/2024] [Indexed: 09/30/2024] Open
Abstract
The advancement in extraterrestrial exploration has highlighted the crucial need for studying how the human cardiovascular system adapts to space conditions. Human development occurs under the influence of gravity, shielded from space radiation by Earth's magnetic field, and within an environment characterized by 24-hour day-night cycles resulting from Earth's rotation, thus deviating from these conditions necessitates adaptive responses for survival. With upcoming manned lunar and Martian missions approaching rapidly, it is essential to understand the impact of various stressors induced by outer-space environments on cardiovascular health. This comprehensive review integrates insights from both actual space missions and simulated experiments on Earth, to analyze how microgravity, space radiation, and disrupted circadian affect cardiovascular well-being. Prolonged exposure to microgravity induces myocardial atrophy and endothelial dysfunction, which may be exacerbated by space radiation. Mitochondrial dysfunction and oxidative stress emerge as key underlying mechanisms along with disturbances in ion channel perturbations, cytoskeletal damage, and myofibril changes. Disruptions in circadian rhythms caused by factors such as microgravity, light exposure, and irregular work schedules, could further exacerbate cardiovascular issues. However, current research tends to predominantly focus on disruptions in the core clock gene, overlooking the multifactorial nature of circadian rhythm disturbances in space. Future space missions should prioritize targeted prevention strategies and early detection methods for identifying cardiovascular risks, to preserve astronaut health and ensure mission success.
Collapse
Affiliation(s)
- Han Han
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yi-Fan Wang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Jiang-Ping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease; Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
13
|
Su M, Qiu F, Li Y, Che T, Li N, Zhang S. Mechanisms of the NAD + salvage pathway in enhancing skeletal muscle function. Front Cell Dev Biol 2024; 12:1464815. [PMID: 39372950 PMCID: PMC11450036 DOI: 10.3389/fcell.2024.1464815] [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: 07/15/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is crucial for cellular energy production, serving as a coenzyme in oxidation-reduction reactions. It also supports enzymes involved in processes such as DNA repair, aging, and immune responses. Lower NAD+ levels have been associated with various diseases, highlighting the importance of replenishing NAD+. Nicotinamide phosphoribosyltransferase (NAMPT) plays a critical role in the NAD+ salvage pathway, which helps sustain NAD+ levels, particularly in high-energy tissues like skeletal muscle.This review explores how the NAMPT-driven NAD+ salvage pathway influences skeletal muscle health and functionality in aging, type 2 diabetes mellitus (T2DM), and skeletal muscle injury. The review offers insights into enhancing the salvage pathway through exercise and NAD+ boosters as strategies to improve muscle performance. The findings suggest significant potential for using this pathway in the diagnosis, monitoring, and treatment of skeletal muscle conditions.
Collapse
Affiliation(s)
- Mengzhu Su
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, China
- School of Physical Education, Qingdao University, Qingdao, China
| | - Fanghui Qiu
- School of Physical Education, Qingdao University, Qingdao, China
| | - Yansong Li
- School of Physical Education, Qingdao University, Qingdao, China
| | - Tongtong Che
- School of Physical Education, Qingdao University, Qingdao, China
| | - Ningning Li
- School of Physical Education, Qingdao University, Qingdao, China
| | - Shuangshuang Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, China
- School of Physical Education, Qingdao University, Qingdao, China
| |
Collapse
|
14
|
Viña J, Borrás C. Unlocking the biochemical secrets of longevity: balancing healthspan and lifespan. FEBS Lett 2024; 598:2135-2144. [PMID: 38956807 DOI: 10.1002/1873-3468.14963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 07/04/2024]
Abstract
In an era of rising global life expectancies, research focuses on enhancing the quality of extended years. This review examines the link between mitochondrial function and aging, highlighting the importance of healthspan alongside lifespan. This involves significant human and economic challenges, with longer lifespans often accompanied by reduced well-being. Addressing mitochondrial decline, exploring targeted interventions, and understanding the complexities of research models are vital for advancing our knowledge in this field. Additionally, promoting physical exercise and adopting personalized supplementation strategies based on individual needs can contribute to healthy aging. The insights from this Perspective article offer a hopeful outlook for future advances in extending both lifespan and healthspan, aiming to improve the overall quality of life in aging populations.
Collapse
Affiliation(s)
- Jose Viña
- Freshage Research Group, Department of Physiology, Faculty of Medicine, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), Institute of Health Research-INCLIVA, University of Valencia, Spain
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), Institute of Health Research-INCLIVA, University of Valencia, Spain
| |
Collapse
|
15
|
Broome SC, Whitfield J, Karagounis LG, Hawley JA. Mitochondria as Nutritional Targets to Maintain Muscle Health and Physical Function During Ageing. Sports Med 2024; 54:2291-2309. [PMID: 39060742 PMCID: PMC11393155 DOI: 10.1007/s40279-024-02072-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 07/28/2024]
Abstract
The age-related loss of skeletal muscle mass and physical function leads to a loss of independence and an increased reliance on health-care. Mitochondria are crucial in the aetiology of sarcopenia and have been identified as key targets for interventions that can attenuate declines in physical capacity. Exercise training is a primary intervention that reduces many of the deleterious effects of ageing in skeletal muscle quality and function. However, habitual levels of physical activity decline with age, making it necessary to implement adjunct treatments to maintain skeletal muscle mitochondrial health and physical function. This review provides an overview of the effects of ageing and exercise training on human skeletal muscle mitochondria and considers several supplements that have plausible mechanistic underpinning to improve physical function in ageing through their interactions with mitochondria. Several supplements, including MitoQ, urolithin A, omega-3 polyunsaturated fatty acids (n3-PUFAs), and a combination of glycine and N-acetylcysteine (GlyNAC) can improve physical function in older individuals through a variety of inter-dependent mechanisms including increases in mitochondrial biogenesis and energetics, decreases in mitochondrial reactive oxygen species emission and oxidative damage, and improvements in mitochondrial quality control. While there is evidence that some nicotinamide adenine dinucleotide precursors can improve physical function in older individuals, such an outcome seems unrelated to and independent of changes in skeletal muscle mitochondrial function. Future research should investigate the safety and efficacy of compounds that can improve skeletal muscle health in preclinical models through mechanisms involving mitochondria, such as mitochondrial-derived peptides and mitochondrial uncouplers, with a view to extending the human health-span.
Collapse
Affiliation(s)
- Sophie C Broome
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, 3000, Australia.
| | - Jamie Whitfield
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, 3000, Australia
| | - Leonidas G Karagounis
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, 3000, Australia
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
| | - John A Hawley
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, 3000, Australia
| |
Collapse
|
16
|
Deane CS, Willis CRG, Gallagher IJ, Brook MS, Gharahdaghi N, Wylie LJ, Wilkinson DJ, Smith K, Atherton PJ, Etheridge T. Nicotinic acid improves mitochondrial function and associated transcriptional pathways in older inactive males. TRANSLATIONAL EXERCISE BIOMEDICINE 2024; 1:277-294. [PMID: 39703532 PMCID: PMC11653476 DOI: 10.1515/teb-2024-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/30/2024] [Indexed: 12/21/2024]
Abstract
Objectives To examine the effect of the NAD+ precursor, nicotinic acid (NA), for improving skeletal muscle status in sedentary older people. Methods In a double-blind, randomised, placebo-controlled design, 18 sedentary yet otherwise healthy older (65-75 y) males were assigned to 2-weeks of NA (acipimox; 250 mg × 3 daily, n=8) or placebo (PLA, n=10) supplementation. At baseline, and after week 1 and week 2 of supplementation, a battery of functional, metabolic, and molecular readouts were measured. Results Resting and submaximal respiratory exchange ratio was lower (p<0.05) after 2 weeks in the NA group only, but maximal aerobic and anaerobic function and glucose handling were unchanged (p>0.05). Bayesian statistical modelling identified that leak, maximal coupled and maximal uncoupled mitochondrial respiratory states, increased over the 2-week supplemental period in the NA group (probability for a positive change (pd) 85.2, 90.8 and 95.9 %, respectively) but not in PLA. Citrate synthase and protein content of complex II (SDHB) and V (ATP5A) electron transport chain (ETC) components increased over the 2-week period in the NA group only (pd 95.1, 74.5 and 82.3 %, respectively). Mitochondrial and myofibrillar protein synthetic rates remained unchanged in both groups. NA intake altered the muscle transcriptome by increasing the expression of gene pathways related to cell adhesion/cytoskeleton organisation and inflammation/immunity and decreasing pathway expression of ETC and aerobic respiration processes. NAD+-specific pathways (e.g., de novo NAD+ biosynthetic processes) and genes (e.g., NADSYN1) were uniquely regulated by NA. Conclusions NA might be an effective strategy for improving ageing muscle mitochondrial health.
Collapse
Affiliation(s)
- Colleen S. Deane
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
- Human Development & Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Craig R. G. Willis
- School of Chemistry and Biosciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Iain J. Gallagher
- Centre for Biomedicine & Global Health, Edinburgh Napier University, Edinburgh, UK
| | - Matthew S. Brook
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, UK
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Nima Gharahdaghi
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Lee J. Wylie
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Daniel J. Wilkinson
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Kenneth Smith
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Philip J. Atherton
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research and National Institute for Health Research Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, Derby, UK
| | - Timothy Etheridge
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
17
|
Yoshida T, Myakala K, Jones BA, Wang XX, Shrivastav S, Santo BA, Patel TR, Zhao Y, Tutino VM, Sarder P, Rosenberg AZ, Winkler CA, Levi M, Kopp JB. NAD deficiency contributes to progressive kidney disease in HIV-nephropathy mice. Am J Physiol Renal Physiol 2024; 327:F450-F462. [PMID: 38961841 PMCID: PMC11444509 DOI: 10.1152/ajprenal.00061.2024] [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: 03/13/2024] [Revised: 06/04/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024] Open
Abstract
HIV disease remains prevalent in the United States and is particularly prevalent in sub-Saharan Africa. Recent investigations revealed that mitochondrial dysfunction in kidney contributes to HIV-associated nephropathy (HIVAN) in Tg26 transgenic mice. We hypothesized that nicotinamide adenine dinucleotide (NAD) deficiency contributes to energetic dysfunction and progressive tubular injury. We investigated metabolomic mechanisms of HIVAN tubulopathy. Tg26 and wild-type (WT) mice were treated with the farnesoid X receptor (FXR) agonist INT-747 or nicotinamide riboside (NR) from 6 to 12 wk of age. Multiomic approaches were used to characterize kidney tissue transcriptomes and metabolomes. Treatment with INT-747 or NR ameliorated kidney tubular injury, as shown by serum creatinine, the tubular injury marker urinary neutrophil-associated lipocalin, and tubular morphometry. Integrated analysis of metabolomic and transcriptomic measurements showed that NAD levels and production were globally downregulated in Tg26 mouse kidneys, especially nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in the NAD salvage pathway. Furthermore, NAD-dependent deacetylase sirtuin3 activity and mitochondrial oxidative phosphorylation activity were lower in ex vivo proximal tubules from Tg26 mouse kidneys compared with those of WT mice. Restoration of NAD levels in the kidney improved these abnormalities. These data suggest that NAD deficiency might be a treatable target for HIVAN.NEW & NOTEWORTHY The study describes a novel investigation that identified nicotinamide adenine dinucleotide (NAD) deficiency in a widely used HIV-associated nephropathy (HIVAN) transgenic mouse model. We show that INT-747, a farnesoid X receptor agonist, and nicotinamide riboside (NR), a precursor of nicotinamide, each ameliorated HIVAN tubulopathy. Multiomic analysis of mouse kidneys revealed that NAD deficiency was an upstream metabolomic mechanism contributing to HIVAN tubulopathy.
