1
|
Su Y, Sun J, Li X, Huang F, Kong Y, Chen Z, Zhang J, Qin D, Chen X, Wang Z, Pei Y, Gong M, Yang K, Xu M, Dong Y, He Q, Zhang ZN, Sheng Z, Deng Q, Wang H, Wang G, Hu P, Le R, Gao S, Li W. CD47-blocking antibody confers metabolic benefits against obesity. Cell Rep Med 2025; 6:102089. [PMID: 40267910 DOI: 10.1016/j.xcrm.2025.102089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 07/22/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025]
Abstract
CD47-blocking antibody is a well-known potential antibody drug for tumor immunotherapy. However, it is unclear whether CD47-blocking antibody can protect against metabolic disorders. We report that high-fat diet (HFD)-induced obesity increases CD47 expression, while exercise downregulates it in skeletal muscle. Administration of CD47-blocking antibody in mice prevents HFD-induced weight gain and glucose intolerance, enhances exercise capacity, and improves body composition and skeletal muscle mitochondrial function. Mechanistically, the protective effects conferred by CD47-blocking antibody are mediated through activation of AMP-activated protein kinase (AMPK) in skeletal muscle. Consistently, muscle-specific CD47-knockout mice show similar metabolic improvements, indicating a direct muscle-specific role of CD47 in regulating AMPK activation in vivo. Furthermore, the CD47-blocking antibody reduces the phosphorylation of heat shock protein 90α (HSP90α) to activate AMPK in skeletal muscle. In conclusion, CD47-blocking antibody confers metabolic benefits by activating the AMPK pathway in skeletal muscle.
Collapse
Affiliation(s)
- Yajuan Su
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China; Tsingtao Advanced Research Institute, Tongji University, Qingdao 266071, China
| | - Jingyu Sun
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Xiaobo Li
- Tsingtao Advanced Research Institute, Tongji University, Qingdao 266071, China
| | - Feier Huang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Yunhui Kong
- Institute of Modern Biology, Nanjing University, Nanjing 210023, China
| | - Zian Chen
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Jingzhi Zhang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Duran Qin
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Xiangyi Chen
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Zhaoyue Wang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Yu Pei
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Solna, Sweden
| | - Mengting Gong
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Kaijiang Yang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Minglu Xu
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Yu Dong
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Qing He
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Zhen-Ning Zhang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Zhejin Sheng
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Solna, Sweden
| | - Hong Wang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Gaowei Wang
- Institute of Modern Biology, Nanjing University, Nanjing 210023, China
| | - Ping Hu
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai TenthPeople's Hospital Affiliated to Tongji University, Shanghai 200031, China; Guangzhou Laboratory, No. 9 XingDaoHuan Road, Guanghzou International Bio lsland, Guangzhou 510005, China
| | - Rongrong Le
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China.
| | - Shaorong Gao
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China; Tsingtao Advanced Research Institute, Tongji University, Qingdao 266071, China.
| | - Weida Li
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China; Tsingtao Advanced Research Institute, Tongji University, Qingdao 266071, China.
| |
Collapse
|
2
|
Machado IF, Palmeira CM, Rolo AP. Sestrin2 is a central regulator of mitochondrial stress responses in disease and aging. Ageing Res Rev 2025; 109:102762. [PMID: 40320152 DOI: 10.1016/j.arr.2025.102762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/09/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025]
Abstract
Mitochondria supply most of the energy for cellular functions and coordinate numerous cellular pathways. Their dynamic nature allows them to adjust to stress and cellular metabolic demands, thus ensuring the preservation of cellular homeostasis. Loss of normal mitochondrial function compromises cell survival and has been implicated in the development of many diseases and in aging. Although exposure to continuous or severe stress has adverse effects on cells, mild mitochondrial stress enhances mitochondrial function and potentially extends health span through mitochondrial adaptive responses. Over the past few decades, sestrin2 (SESN2) has emerged as a pivotal regulator of stress responses. For instance, SESN2 responds to genotoxic, oxidative, and metabolic stress, promoting cellular defense against stress-associated damage. Here, we focus on recent findings that establish SESN2 as an orchestrator of mitochondrial stress adaptation, which is supported by its involvement in the integrated stress response, mitochondrial biogenesis, and mitophagy. Additionally, we discuss the integral role of SESN2 in mediating the health benefits of exercise as well as its impact on skeletal muscle, liver and heart injury, and aging.
Collapse
Affiliation(s)
- Ivo F Machado
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CiBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute of Interdisciplinary Research, Doctoral Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | - Carlos M Palmeira
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CiBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Anabela P Rolo
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CiBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
3
|
Peng B, Wang Y, Zhang H. Mitonuclear Communication in Stem Cell Function. Cell Prolif 2025; 58:e13796. [PMID: 39726221 PMCID: PMC12099226 DOI: 10.1111/cpr.13796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024] Open
Abstract
Mitochondria perform multiple functions within the cell, including the production of ATP and a great deal of metabolic intermediates, while also contributing to the cellular stress response. The majority of mitochondrial proteins are encoded by nuclear genomes, highlighting the importance of mitonuclear communication for sustaining mitochondrial homeostasis and functional. As a crucial part of the intracellular signalling network, mitochondria can impact stem cell fate determinations. Considering the essential function of stem cells in tissue maintenance, regeneration and aging, it is important to understand how mitochondria influence stem cell fate. This review explores the significant roles of mitonuclear communication and mitochondrial proteostasis, highlighting their influence on stem cells. We also examine how mitonuclear interactions contribute to cellular homeostasis, stem cell therapies, and the potential for extending lifespan.