Collapse
Affiliation(s)
- Teruhiko Yoshida
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Komuraiah Myakala
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States
| | - Bryce A Jones
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States
| | - Xiaoxin X Wang
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States
| | - Shashi Shrivastav
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Briana A Santo
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States
| | - Tatsat R Patel
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States
| | - Yongmei Zhao
- Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States
| | - Vincent M Tutino
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States
| | - Pinaki Sarder
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland, United States
| | - Cheryl A Winkler
- Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia, United States
| | - Jeffrey B Kopp
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
18
|
Mostafavi Abdolmaleky H, Zhou JR. Gut Microbiota Dysbiosis, Oxidative Stress, Inflammation, and Epigenetic Alterations in Metabolic Diseases. Antioxidants (Basel) 2024; 13:985. [PMID: 39199231 PMCID: PMC11351922 DOI: 10.3390/antiox13080985] [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: 07/08/2024] [Revised: 08/05/2024] [Accepted: 08/11/2024] [Indexed: 09/01/2024] Open
Abstract
Gut dysbiosis, resulting from an imbalance in the gut microbiome, can induce excessive production of reactive oxygen species (ROS), leading to inflammation, DNA damage, activation of the immune system, and epigenetic alterations of critical genes involved in the metabolic pathways. Gut dysbiosis-induced inflammation can also disrupt the gut barrier integrity and increase intestinal permeability, which allows gut-derived toxic products to enter the liver and systemic circulation, further triggering oxidative stress, inflammation, and epigenetic alterations associated with metabolic diseases. However, specific gut-derived metabolites, such as short-chain fatty acids (SCFAs), lactate, and vitamins, can modulate oxidative stress and the immune system through epigenetic mechanisms, thereby improving metabolic function. Gut microbiota and diet-induced metabolic diseases, such as obesity, insulin resistance, dyslipidemia, and hypertension, can transfer to the next generation, involving epigenetic mechanisms. In this review, we will introduce the key epigenetic alterations that, along with gut dysbiosis and ROS, are engaged in developing metabolic diseases. Finally, we will discuss potential therapeutic interventions such as dietary modifications, prebiotics, probiotics, postbiotics, and fecal microbiota transplantation, which may reduce oxidative stress and inflammation associated with metabolic syndrome by altering gut microbiota and epigenetic alterations. In summary, this review highlights the crucial role of gut microbiota dysbiosis, oxidative stress, and inflammation in the pathogenesis of metabolic diseases, with a particular focus on epigenetic alterations (including histone modifications, DNA methylomics, and RNA interference) and potential interventions that may prevent or improve metabolic diseases.
Collapse
Affiliation(s)
- Hamid Mostafavi Abdolmaleky
- Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
19
|
Yusri K, Kumar S, Fong S, Gruber J, Sorrentino V. Towards Healthy Longevity: Comprehensive Insights from Molecular Targets and Biomarkers to Biological Clocks. Int J Mol Sci 2024; 25:6793. [PMID: 38928497 PMCID: PMC11203944 DOI: 10.3390/ijms25126793] [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: 05/23/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Aging is a complex and time-dependent decline in physiological function that affects most organisms, leading to increased risk of age-related diseases. Investigating the molecular underpinnings of aging is crucial to identify geroprotectors, precisely quantify biological age, and propose healthy longevity approaches. This review explores pathways that are currently being investigated as intervention targets and aging biomarkers spanning molecular, cellular, and systemic dimensions. Interventions that target these hallmarks may ameliorate the aging process, with some progressing to clinical trials. Biomarkers of these hallmarks are used to estimate biological aging and risk of aging-associated disease. Utilizing aging biomarkers, biological aging clocks can be constructed that predict a state of abnormal aging, age-related diseases, and increased mortality. Biological age estimation can therefore provide the basis for a fine-grained risk stratification by predicting all-cause mortality well ahead of the onset of specific diseases, thus offering a window for intervention. Yet, despite technological advancements, challenges persist due to individual variability and the dynamic nature of these biomarkers. Addressing this requires longitudinal studies for robust biomarker identification. Overall, utilizing the hallmarks of aging to discover new drug targets and develop new biomarkers opens new frontiers in medicine. Prospects involve multi-omics integration, machine learning, and personalized approaches for targeted interventions, promising a healthier aging population.
Collapse
Affiliation(s)
- Khalishah Yusri
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Sanjay Kumar
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Sheng Fong
- Department of Geriatric Medicine, Singapore General Hospital, Singapore 169608, Singapore
- Clinical and Translational Sciences PhD Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jan Gruber
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Science Division, Yale-NUS College, Singapore 138527, Singapore
| | - Vincenzo Sorrentino
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism and Amsterdam Neuroscience Cellular & Molecular Mechanisms, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| |
Collapse
|
20
|
Yli-Öyrä J, Juvonen RO, Lehtonen M, Herrala M, Finel M, Räisänen R, Rysä J. Anthraquinone biocolourant dermocybin is metabolized whereas dermorubin is not in in vitro liver fractions and recombinant metabolic enzymes. Basic Clin Pharmacol Toxicol 2024; 134:846-857. [PMID: 38664998 DOI: 10.1111/bcpt.14013] [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: 10/25/2023] [Revised: 02/29/2024] [Accepted: 04/08/2024] [Indexed: 05/18/2024]
Abstract
Fungal anthraquinones dermocybin and dermorubin are attractive alternatives for synthetic dyes but their metabolism is largely unknown. We conducted a qualitative in vitro study to identify their metabolism using human liver microsomes and cytosol, as well as recombinant human cytochrome P450 (CYP), UDP-glucuronosyltransferase (UGT) and sulfotransferase (SULT) enzymes. Additionally, liver microsomal and cytosolic fractions from rat, mouse and pig were used. Following incubations of the biocolourants with the enzymes in the presence of nicotinamide adenine dinucleotide phosphate, UDP-glucuronic acid, 3'-phosphoadenosine-5'-phosphosulfate (PAPS) or S-adenosyl methionine (SAM) to enable CYP oxidation, glucuronidation, sulfonation or methylation, we observed several oxidation and conjugation metabolites for dermocybin but none for dermorubin. Human CYP1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, 3A4 and 3A7 catalysed dermocybin oxidation. The formation of dermocybin glucuronides was catalysed by human UGT1A1, 1A3, 1A7, 1A8, 1A9, 1A10 and 2B15. Human SULT1B1, 1C2 and 2A1 sulfonated dermocybin. Dermocybin oxidation was faster than conjugation in human liver microsomes. Species differences were seen in dermocybin glucuronidation between human, rat, mouse and pig. In conclusion, many CYP and conjugation enzymes metabolized dermocybin, whereas dermorubin was not metabolized in human liver fractions in vitro. The results indicate that dermocybin would be metabolized in humans in vivo.
Collapse
Affiliation(s)
- Johanna Yli-Öyrä
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Risto O Juvonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Mikko Herrala
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Moshe Finel
- Centre for Drug Research, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | - Jaana Rysä
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
21
|
Wu J, Han K, Sack MN. Targeting NAD+ Metabolism to Modulate Autoimmunity and Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1043-1050. [PMID: 38498807 PMCID: PMC10954088 DOI: 10.4049/jimmunol.2300693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/18/2023] [Indexed: 03/20/2024]
Abstract
NAD+ biology is involved in controlling redox balance, functioning as a coenzyme in numerous enzymatic reactions, and is a cofactor for Sirtuin enzymes and a substrate for multiple regulatory enzyme reactions within and outside the cell. At the same time, NAD+ levels are diminished with aging and are consumed during the development of inflammatory and autoimmune diseases linked to aberrant immune activation. Direct NAD+ augmentation via the NAD+ salvage and Priess-Handler pathways is being investigated as a putative therapeutic intervention to improve the healthspan in inflammation-linked diseases. In this review, we survey NAD+ biology and its pivotal roles in the regulation of immunity and inflammation. Furthermore, we discuss emerging studies evaluate NAD+ boosting in murine models and in human diseases, and we highlight areas of research that remain unresolved in understanding the mechanisms of action of these nutritional supplementation strategies.
Collapse
Affiliation(s)
- Jing Wu
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Kim Han
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Michael N Sack
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
22
|
Petr MA, Matiyevskaya F, Osborne B, Berglind M, Reves S, Zhang B, Ezra MB, Carmona-Marin LM, Syadzha MF, Mediavilla MC, Keijzers G, Bakula D, Mkrtchyan GV, Scheibye-Knudsen M. Pharmacological interventions in human aging. Ageing Res Rev 2024; 95:102213. [PMID: 38309591 DOI: 10.1016/j.arr.2024.102213] [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: 08/09/2023] [Revised: 01/15/2024] [Accepted: 01/30/2024] [Indexed: 02/05/2024]
Abstract
Pharmacological interventions are emerging as potential avenues of alleviating age-related disease. However, the knowledge of ongoing clinical trials as they relate to aging and pharmacological interventions is dispersed across a variety of mediums. In this review we summarize 136 age-related clinical trials that have been completed or are ongoing. Furthermore, we establish a database that describe the trials (AgingDB, www.agingdb.com) keeping track of the previous and ongoing clinical trials, alongside their outcomes. The aim of this review and database is to give people the ability to easily query for their trial of interest and stay up to date on the latest results. In sum, herein we give an overview of the current pharmacological strategies that have been applied to target human aging.
Collapse
Affiliation(s)
- Michael Angelo Petr
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Frida Matiyevskaya
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Brenna Osborne
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Magnus Berglind
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Simon Reves
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Bin Zhang
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Michael Ben Ezra
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Lina Maria Carmona-Marin
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Muhammad Farraz Syadzha
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Marta Cortés Mediavilla
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Guido Keijzers
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Daniela Bakula
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Garik V Mkrtchyan
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
23
|
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
|
24
|
Kiiski JI, Neuvonen M, Kurkela M, Hirvensalo P, Hämäläinen K, Tarkiainen EK, Sistonen J, Korhonen M, Khan S, Orpana A, Filppula AM, Lehtonen M, Niemi M. Solanidine is a sensitive and specific dietary biomarker for CYP2D6 activity. Hum Genomics 2024; 18:11. [PMID: 38303026 PMCID: PMC10835938 DOI: 10.1186/s40246-024-00579-8] [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] [Received: 11/02/2023] [Accepted: 01/24/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Individual assessment of CYP enzyme activities can be challenging. Recently, the potato alkaloid solanidine was suggested as a biomarker for CYP2D6 activity. Here, we aimed to characterize the sensitivity and specificity of solanidine as a CYP2D6 biomarker among Finnish volunteers with known CYP2D6 genotypes. RESULTS Using non-targeted metabolomics analysis, we identified 9152 metabolite features in the fasting plasma samples of 356 healthy volunteers. Machine learning models suggested strong association between CYP2D6 genotype-based phenotype classes with a metabolite feature identified as solanidine. Plasma solanidine concentration was 1887% higher in genetically poor CYP2D6 metabolizers (gPM) (n = 9; 95% confidence interval 755%, 4515%; P = 1.88 × 10-11), 74% higher in intermediate CYP2D6 metabolizers (gIM) (n = 89; 27%, 138%; P = 6.40 × 10-4), and 35% lower in ultrarapid CYP2D6 metabolizers (gUM) (n = 20; 64%, - 17%; P = 0.151) than in genetically normal CYP2D6 metabolizers (gNM; n = 196). The solanidine metabolites m/z 444 and 430 to solanidine concentration ratios showed even stronger associations with CYP2D6 phenotypes. Furthermore, the areas under the receiver operating characteristic and precision-recall curves for these metabolic ratios showed equal or better performances for identifying the gPM, gIM, and gUM phenotype groups than the other metabolites, their ratios to solanidine, or solanidine alone. In vitro studies with human recombinant CYP enzymes showed that solanidine was metabolized mainly by CYP2D6, with a minor contribution from CYP3A4/5. In human liver microsomes, the CYP2D6 inhibitor paroxetine nearly completely (95%) inhibited the metabolism of solanidine. In a genome-wide association study, several variants near the CYP2D6 gene associated with plasma solanidine metabolite ratios. CONCLUSIONS These results are in line with earlier studies and further indicate that solanidine and its metabolites are sensitive and specific biomarkers for measuring CYP2D6 activity. Since potato consumption is common worldwide, this biomarker could be useful for evaluating CYP2D6-mediated drug-drug interactions and to improve prediction of CYP2D6 activity in addition to genotyping.
Collapse
Affiliation(s)
- Johanna I Kiiski
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
| | - Mikko Neuvonen
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
| | - Mika Kurkela
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
| | - Päivi Hirvensalo
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
| | - Kreetta Hämäläinen
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
| | - E Katriina Tarkiainen
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Johanna Sistonen
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Genetics Laboratory, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Mari Korhonen
- Genetics Laboratory, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Sofia Khan
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Genetics Laboratory, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Arto Orpana
- Genetics Laboratory, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Anne M Filppula
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, Finland
| | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Mikko Niemi
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland.
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
25
|
Ali A, Esmaeil A, Behbehani R. Mitochondrial Chronic Progressive External Ophthalmoplegia. Brain Sci 2024; 14:135. [PMID: 38391710 PMCID: PMC10887352 DOI: 10.3390/brainsci14020135] [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: 12/25/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Chronic progressive external ophthalmoplegia (CPEO) is a rare disorder that can be at the forefront of several mitochondrial diseases. This review overviews mitochondrial CPEO encephalomyopathies to enhance accurate recognition and diagnosis for proper management. METHODS This study is conducted based on publications and guidelines obtained by selective review in PubMed. Randomized, double-blind, placebo-controlled trials, Cochrane reviews, and literature meta-analyses were particularly sought. DISCUSSION CPEO is a common presentation of mitochondrial encephalomyopathies, which can result from alterations in mitochondrial or nuclear DNA. Genetic sequencing is the gold standard for diagnosing mitochondrial encephalomyopathies, preceded by non-invasive tests such as fibroblast growth factor-21 and growth differentiation factor-15. More invasive options include a muscle biopsy, which can be carried out after uncertain diagnostic testing. No definitive treatment option is available for mitochondrial diseases, and management is mainly focused on lifestyle risk modification and supplementation to reduce mitochondrial load and symptomatic relief, such as ptosis repair in the case of CPEO. Nevertheless, various clinical trials and endeavors are still at large for achieving beneficial therapeutic outcomes for mitochondrial encephalomyopathies. KEY MESSAGES Understanding the varying presentations and genetic aspects of mitochondrial CPEO is crucial for accurate diagnosis and management.
Collapse
Affiliation(s)
| | | | - Raed Behbehani
- Neuro-Ophthalmology Unit, Ibn Sina Hospital, Al-Bahar Ophthalmology Center, Kuwait City 70035, Kuwait; (A.A.); (A.E.)
| |
Collapse
|
26
|
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
|
27
|
Drouard G, Hagenbeek FA, Whipp AM, Pool R, Hottenga JJ, Jansen R, Hubers N, Afonin A, Willemsen G, de Geus EJC, Ripatti S, Pirinen M, Kanninen KM, Boomsma DI, van Dongen J, Kaprio J. Longitudinal multi-omics study reveals common etiology underlying association between plasma proteome and BMI trajectories in adolescent and young adult twins. BMC Med 2023; 21:508. [PMID: 38129841 PMCID: PMC10740308 DOI: 10.1186/s12916-023-03198-7] [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: 07/03/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND The influence of genetics and environment on the association of the plasma proteome with body mass index (BMI) and changes in BMI remains underexplored, and the links to other omics in these associations remain to be investigated. We characterized protein-BMI trajectory associations in adolescents and adults and how these connect to other omics layers. METHODS Our study included two cohorts of longitudinally followed twins: FinnTwin12 (N = 651) and the Netherlands Twin Register (NTR) (N = 665). Follow-up comprised 4 BMI measurements over approximately 6 (NTR: 23-27 years old) to 10 years (FinnTwin12: 12-22 years old), with omics data collected at the last BMI measurement. BMI changes were calculated in latent growth curve models. Mixed-effects models were used to quantify the associations between the abundance of 439 plasma proteins with BMI at blood sampling and changes in BMI. In FinnTwin12, the sources of genetic and environmental variation underlying the protein abundances were quantified by twin models, as were the associations of proteins with BMI and BMI changes. In NTR, we investigated the association of gene expression of genes encoding proteins identified in FinnTwin12 with BMI and changes in BMI. We linked identified proteins and their coding genes to plasma metabolites and polygenic risk scores (PRS) applying mixed-effects models and correlation networks. RESULTS We identified 66 and 14 proteins associated with BMI at blood sampling and changes in BMI, respectively. The average heritability of these proteins was 35%. Of the 66 BMI-protein associations, 43 and 12 showed genetic and environmental correlations, respectively, including 8 proteins showing both. Similarly, we observed 7 and 3 genetic and environmental correlations between changes in BMI and protein abundance, respectively. S100A8 gene expression was associated with BMI at blood sampling, and the PRG4 and CFI genes were associated with BMI changes. Proteins showed strong connections with metabolites and PRSs, but we observed no multi-omics connections among gene expression and other omics layers. CONCLUSIONS Associations between the proteome and BMI trajectories are characterized by shared genetic, environmental, and metabolic etiologies. We observed few gene-protein pairs associated with BMI or changes in BMI at the proteome and transcriptome levels.
Collapse
Affiliation(s)
- Gabin Drouard
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Fiona A Hagenbeek
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Alyce M Whipp
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - René Pool
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Jouke Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Rick Jansen
- Department of Psychiatry, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress Program, Amsterdam, The Netherlands
| | - Nikki Hubers
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam, The Netherlands
| | - Aleksei Afonin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Matti Pirinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
| | - Katja M Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam, The Netherlands
| | - Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam, The Netherlands
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
28
|
Picca A, Faitg J, Auwerx J, Ferrucci L, D'Amico D. Mitophagy in human health, ageing and disease. Nat Metab 2023; 5:2047-2061. [PMID: 38036770 DOI: 10.1038/s42255-023-00930-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 10/13/2023] [Indexed: 12/02/2023]
Abstract
Maintaining optimal mitochondrial function is a feature of health. Mitophagy removes and recycles damaged mitochondria and regulates the biogenesis of new, fully functional ones preserving healthy mitochondrial functions and activities. Preclinical and clinical studies have shown that impaired mitophagy negatively affects cellular health and contributes to age-related chronic diseases. Strategies to boost mitophagy have been successfully tested in model organisms, and, recently, some have been translated into clinics. In this Review, we describe the basic mechanisms of mitophagy and how mitophagy can be assessed in human blood, the immune system and tissues, including muscle, brain and liver. We outline mitophagy's role in specific diseases and describe mitophagy-activating approaches successfully tested in humans, including exercise and nutritional and pharmacological interventions. We describe how mitophagy is connected to other features of ageing through general mechanisms such as inflammation and oxidative stress and forecast how strengthening research on mitophagy and mitophagy interventions may strongly support human health.
Collapse
Affiliation(s)
- Anna Picca
- Department of Medicine and Surgery, LUM University, Casamassima, Italy
- Fondazione Policlinico Universitario 'A. Gemelli' IRCCS, Rome, Italy
| | - Julie Faitg
- Amazentis, EPFL Innovation Park, Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Luigi Ferrucci
- Division of Intramural Research, National Institute on Aging, Baltimore, MD, USA.
| | | |
Collapse
|
29
|
Freeberg KA, Udovich CC, Martens CR, Seals DR, Craighead DH. Dietary Supplementation With NAD+-Boosting Compounds in Humans: Current Knowledge and Future Directions. J Gerontol A Biol Sci Med Sci 2023; 78:2435-2448. [PMID: 37068054 PMCID: PMC10692436 DOI: 10.1093/gerona/glad106] [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: 10/24/2022] [Indexed: 04/18/2023] Open
Abstract
Advancing age and many disease states are associated with declines in nicotinamide adenine dinucleotide (NAD+) levels. Preclinical studies suggest that boosting NAD+ abundance with precursor compounds, such as nicotinamide riboside or nicotinamide mononucleotide, has profound effects on physiological function in models of aging and disease. Translation of these compounds for oral supplementation in humans has been increasingly studied within the last 10 years; however, the clinical evidence that raising NAD+ concentrations can improve physiological function is unclear. The goal of this review was to synthesize the published literature on the effects of chronic oral supplementation with NAD+ precursors on healthy aging and age-related chronic diseases. We identified nicotinamide riboside, nicotinamide riboside co-administered with pterostilbene, and nicotinamide mononucleotide as the most common candidates in investigations of NAD+-boosting compounds for improving physiological function in humans. Studies have been performed in generally healthy midlife and older adults, adults with cardiometabolic disease risk factors such as overweight and obesity, and numerous patient populations. Supplementation with these compounds is safe, tolerable, and can increase the abundance of NAD+ and related metabolites in multiple tissues. Dosing regimens and study durations vary greatly across interventions, and small sample sizes limit data interpretation of physiological outcomes. Limitations are identified and future research directions are suggested to further our understanding of the potential efficacy of NAD+-boosting compounds for improving physiological function and extending human health span.
Collapse
Affiliation(s)
- Kaitlin A Freeberg
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - CeAnn C Udovich
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Christopher R Martens
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, USA
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Daniel H Craighead
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
30
|
Berven H, Kverneng S, Sheard E, Søgnen M, Af Geijerstam SA, Haugarvoll K, Skeie GO, Dölle C, Tzoulis C. NR-SAFE: a randomized, double-blind safety trial of high dose nicotinamide riboside in Parkinson's disease. Nat Commun 2023; 14:7793. [PMID: 38016950 PMCID: PMC10684646 DOI: 10.1038/s41467-023-43514-6] [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: 03/24/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) replenishment therapy using nicotinamide riboside (NR) shows promise for Parkinson's disease (PD) and other neurodegenerative disorders. However, the optimal dose of NR remains unknown, and doses exceeding 2000 mg daily have not been tested in humans. To evaluate the safety of high-dose NR therapy, we conducted a single-center, randomized, placebo-controlled, double-blind, phase I trial on 20 individuals with PD, randomized 1:1 on NR 1500 mg twice daily (n = 10) or placebo (n = 10) for four weeks. The trial was conducted at the Department of Neurology, Haukeland University Hospital, Bergen, Norway. The primary outcome was safety, defined as the frequency of moderate and severe adverse events. Secondary outcomes were tolerability defined as frequency of mild adverse events, change in the whole blood and urine NAD metabolome, and change in the clinical severity of PD, measured by MDS-UPDRS. All 20 participants completed the trial. The trial met all prespecified outcomes. NR therapy was well tolerated with no moderate or severe adverse events, and no significant difference in mild adverse events. NR therapy was associated with clinical improvement of total MDS-UPDRS scores. However, this change was also associated with a shorter interval since the last levodopa dose. NR greatly augmented the blood NAD metabolome with up to 5-fold increase in blood NAD+ levels. While NR-recipients exhibited a slight initial rise in serum homocysteine levels, the integrity of the methyl donor pool remained intact. Our results support extending the dose range of NR in phase II clinical trials to 3000 mg per day, with appropriate safety monitoring. Clinicaltrials.gov identifier: NCT05344404.
Collapse
Affiliation(s)
- Haakon Berven
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Simon Kverneng
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Erika Sheard
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
| | - Mona Søgnen
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
| | | | - Kristoffer Haugarvoll
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
| | - Geir-Olve Skeie
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Christian Dölle
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway.
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway.
- K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, 5020, Bergen, Norway.
| | - Charalampos Tzoulis
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway.
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway.
- K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, 5020, Bergen, Norway.
| |
Collapse
|
31
|
Nevi L, Pöllänen N, Penna F, Caretti G. Targeting Epigenetic Regulators with HDAC and BET Inhibitors to Modulate Muscle Wasting. Int J Mol Sci 2023; 24:16404. [PMID: 38003594 PMCID: PMC10671811 DOI: 10.3390/ijms242216404] [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: 10/13/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Epigenetic changes contribute to the profound alteration in the transcriptional program associated with the onset and progression of muscle wasting in several pathological conditions. Although HDACs and their inhibitors have been extensively studied in the field of muscular dystrophies, the potential of epigenetic inhibitors has only been marginally explored in other disorders associated with muscle atrophy, such as in cancer cachexia and sarcopenia. BET inhibitors represent a novel class of recently developed epigenetic drugs that display beneficial effects in a variety of diseases beyond malignancies. Based on the preliminary in vitro and preclinical data, HDACs and BET proteins contribute to the pathogenesis of cancer cachexia and sarcopenia, modulating processes related to skeletal muscle mass maintenance and/or metabolism. Thus, epigenetic drugs targeting HDACs and BET proteins may emerge as promising strategies to reverse the catabolic phenotype associated with cachexia and sarcopenia. Further preclinical studies are warranted to delve deeper into the molecular mechanisms associated with the functions of HDACs and BET proteins in muscle atrophy and to establish whether their epigenetic inhibitors represent a prospective therapeutic avenue to alleviate muscle wasting.
Collapse
Affiliation(s)
- Lorenzo Nevi
- Department of Biosciences, University of Milan, 20133 Milan, Italy;
| | - Noora Pöllänen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Torino, 10125 Torino, Italy
| | | |
Collapse
|
32
|
Bhasin S, Seals D, Migaud M, Musi N, Baur JA. Nicotinamide Adenine Dinucleotide in Aging Biology: Potential Applications and Many Unknowns. Endocr Rev 2023; 44:1047-1073. [PMID: 37364580 DOI: 10.1210/endrev/bnad019] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/28/2023] [Accepted: 06/22/2023] [Indexed: 06/28/2023]
Abstract
Recent research has unveiled an expansive role of NAD+ in cellular energy generation, redox reactions, and as a substrate or cosubstrate in signaling pathways that regulate health span and aging. This review provides a critical appraisal of the clinical pharmacology and the preclinical and clinical evidence for therapeutic effects of NAD+ precursors for age-related conditions, with a particular focus on cardiometabolic disorders, and discusses gaps in current knowledge. NAD+ levels decrease throughout life; age-related decline in NAD+ bioavailability has been postulated to be a contributor to many age-related diseases. Raising NAD+ levels in model organisms by administration of NAD+ precursors improves glucose and lipid metabolism; attenuates diet-induced weight gain, diabetes, diabetic kidney disease, and hepatic steatosis; reduces endothelial dysfunction; protects heart from ischemic injury; improves left ventricular function in models of heart failure; attenuates cerebrovascular and neurodegenerative disorders; and increases health span. Early human studies show that NAD+ levels can be raised safely in blood and some tissues by oral NAD+ precursors and suggest benefit in preventing nonmelanotic skin cancer, modestly reducing blood pressure and improving lipid profile in older adults with obesity or overweight; preventing kidney injury in at-risk patients; and suppressing inflammation in Parkinson disease and SARS-CoV-2 infection. Clinical pharmacology, metabolism, and therapeutic mechanisms of NAD+ precursors remain incompletely understood. We suggest that these early findings provide the rationale for adequately powered randomized trials to evaluate the efficacy of NAD+ augmentation as a therapeutic strategy to prevent and treat metabolic disorders and age-related conditions.
Collapse
Affiliation(s)
- Shalender Bhasin
- Department of Medicine, Harvard Medical School, Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Douglas Seals
- Department of Integrative Physiology and Medicine, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Marie Migaud
- Department of Pharmacology, Mitchell Cancer Institute, College of Medicine, University of Southern Alabama, Mobile, AL 36688, USA
| | - Nicolas Musi
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joseph A Baur
- Department of Physiology, Institute for Diabetes, Obesity & Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
33
|
Jia D, Tian Z, Wang R. Exercise mitigates age-related metabolic diseases by improving mitochondrial dysfunction. Ageing Res Rev 2023; 91:102087. [PMID: 37832607 DOI: 10.1016/j.arr.2023.102087] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 10/15/2023]
Abstract
The benefits of regular physical activity are related to delaying and reversing the onset of ageing and age-related disorders, including cardiomyopathy, neurodegenerative diseases, cancer, obesity, diabetes, and fatty liver diseases. However, the molecular mechanisms of the benefits of exercise or physical activity on ageing and age-related disorders remain poorly understood. Mitochondrial dysfunction is implicated in the pathogenesis of ageing and age-related metabolic diseases. Mitochondrial health is an important mediator of cellular function. Therefore, exercise alleviates metabolic diseases in individuals with advancing ageing and age-related diseases by the remarkable promotion of mitochondrial biogenesis and function. Exerkines are identified as signaling moieties released in response to exercise. Exerkines released by exercise have potential roles in improving mitochondrial dysfunction in response to age-related disorders. This review comprehensive summarizes the benefits of exercise in metabolic diseases, linking mitochondrial dysfunction to the onset of age-related diseases. Using relevant examples utilizing this approach, the possibility of designing therapeutic interventions based on these molecular mechanisms is addressed.
Collapse
Affiliation(s)
- Dandan Jia
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
34
|
Song Q, Zhou X, Xu K, Liu S, Zhu X, Yang J. The Safety and Antiaging Effects of Nicotinamide Mononucleotide in Human Clinical Trials: an Update. Adv Nutr 2023; 14:1416-1435. [PMID: 37619764 PMCID: PMC10721522 DOI: 10.1016/j.advnut.2023.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 08/02/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
The importance of nicotinamide adenine dinucleotide (NAD+) in human physiology is well recognized. As the NAD+ concentration in human skin, blood, liver, muscle, and brain are thought to decrease with age, finding ways to increase NAD+ status could possibly influence the aging process and associated metabolic sequelae. Nicotinamide mononucleotide (NMN) is a precursor for NAD+ biosynthesis, and in vitro/in vivo studies have demonstrated that NMN supplementation increases NAD+ concentration and could mitigate aging-related disorders such as oxidative stress, DNA damage, neurodegeneration, and inflammatory responses. The promotion of NMN as an antiaging health supplement has gained popularity due to such findings; however, since most studies evaluating the effects of NMN have been conducted in cell or animal models, a concern remains regarding the safety and physiological effects of NMN supplementation in the human population. Nonetheless, a dozen human clinical trials with NMN supplementation are currently underway. This review summarizes the current progress of these trials and NMN/NAD+ biology to clarify the potential effects of NMN supplementation and to shed light on future study directions.
Collapse
Affiliation(s)
- Qin Song
- Department of Occupational and Environmental Health, Hangzhou Normal University School of Public Health, Hangzhou, China
| | - Xiaofeng Zhou
- Department of Radiotherapy, The 2(nd) Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kexin Xu
- Department of Nutritional and Toxicological Science, Hangzhou Normal University School of Public Health, Hangzhou, China
| | - Sishi Liu
- Department of Nutritional and Toxicological Science, Hangzhou Normal University School of Public Health, Hangzhou, China
| | - Xinqiang Zhu
- Core Facility, The 4(th) Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China.
| | - Jun Yang
- Department of Nutritional and Toxicological Science, Hangzhou Normal University School of Public Health, Hangzhou, China; Zhejiang Provincial Center for Uterine Cancer Diagnosis and Therapy Research, The Affiliated Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
35
|
Xu Y, Xiao W. NAD+: An Old but Promising Therapeutic Agent for Skeletal Muscle Ageing. Ageing Res Rev 2023; 92:102106. [PMID: 39492424 DOI: 10.1016/j.arr.2023.102106] [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: 06/04/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/05/2024]
Abstract
More than a century after the discovery of nicotinamide adenine dinucleotide (NAD+), our understanding of the molecule's role in the biology of ageing continues to evolve. As a coenzyme or substrate for many enzymes, NAD+ governs a wide range of biological processes, including energy metabolism, genomic stability, signal transduction, and cell fate. NAD+ deficiency has been recognised as a bona fide hallmark of tissue degeneration, and restoring NAD+ homeostasis helps to rejuvenate multiple mechanisms associated with tissue ageing. The progressive loss of skeletal muscle homeostasis with age is directly associated with high morbidity, disability and mortality. The aetiology of skeletal muscle ageing is complex, involving mitochondrial dysfunction, senescence and stem cell depletion, autophagy defects, chronic cellular stress, intracellular ion overload, immune cell dysfunction, circadian clock disruption, microcirculation disorders, persistent denervation, and gut microbiota dysbiosis. This review focuses on the therapeutic potential of NAD+ restoration to alleviate the above pathological factors and discusses the effects of in vivo administration of different NAD+ boosting strategies on skeletal muscle homeostasis, aiming to provide a reference for combating skeletal muscle ageing.
Collapse
Affiliation(s)
- Yingying Xu
- Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Weihua Xiao
- Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
36
|
Hong S, Kim S, Kim K, Lee H. Clinical Approaches for Mitochondrial Diseases. Cells 2023; 12:2494. [PMID: 37887337 PMCID: PMC10605124 DOI: 10.3390/cells12202494] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
Mitochondria are subcontractors dedicated to energy production within cells. In human mitochondria, almost all mitochondrial proteins originate from the nucleus, except for 13 subunit proteins that make up the crucial system required to perform 'oxidative phosphorylation (OX PHOS)', which are expressed by the mitochondria's self-contained DNA. Mitochondrial DNA (mtDNA) also encodes 2 rRNA and 22 tRNA species. Mitochondrial DNA replicates almost autonomously, independent of the nucleus, and its heredity follows a non-Mendelian pattern, exclusively passing from mother to children. Numerous studies have identified mtDNA mutation-related genetic diseases. The consequences of various types of mtDNA mutations, including insertions, deletions, and single base-pair mutations, are studied to reveal their relationship to mitochondrial diseases. Most mitochondrial diseases exhibit fatal symptoms, leading to ongoing therapeutic research with diverse approaches such as stimulating the defective OXPHOS system, mitochondrial replacement, and allotropic expression of defective enzymes. This review provides detailed information on two topics: (1) mitochondrial diseases caused by mtDNA mutations, and (2) the mechanisms of current treatments for mitochondrial diseases and clinical trials.
Collapse
Affiliation(s)
- Seongho Hong
- Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea;
- Department of Medicine, Korea University College of Medicine, Seoul 02708, Republic of Korea
| | - Sanghun Kim
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea;
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Kyoungmi Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Department of Physiology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hyunji Lee
- Department of Medicine, Korea University College of Medicine, Seoul 02708, Republic of Korea
| |
Collapse
|
37
|
Kaw A, Wu T, Starosolski Z, Zhou Z, Pedroza AJ, Majumder S, Duan X, Kaw K, Pinelo JEE, Fischbein MP, Lorenzi PL, Tan L, Martinez SA, Mahmud I, Devkota L, Taegtmeyer H, Ghaghada KB, Marrelli SP, Kwartler CS, Milewicz DM. Augmenting Mitochondrial Respiration in Immature Smooth Muscle Cells with an ACTA2 Pathogenic Variant Mitigates Moyamoya-like Cerebrovascular Disease. RESEARCH SQUARE 2023:rs.3.rs-3304679. [PMID: 37886459 PMCID: PMC10602100 DOI: 10.21203/rs.3.rs-3304679/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
ACTA2 pathogenic variants altering arginine 179 cause childhood-onset strokes due to moyamoya disease (MMD)-like occlusion of the distal internal carotid arteries. A smooth muscle cell (SMC)-specific knock-in mouse model (Acta2SMC-R179C/+) inserted the mutation into 67% of aortic SMCs, whereas explanted SMCs were uniformly heterozygous. Acta2R179C/+ SMCs fail to fully differentiate and maintain stem cell-like features, including high glycolytic flux, and increasing oxidative respiration (OXPHOS) with nicotinamide riboside (NR) drives the mutant SMCs to differentiate and decreases migration. Acta2SMC-R179C/+ mice have intraluminal MMD-like occlusive lesions and strokes after carotid artery injury, whereas the similarly treated WT mice have no strokes and patent lumens. Treatment with NR prior to the carotid artery injury attenuates the strokes, MMD-like lumen occlusions, and aberrant vascular remodeling in the Acta2SMC-R179C/+ mice. These data highlight the role of immature SMCs in MMD-associated occlusive disease and demonstrate that altering SMC metabolism to drive quiescence of Acta2R179C/+ SMCs attenuates strokes and aberrant vascular remodeling in the Acta2SMC-R179C/+ mice.
Collapse
Affiliation(s)
- Anita Kaw
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Ting Wu
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, McGovern Medical School, 6431 Fannin Street, Houston, TX 77030, USA
| | - Zbigniew Starosolski
- Department of Radiology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Zhen Zhou
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Suravi Majumder
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Xueyan Duan
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Kaveeta Kaw
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Jose E. E. Pinelo
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Philip L. Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sara A. Martinez
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Iqbal Mahmud
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laxman Devkota
- Department of Radiology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Heinrich Taegtmeyer
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Ketan B. Ghaghada
- Department of Radiology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Sean P. Marrelli
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, McGovern Medical School, 6431 Fannin Street, Houston, TX 77030, USA
| | - Callie S. Kwartler
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Dianna M. Milewicz
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| |
Collapse
|
38
|
Baichuan Y, Gomes Reis M, Tavakoli S, Khodadadi N, Sohouli MH, Sernizon Guimarães N. The effects of NAD+ precursor (nicotinic acid and nicotinamide) supplementation on weight loss and related hormones: a systematic review and meta-regression analysis of randomized controlled trials. Front Nutr 2023; 10:1208734. [PMID: 37854354 PMCID: PMC10579603 DOI: 10.3389/fnut.2023.1208734] [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: 04/19/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Background Despite the fact that obesity and overweight are serious major health problems worldwide, fighting against them is also considered a challenging issue. Several interventional studies have evaluated the potential weight-reduction effect of nicotinamide adenine dinucleotide (NAD+) precursor. In order to obtain a better viewpoint from them, this study aimed to comprehensively investigate the effects of NAD+ precursor supplementation on weight loss, adiponectin, and leptin. Methods Scopus, PubMed/Medline, Web of Science, Cochrane, and Embase databases were searched using standard keywords to identify all controlled trials investigating the weight loss and related hormones effects of NAD+ precursor. Pooled weighted mean difference and 95% confidence intervals were achieved by random-effects model analysis for the best estimation of outcomes. Results Twenty two treatment arms with 5,144 participants' were included in this systematic review and meta-regression analysis. The pooled findings showed that NAD+ precursor supplementation has an effect on lowering BMI (weighted mean difference (WMD): -0.19 kg/m2, 95% confidence interval (CI): -0.29 to -0.09, p < 0.001) and increasing adiponectin (WMD: 1.59 μg/mL, 95% CI: 0.49 to 2.68, p = 0.004) in humans compared with control groups. However, no significant effect was observed on body weight and leptin. There was a significant relationship between doses of intervention with changes in BMI. In addition, subgroup analysis showed that BMI reduction was greater when receiving nicotinic acid (NA) supplementation than nicotinamide (NE) supplementation. Conclusion NAD+ precursor had significant effects on weight management with the reduction of BMI and increasing adiponectin.
Collapse
Affiliation(s)
- You Baichuan
- SDU-ANU Joint Science College, Shandong University, Weihai, China
| | - Marcela Gomes Reis
- Master in Health Science at Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- OPENS: Observatory of Epidemiology, Nutrition and Health Research, Faculdade Ciências Médicas de Minas Gerais/FELUMA, Belo Horizonte, Minas Gerais, Brazil
| | - Sogand Tavakoli
- Student Research Committee, Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navideh Khodadadi
- Student Research Committee, Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassan Sohouli
- Student Research Committee, Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nathalia Sernizon Guimarães
- OPENS: Observatory of Epidemiology, Nutrition and Health Research, Faculdade Ciências Médicas de Minas Gerais/FELUMA, Belo Horizonte, Minas Gerais, Brazil
- Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
39
|
Perez-Sanchez C, Escudero-Contreras A, Cerdó T, Sánchez-Mendoza LM, Llamas-Urbano A, la Rosa IAD, Pérez-Rodriguez M, Muñoz-Barrera L, Del Carmen Abalos-Aguilera M, Barbarroja N, Calvo J, Ortega-Castro R, Ruiz-Vilchez D, Moreno JA, Burón MI, González-Reyes JA, Collantes-Estevez E, Lopez-Pedrera C, Villalba JM. Preclinical Characterization of Pharmacologic NAD + Boosting as a Promising Therapeutic Approach in Rheumatoid Arthritis. Arthritis Rheumatol 2023; 75:1749-1761. [PMID: 37094367 DOI: 10.1002/art.42528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/08/2023] [Accepted: 04/04/2023] [Indexed: 04/26/2023]
Abstract
OBJECTIVE We analyzed NAD+ metabolism in patients with rheumatoid arthritis (RA), its association with disease activity and clinical outcomes of RA, and the therapeutic potential of pharmacologic NAD+ boosting. METHODS Our study included 253 participants. In the first cohort, comprising 153 RA patients and 56 healthy donors, we assessed NAD+ levels and NAD+ -related gene pathways. We analyzed 92 inflammatory molecules by proximity extension assay. In the second cohort, comprising 44 RA patients starting anti-tumor necrosis factor (anti-TNF) drugs, we evaluated changes in NAD+ levels and their association with clinical response after 3 months. Mechanistic studies were performed ex vivo on peripheral blood mononuclear cells (PBMCs) from patients with RA to test the beneficial effects of NAD+ boosters, such as nicotinamide and nicotinamide riboside. RESULTS Reduced NAD+ levels were found in RA samples, in line with altered activity and expression of genes involved in NAD+ consumption (sirtuins, poly[ADP-ribose] polymerase, CD38), transport (connexin 43), and biosynthesis (NAMPT, NMNATs). Unsupervised clustering analysis identified a group of RA patients with the highest inflammatory profile, the lowest NAD+ levels, and the highest disease activity (as shown by the Disease Activity Score in 28 joints). NAD+ levels were modulated by anti-TNF therapy in parallel with the clinical response. In vitro studies using PBMCs from RA patients showed that nicotinamide riboside and nicotinamide increased NAD+ levels via NAMPT and NMNAT and reduced their prooxidative, proapoptotic, and proinflammatory status. CONCLUSION RA patients display altered NAD+ metabolism, directly linked to their inflammatory and disease activity status, which was reverted by anti-TNF therapy. The preclinical beneficial effects of NAD+ boosters, as shown in leukocytes from RA patients, along with their proven clinical safety, might pave the way for the development of clinical trials using these compounds.
Collapse
Affiliation(s)
- Carlos Perez-Sanchez
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, and Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, Campus de Excelencia Internacional Agroalimentario (ceiA3), Córdoba, Spain; Cobiomic Bioscience
| | | | - Tomás Cerdó
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Luz Marina Sánchez-Mendoza
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, Córdoba, Spain
| | - Adrián Llamas-Urbano
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Iván Arias-de la Rosa
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Miguel Pérez-Rodriguez
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, Córdoba, Spain
| | - Laura Muñoz-Barrera
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | | | - Nuria Barbarroja
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Jerusalem Calvo
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Rafaela Ortega-Castro
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Desiree Ruiz-Vilchez
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Juan Antonio Moreno
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, and Laboratory GE-06, IMIBIC, Nephrology Service, Reina Sofia University Hospital, ceiA3, Córdoba, Spain
| | - María Isabel Burón
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, Córdoba, Spain
| | - José Antonio González-Reyes
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, Córdoba, Spain
| | - Eduardo Collantes-Estevez
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Chary Lopez-Pedrera
- Rheumatology Service, IMIBIC, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - José Manuel Villalba
- Department of Cell Biology, Immunology and Physiology, Agrifood Campus of International Excellence, University of Córdoba, ceiA3, Córdoba, Spain
| |
Collapse
|
40
|
U-Din M, de Mello VD, Tuomainen M, Raiko J, Niemi T, Fromme T, Klåvus A, Gautier N, Haimilahti K, Lehtonen M, Kristiansen K, Newman JW, Pietiläinen KH, Pihlajamäki J, Amri EZ, Klingenspor M, Nuutila P, Pirinen E, Hanhineva K, Virtanen KA. Cold-stimulated brown adipose tissue activation is related to changes in serum metabolites relevant to NAD + metabolism in humans. Cell Rep 2023; 42:113131. [PMID: 37708023 DOI: 10.1016/j.celrep.2023.113131] [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: 05/31/2022] [Revised: 07/06/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023] Open
Abstract
Cold-induced brown adipose tissue (BAT) activation is considered to improve metabolic health. In murine BAT, cold increases the fundamental molecule for mitochondrial function, nicotinamide adenine dinucleotide (NAD+), but limited knowledge of NAD+ metabolism during cold in human BAT metabolism exists. We show that cold increases the serum metabolites of the NAD+ salvage pathway (nicotinamide and 1-methylnicotinamide) in humans. Additionally, individuals with cold-stimulated BAT activation have decreased levels of metabolites from the de novo NAD+ biosynthesis pathway (tryptophan, kynurenine). Serum nicotinamide correlates positively with cold-stimulated BAT activation, whereas tryptophan and kynurenine correlate negatively. Furthermore, the expression of genes involved in NAD+ biosynthesis in BAT is related to markers of metabolic health. Our data indicate that cold increases serum tryptophan conversion to nicotinamide to be further utilized by BAT. We conclude that NAD+ metabolism is activated upon cold in humans and is probably regulated in a coordinated fashion by several tissues.
Collapse
Affiliation(s)
- Mueez U-Din
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland
| | - Vanessa D de Mello
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Marjo Tuomainen
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Juho Raiko
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Tarja Niemi
- Department of Surgery, Turku University Hospital, Turku, Finland
| | - Tobias Fromme
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Anton Klåvus
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | | | - Kimmo Haimilahti
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; Research Program for Stem Cells and Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Marko Lehtonen
- Department of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | | | - John W Newman
- Obesity and Metabolism Research Unit, USDA-ARS Western Human Nutrition Research Center, Davis, CA, USA; West Coast Metabolomics Center, Davis Genome Center, University of California, Davis, Davis, CA 95616, USA; Department of Nutrition, University of California, Davis, Davis, CA 95616, USA
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Obesity Center, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Jussi Pihlajamäki
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Endocrinology and Clinical Nutrition, Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | | | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Pirjo Nuutila
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland; Department of Endocrinology, Turku University Hospital, Turku, Finland
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; Research Unit for Internal Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Kati Hanhineva
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Life Technologies, Food Chemistry and Food Development Unit, University of Turku, Turku, Finland; Department of Biology and Biological Engineering, Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Kirsi A Virtanen
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland; Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Endocrinology and Clinical Nutrition, Department of Medicine, Kuopio University Hospital, Kuopio, Finland; Department of Endocrinology, Turku University Hospital, Turku, Finland.
| |
Collapse
|
41
|
Alegre GFS, Pastore GM. NAD+ Precursors Nicotinamide Mononucleotide (NMN) and Nicotinamide Riboside (NR): Potential Dietary Contribution to Health. Curr Nutr Rep 2023; 12:445-464. [PMID: 37273100 PMCID: PMC10240123 DOI: 10.1007/s13668-023-00475-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 06/06/2023]
Abstract
PURPOSE OF REVIEW NAD+ is a vital molecule that takes part as a redox cofactor in several metabolic reactions besides being used as a substrate in important cellular signaling in regulation pathways for energetic, genotoxic, and infectious stress. In stress conditions, NAD+ biosynthesis and levels decrease as well as the activity of consuming enzymes rises. Dietary precursors can promote NAD+ biosynthesis and increase intracellular levels, being a potential strategy for reversing physiological decline and preventing diseases. In this review, we will show the biochemistry and metabolism of NAD+ precursors NR (nicotinamide riboside) and NMN (nicotinamide mononucleotide), the latest findings on their beneficial physiological effects, their interplay with gut microbiota, and the future perspectives for research in nutrition and food science fields. RECENT FINDINGS NMN and NR demonstrated protect against diabetes, Alzheimer disease, endothelial dysfunction, and inflammation. They also reverse gut dysbiosis and promote beneficial effects at intestinal and extraintestinal levels. NR and NMN have been found in vegetables, meat, and milk, and microorganisms in fermented beverages can also produce them. NMN and NR can be obtained through the diet either in their free form or as metabolites derivate from the digestion of NAD+. The prospection of NR and NMN to find potential food sources and their dietary contribution in increasing NAD+ levels are still an unexplored field of research. Moreover, it could enable the development of new functional foods and processing strategies to maintain and enhance their physiological benefits, besides the studies of new raw materials for extraction and biotechnological development.
Collapse
Affiliation(s)
- Gabriela Fabiana Soares Alegre
- Department of Food Science and Nutrition, Faculty of Food Engineering, State University of Campinas, Campinas, Brazil.
- Laboratory of Bioflavours and Bioactive Compounds-Rua Monteiro Lobato, Cidade Universitária "Zeferino Vaz" Barão Geraldo, 80-CEP 13083-862, Campinas, SP, Brazil.
| | - Glaucia Maria Pastore
- Department of Food Science and Nutrition, Faculty of Food Engineering, State University of Campinas, Campinas, Brazil
- Laboratory of Bioflavours and Bioactive Compounds-Rua Monteiro Lobato, Cidade Universitária "Zeferino Vaz" Barão Geraldo, 80-CEP 13083-862, Campinas, SP, Brazil
| |
Collapse
|
42
|
Biţă A, Scorei IR, Ciocîlteu MV, Nicolaescu OE, Pîrvu AS, Bejenaru LE, Rău G, Bejenaru C, Radu A, Neamţu J, Mogoşanu GD, Benner SA. Nicotinamide Riboside, a Promising Vitamin B 3 Derivative for Healthy Aging and Longevity: Current Research and Perspectives. Molecules 2023; 28:6078. [PMID: 37630330 PMCID: PMC10459282 DOI: 10.3390/molecules28166078] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Many studies have suggested that the oxidized form of nicotinamide adenine dinucleotide (NAD+) is involved in an extensive spectrum of human pathologies, including neurodegenerative disorders, cardiomyopathy, obesity, and diabetes. Further, healthy aging and longevity appear to be closely related to NAD+ and its related metabolites, including nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN). As a dietary supplement, NR appears to be well tolerated, having better pharmacodynamics and greater potency. Unfortunately, NR is a reactive molecule, often unstable during its manufacturing, transport, and storage. Recently, work related to prebiotic chemistry discovered that NR borate is considerably more stable than NR itself. However, immediately upon consumption, the borate dissociates from the NR borate and is lost in the body through dilution and binding to other species, notably carbohydrates such as fructose and glucose. The NR left behind is expected to behave pharmacologically in ways identical to NR itself. This review provides a comprehensive summary (through Q1 of 2023) of the literature that makes the case for the consumption of NR as a dietary supplement. It then summarizes the challenges of delivering quality NR to consumers using standard synthesis, manufacture, shipping, and storage approaches. It concludes by outlining the advantages of NR borate in these processes.
Collapse
Affiliation(s)
- Andrei Biţă
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Dolj County, Romania; (A.B.); (L.E.B.); (G.D.M.)
- Department of Biochemistry, BioBoron Research Institute, S.C. Natural Research S.R.L., 31B Dunării Street, 207465 Podari, Dolj County, Romania; (M.V.C.); (G.R.); (J.N.)
| | - Ion Romulus Scorei
- Department of Biochemistry, BioBoron Research Institute, S.C. Natural Research S.R.L., 31B Dunării Street, 207465 Podari, Dolj County, Romania; (M.V.C.); (G.R.); (J.N.)
| | - Maria Viorica Ciocîlteu
- Department of Biochemistry, BioBoron Research Institute, S.C. Natural Research S.R.L., 31B Dunării Street, 207465 Podari, Dolj County, Romania; (M.V.C.); (G.R.); (J.N.)
- Department of Analytical Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Dolj County, Romania
| | - Oana Elena Nicolaescu
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Dolj County, Romania;
| | - Andreea Silvia Pîrvu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Dolj County, Romania;
| | - Ludovic Everard Bejenaru
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Dolj County, Romania; (A.B.); (L.E.B.); (G.D.M.)
| | - Gabriela Rău
- Department of Biochemistry, BioBoron Research Institute, S.C. Natural Research S.R.L., 31B Dunării Street, 207465 Podari, Dolj County, Romania; (M.V.C.); (G.R.); (J.N.)
- Department of Organic Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Dolj County, Romania
| | - Cornelia Bejenaru
- Department of Pharmaceutical Botany, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Dolj County, Romania; (C.B.); (A.R.)
| | - Antonia Radu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Dolj County, Romania; (C.B.); (A.R.)
| | - Johny Neamţu
- Department of Biochemistry, BioBoron Research Institute, S.C. Natural Research S.R.L., 31B Dunării Street, 207465 Podari, Dolj County, Romania; (M.V.C.); (G.R.); (J.N.)
- Department of Physics, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Dolj County, Romania
| | - George Dan Mogoşanu
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Dolj County, Romania; (A.B.); (L.E.B.); (G.D.M.)
- Department of Biochemistry, BioBoron Research Institute, S.C. Natural Research S.R.L., 31B Dunării Street, 207465 Podari, Dolj County, Romania; (M.V.C.); (G.R.); (J.N.)
| | - Steven A. Benner
- Foundation for Applied Molecular Evolution (FfAME), 13709 Progress Avenue, Room N112, Alachua, FL 32615, USA;
| |
Collapse
|
43
|
Chinnapaka S, Malekzadeh H, Tirmizi Z, Arellano JA, Ejaz A. Nicotinamide Riboside Improves Stemness of Human Adipose-Derived Stem Cells and Inhibits Terminal Adipocyte Differentiation. Pharmaceuticals (Basel) 2023; 16:1134. [PMID: 37631051 PMCID: PMC10458272 DOI: 10.3390/ph16081134] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Adipose tissue plays a crucial role in maintaining metabolic homeostasis by serving as a storage site for excess fat and protecting other organs from the detrimental effects of lipotoxicity. However, the aging process is accompanied by a redistribution of fat, characterized by a decrease in insulin-sensitive subcutaneous adipose depot and an increase in insulin-resistant visceral adipose depot. This age-related alteration in adipose tissue distribution has implications for metabolic health. Adipose-derived stem cells (ASCs) play a vital role in the regeneration of adipose tissue. However, aging negatively impacts the stemness and regenerative potential of ASCs. The accumulation of oxidative stress and mitochondrial dysfunction-associated cellular damage contributes to the decline in stemness observed in aged ASCs. Nicotinamide adenine dinucleotide (NAD+) is a crucial metabolite that is involved in maintaining cellular homeostasis and stemness. The dysregulation of NAD+ levels with age has been associated with metabolic disorders and the loss of stemness. In this study, we aimed to investigate the effects of nicotinamide riboside (NR), a precursor of NAD+, on the stemness of human ASCs in cell culture. Our findings reveal that adipogenesis is accompanied by an increase in mitochondrial activity and the production of reactive oxygen species (ROS). However, treatment with NR leads to a reduction in mitochondrial activity and ROS production in ASCs. Furthermore, NR administration improves the stemness-related genes expression in ASCs and mitigates their propensity for adipocyte differentiation. These results suggest that NR treatment holds promise as a potential strategy to rejuvenate the stemness of aged ASCs. Further investigations, including in vivo evaluations using animal models and human studies, will be necessary to validate these findings and establish the clinical potential of this well-established drug for enhancing the stemness of aged stem cells.
Collapse
Affiliation(s)
| | | | | | | | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
44
|
Damgaard MV, Treebak JT. What is really known about the effects of nicotinamide riboside supplementation in humans. SCIENCE ADVANCES 2023; 9:eadi4862. [PMID: 37478182 PMCID: PMC10361580 DOI: 10.1126/sciadv.adi4862] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/21/2023] [Indexed: 07/23/2023]
Abstract
Nicotinamide riboside is a precursor to the important cofactor nicotinamide adenine dinucleotide and has elicited metabolic benefits in multiple preclinical studies. In 2016, the first clinical trial of nicotinamide riboside was conducted to test the safety and efficacy of human supplementation. Many trials have since been conducted aiming to delineate benefits to metabolic health and severe diseases in humans. This review endeavors to summarize and critically assess the 25 currently published research articles on human nicotinamide riboside supplementation to identify any poorly founded claims and assist the field in elucidating the actual future potential for nicotinamide riboside. Collectively, oral nicotinamide riboside supplementation has displayed few clinically relevant effects, and there is an unfortunate tendency in the literature to exaggerate the importance and robustness of reported effects. Even so, nicotinamide riboside may play a role in the reduction of inflammatory states and has shown some potential in the treatment of diverse severe diseases.
Collapse
Affiliation(s)
- Mads V. Damgaard
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jonas T. Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
45
|
Hintikka JE, Ahtiainen JP, Permi P, Jalkanen S, Lehtonen M, Pekkala S. Aerobic exercise training and gut microbiome-associated metabolic shifts in women with overweight: a multi-omic study. Sci Rep 2023; 13:11228. [PMID: 37433843 DOI: 10.1038/s41598-023-38357-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/06/2023] [Indexed: 07/13/2023] Open
Abstract
Physical activity is essential in weight management, improves overall health, and mitigates obesity-related risk markers. Besides inducing changes in systemic metabolism, habitual exercise may improve gut's microbial diversity and increase the abundance of beneficial taxa in a correlated fashion. Since there is a lack of integrative omics studies on exercise and overweight populations, we studied the metabolomes and gut microbiota associated with programmed exercise in obese individuals. We measured the serum and fecal metabolites of 17 adult women with overweight during a 6-week endurance exercise program. Further, we integrated the exercise-responsive metabolites with variations in the gut microbiome and cardiorespiratory parameters. We found clear correlation with several serum and fecal metabolites, and metabolic pathways, during the exercise period in comparison to the control period, indicating increased lipid oxidation and oxidative stress. Especially, exercise caused co-occurring increase in levels of serum lyso-phosphatidylcholine moieties and fecal glycerophosphocholine. This signature was associated with several microbial metagenome pathways and the abundance of Akkermansia. The study demonstrates that, in the absence of body composition changes, aerobic exercise can induce metabolic shifts that provide substrates for beneficial gut microbiota in overweight individuals.
Collapse
Affiliation(s)
- Jukka E Hintikka
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.
| | - Juha P Ahtiainen
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Perttu Permi
- Department of Biological and Environmental Science, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
- Department of Chemistry, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Sirpa Jalkanen
- MediCity and InFLAMES Flagship, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Marko Lehtonen
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Satu Pekkala
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
46
|
Drouard G, Hagenbeek FA, Whipp A, Pool R, Hottenga JJ, Jansen R, Hubers N, Afonin A, Willemsen G, de Geus EJC, Ripatti S, Pirinen M, Kanninen KM, Boomsma DI, van Dongen J, Kaprio J. Longitudinal multi-omics study reveals common etiology underlying association between plasma proteome and BMI trajectories in adolescent and young adult twins. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.28.23291995. [PMID: 37425750 PMCID: PMC10327285 DOI: 10.1101/2023.06.28.23291995] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Background The influence of genetics and environment on the association of the plasma proteome with body mass index (BMI) and changes in BMI remain underexplored, and the links to other omics in these associations remain to be investigated. We characterized protein-BMI trajectory associations in adolescents and adults and how these connect to other omics layers. Methods Our study included two cohorts of longitudinally followed twins: FinnTwin12 (N=651) and the Netherlands Twin Register (NTR) (N=665). Follow-up comprised four BMI measurements over approximately 6 (NTR: 23-27 years old) to 10 years (FinnTwin12: 12-22 years old), with omics data collected at the last BMI measurement. BMI changes were calculated using latent growth curve models. Mixed-effects models were used to quantify the associations between the abundance of 439 plasma proteins with BMI at blood sampling and changes in BMI. The sources of genetic and environmental variation underlying the protein abundances were quantified using twin models, as were the associations of proteins with BMI and BMI changes. In NTR, we investigated the association of gene expression of genes encoding proteins identified in FinnTwin12 with BMI and changes in BMI. We linked identified proteins and their coding genes to plasma metabolites and polygenic risk scores (PRS) using mixed-effect models and correlation networks. Results We identified 66 and 14 proteins associated with BMI at blood sampling and changes in BMI, respectively. The average heritability of these proteins was 35%. Of the 66 BMI-protein associations, 43 and 12 showed genetic and environmental correlations, respectively, including 8 proteins showing both. Similarly, we observed 6 and 4 genetic and environmental correlations between changes in BMI and protein abundance, respectively. S100A8 gene expression was associated with BMI at blood sampling, and the PRG4 and CFI genes were associated with BMI changes. Proteins showed strong connections with many metabolites and PRSs, but we observed no multi-omics connections among gene expression and other omics layers. Conclusions Associations between the proteome and BMI trajectories are characterized by shared genetic, environmental, and metabolic etiologies. We observed few gene-protein pairs associated with BMI or changes in BMI at the proteome and transcriptome levels.
Collapse
Affiliation(s)
- Gabin Drouard
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Fiona A. Hagenbeek
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Alyce Whipp
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - René Pool
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Jouke Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Rick Jansen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Psychiatry, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress Program, Amsterdam, The Netherlands
| | - Nikki Hubers
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam, The Netherlands
| | - Aleksei Afonin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - BIOS Consortium
- Biobank-based Integrative Omics Study Consortium. Lists of authors and their affiliations appear in the supplementary material (see Additional file 1)
| | | | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Eco J. C. de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Matti Pirinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
| | - Katja M. Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Dorret I. Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam, The Netherlands
| | - Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam, The Netherlands
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
47
|
Calvani R, Picca A, Coelho-Júnior HJ, Tosato M, Marzetti E, Landi F. "Diet for the prevention and management of sarcopenia". Metabolism 2023:155637. [PMID: 37352971 DOI: 10.1016/j.metabol.2023.155637] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/25/2023]
Abstract
Sarcopenia is a geriatric condition characterized by a progressive loss of skeletal muscle mass and strength, with an increased risk of adverse health outcomes (e.g., falls, disability, institutionalization, reduced quality of life, mortality). Pharmacological remedies are currently unavailable for preventing the development of sarcopenia, halting its progression, or impeding its negative health outcomes. The most effective strategies to contrast sarcopenia rely on the adoption of healthier lifestyle behaviors, including adherence to high-quality diets and regular physical activity. In this review, the role of nutrition in the prevention and management of sarcopenia is summarized. Special attention is given to current "blockbuster" dietary regimes and agents used to counteract age-related muscle wasting, together with their putative mechanisms of action. Issues related to the design and implementation of effective nutritional strategies are discussed, with a focus on unanswered questions on the most appropriate timing of nutritional interventions to preserve muscle health and function into old age. A brief description is also provided on new technologies that can facilitate the development and implementation of personalized nutrition plans to contrast sarcopenia.
Collapse
Affiliation(s)
- Riccardo Calvani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy.
| | - Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy; Department of Medicine and Surgery, LUM University, 70100 Casamassima, Italy.
| | - Hélio José Coelho-Júnior
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Matteo Tosato
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy.
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy.
| | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy.
| |
Collapse
|
48
|
Ahmadi A, Begue G, Valencia AP, Norman JE, Lidgard B, Bennett BJ, Van Doren MP, Marcinek DJ, Fan S, Prince DK, Gamboa J, Himmelfarb J, de Boer IH, Kestenbaum BR, Roshanravan B. Randomized crossover clinical trial of coenzyme Q10 and nicotinamide riboside in chronic kidney disease. JCI Insight 2023; 8:e167274. [PMID: 37159264 PMCID: PMC10393227 DOI: 10.1172/jci.insight.167274] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 05/03/2023] [Indexed: 05/10/2023] Open
Abstract
BackgroundCurrent studies suggest mitochondrial dysfunction is a major contributor to impaired physical performance and exercise intolerance in chronic kidney disease (CKD). We conducted a clinical trial of coenzyme Q10 (CoQ10) and nicotinamide riboside (NR) to determine their impact on exercise tolerance and metabolic profile in patients with CKD.MethodsWe conducted a randomized, placebo-controlled, double-blind, crossover trial comparing CoQ10, NR, and placebo in 25 patients with an estimated glomerular filtration rate (eGFR) of less than 60mL/min/1.73 m2. Participants received NR (1,000 mg/day), CoQ10 (1,200 mg/day), or placebo for 6 weeks each. The primary outcomes were aerobic capacity measured by peak rate of oxygen consumption (VO2 peak) and work efficiency measured using graded cycle ergometry testing. We performed semitargeted plasma metabolomics and lipidomics.ResultsParticipant mean age was 61.0 ± 11.6 years and mean eGFR was 36.9 ± 9.2 mL/min/1.73 m2. Compared with placebo, we found no differences in VO2 peak (P = 0.30, 0.17), total work (P = 0.47, 0.77), and total work efficiency (P = 0.46, 0.55) after NR or CoQ10 supplementation. NR decreased submaximal VO2 at 30 W (P = 0.03) and VO2 at 60 W (P = 0.07) compared with placebo. No changes in eGFR were observed after NR or CoQ10 treatment (P = 0.14, 0.88). CoQ10 increased free fatty acids and decreased complex medium- and long-chain triglycerides. NR supplementation significantly altered TCA cycle intermediates and glutamate that were involved in reactions that exclusively use NAD+ and NADP+ as cofactors. NR decreased a broad range of lipid groups including triglycerides and ceramides.ConclusionsSix weeks of treatment with NR or CoQ10 improved markers of systemic mitochondrial metabolism and lipid profiles but did not improve VO2 peak or total work efficiency.Trial registrationClinicalTrials.gov NCT03579693.FundingNational Institutes of Diabetes and Digestive and Kidney Diseases (grants R01 DK101509, R03 DK114502, R01 DK125794, and R01 DK101509).
Collapse
Affiliation(s)
- Armin Ahmadi
- Department of Medicine, Division of Nephrology, UCD, Davis, California, USA
| | - Gwenaelle Begue
- Kinesiology Department, California State University, Sacramento, California, USA
| | - Ana P. Valencia
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Jennifer E. Norman
- Department of Internal Medicine, Division of Cardiovascular Medicine, UCD, Davis, California, USA
| | - Benjamin Lidgard
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Brian J. Bennett
- Obesity and Metabolism Research Unit, Western Human Nutrition Research Center, USDA, ARS, Davis, California, USA
| | | | - David J. Marcinek
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Sili Fan
- Department of Biostatistics, UCD, Davis, California, USA
| | - David K. Prince
- Department of Medicine, Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | - Jorge Gamboa
- School of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Jonathan Himmelfarb
- Department of Medicine, Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | - Ian H. de Boer
- Department of Medicine, Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | - Bryan R. Kestenbaum
- Department of Medicine, Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | - Baback Roshanravan
- Department of Medicine, Division of Nephrology, UCD, Davis, California, USA
| |
Collapse
|
49
|
Beltrà M, Pöllänen N, Fornelli C, Tonttila K, Hsu MY, Zampieri S, Moletta L, Corrà S, Porporato PE, Kivelä R, Viscomi C, Sandri M, Hulmi JJ, Sartori R, Pirinen E, Penna F. NAD + repletion with niacin counteracts cancer cachexia. Nat Commun 2023; 14:1849. [PMID: 37012289 PMCID: PMC10070388 DOI: 10.1038/s41467-023-37595-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 03/21/2023] [Indexed: 04/05/2023] Open
Abstract
Cachexia is a debilitating wasting syndrome and highly prevalent comorbidity in cancer patients. It manifests especially with energy and mitochondrial metabolism aberrations that promote tissue wasting. We recently identified nicotinamide adenine dinucleotide (NAD+) loss to associate with muscle mitochondrial dysfunction in cancer hosts. In this study we confirm that depletion of NAD+ and downregulation of Nrk2, an NAD+ biosynthetic enzyme, are common features of severe cachexia in different mouse models. Testing NAD+ repletion therapy in cachectic mice reveals that NAD+ precursor, vitamin B3 niacin, efficiently corrects tissue NAD+ levels, improves mitochondrial metabolism and ameliorates cancer- and chemotherapy-induced cachexia. In a clinical setting, we show that muscle NRK2 is downregulated in cancer patients. The low expression of NRK2 correlates with metabolic abnormalities underscoring the significance of NAD+ in the pathophysiology of human cancer cachexia. Overall, our results propose NAD+ metabolism as a therapy target for cachectic cancer patients.
Collapse
Affiliation(s)
- Marc Beltrà
- Experimental Medicine and Clinical Pathology Unit, Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Noora Pöllänen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Claudia Fornelli
- Experimental Medicine and Clinical Pathology Unit, Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | - Kialiina Tonttila
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Myriam Y Hsu
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Sandra Zampieri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- CIR-MYO Myology Center, University of Padova, Padova, Italy
| | - Lucia Moletta
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | | | - Paolo E Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Riikka Kivelä
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
- Wihuri Research Institute, Helsinki, Finland
| | - Carlo Viscomi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Study Centre for Neurodegeneration, University of Padova (CESNE), Padova, Italy
| | - Marco Sandri
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Juha J Hulmi
- Faculty of Sport and Health Sciences, NeuroMuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - Roberta Sartori
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
- Veneto Institute of Molecular Medicine, Padova, Italy.
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.
| | - Fabio Penna
- Experimental Medicine and Clinical Pathology Unit, Department of Clinical and Biological Sciences, University of Torino, Turin, Italy.
| |
Collapse
|
50
|
Karim M, Iqbal T, Nawaz A, Yaku K, Nakagawa T. Deletion of Nmnat1 in Skeletal Muscle Leads to the Reduction of NAD + Levels but Has No Impact on Skeletal Muscle Morphology and Fiber Types. J Nutr Sci Vitaminol (Tokyo) 2023; 69:184-189. [PMID: 37394423 DOI: 10.3177/jnsv.69.184] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a coenzyme that mediates many redox reactions in energy metabolism. NAD+ is also a substrate for ADP-ribosylation and deacetylation by poly (ADP-ribose) polymerase and sirtuin, respectively. Nicotinamide mononucleotide adenylyltransferase 1 (Nmnat1) is a NAD+ biosynthesizing enzyme found in the nucleus. Recent research has shown that the maintaining NAD+ levels is critical for sustaining muscle functions both in physiological and pathological conditions. However, the role of Nmnat1 in skeletal muscle remains unexplored. In this study, we generated skeletal muscle-specific Nmnat1 knockout (M-Nmnat1 KO) mice and investigated its role in skeletal muscle. We found that NAD+ levels were significantly lower in the skeletal muscle of M-Nmnat1 KO mice than in control mice. M-Nmnat1 KO mice, in contrast, had similar body weight and normal muscle histology. Furthermore, the distribution of muscle fiber size and gene expressions of muscle fiber type gene expression were comparable in M-Nmnat1 KO and control mice. Finally, we investigated the role of Nmnat1 in muscle regeneration using cardiotoxin-induced muscle injury model, but muscle regeneration appeared almost normal in M-Nmnat1 KO mice. These findings imply that Nmnat1 has a redundancy in the pathophysiology of skeletal muscle.
Collapse
Affiliation(s)
- Mariam Karim
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama
| | - Tooba Iqbal
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama
| | - Allah Nawaz
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama
| | - Keisuke Yaku
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama
- Research Center for Pre-Disease Science, University of Toyama
| |
Collapse
|