Collapse
Affiliation(s)
- Baozhou Peng
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- The Department of Histology and Embryology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Yaning Wang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- The Department of Histology and Embryology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Hongbo Zhang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- The Department of Histology and Embryology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
4
|
Ye D, Zhu J, Su S, Yu Y, Zhang J, Yin Y, Lin C, Xie X, Xiang Q, Yu R. Natural small molecules regulating the mitophagy pathway counteract the pathogenesis of diabetes and chronic complications. Front Pharmacol 2025; 16:1571767. [PMID: 40308774 PMCID: PMC12040946 DOI: 10.3389/fphar.2025.1571767] [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: 02/06/2025] [Accepted: 03/03/2025] [Indexed: 05/02/2025] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder marked by sustained hyperglycemia. These disturbances contribute to extensive damage across various tissues and organs, giving rise to severe complications such as vision loss, kidney failure, amputations, and higher morbidity and mortality rates. Furthermore, DM imposes a substantial economic and emotional burden on patients, families, and healthcare systems. Mitophagy, a selective process that targets the clearance of damaged or dysfunctional mitochondria, is pivotal for sustaining cellular homeostasis through mitochondrial turnover and recycling. Emerging evidence indicates that dysfunctional mitophagy acts as a key pathogenic driver in the pathogenesis of DM and its associated complications. Natural small molecules are particularly attractive in this regard, offering advantages such as low toxicity, favorable pharmacokinetic profiles, excellent biocompatibility, and a broad range of biochemical activities. This review systematically evaluates the mechanistic roles of natural small molecules-including ginsenosides, resveratrol, and berberine-in enhancing mitophagy and restoring mitochondrial homeostasis via activation of core signaling pathways (e.g., PINK1/Parkin, BNIP3/NIX, and FUNDC1). These pathways collectively ameliorate pathological hallmarks of DM, such as oxidative stress, chronic inflammation, and insulin resistance. Furthermore, the integration of nanotechnology with these compounds optimizes their bioavailability and tissue-specific targeting, thereby establishing a transformative therapeutic platform for DM management. Current evidence demonstrates that mitophagy modulation by natural small molecules not only offers novel therapeutic strategies for DM and its chronic complications but also advances the mechanistic foundation for future drug development targeting metabolic disorders.
Collapse
Affiliation(s)
- Du Ye
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Junping Zhu
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siya Su
- The Second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yunfeng Yu
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jun Zhang
- School of Informatics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yuman Yin
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chuanquan Lin
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xuejiao Xie
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qin Xiang
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Rong Yu
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
5
|
Giovarelli M, Zecchini S, Casati SR, Lociuro L, Gjana O, Mollica L, Pisanu E, Mbissam HD, Cappellari O, De Santis C, Arcari A, Bigot A, Clerici G, Catalani E, Del Quondam S, Andolfo A, Braccia C, Cattaneo MG, Banfi C, Brunetti D, Mocciaro E, De Luca A, Clementi E, Cervia D, Perrotta C, De Palma C. The SIRT1 activator SRT2104 exerts exercise mimetic effects and promotes Duchenne muscular dystrophy recovery. Cell Death Dis 2025; 16:259. [PMID: 40195304 PMCID: PMC11977210 DOI: 10.1038/s41419-025-07595-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/12/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating genetic disorder, whose management is still a major challenge, despite progress in genetic and pharmacological disease-modifying treatments have been made. Mitochondrial dysfunctions contribute to DMD, however, there are no effective mitochondrial therapies for DMD. SIRT1 is a NAD+-dependent deacetylase that controls several key processes and whose impairment is involved in determining mitochondrial dysfunction in DMD. In addition to well-known resveratrol, other potent selective activators of SIRT1 exist, with better pharmacokinetics properties and a safer profile. Among these, SRT2104 is the most promising and advanced in clinical studies. Here we unveil the beneficial effects of SRT2104 in flies, mice, and patient-derived myoblasts as different models of DMD, demonstrating an anti-inflammatory, anti-fibrotic, and pro-regenerative action of the drug. We elucidate, by molecular dynamics simulations, that a conformational selection mechanism is responsible for the activation of SIRT1. Further, the impact of SRT2104 in reshaping muscle proteome and acetylome profiles has been investigated, highlighting effects that mimic those induced by exercise. Overall, our data suggest SRT2104 as a possible therapeutic candidate to successfully counteract DMD progression.
Collapse
Affiliation(s)
- Matteo Giovarelli
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan, Italy
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan, Italy
| | - Silvia Rosanna Casati
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano; Segrate, Milan, Italy
| | - Laura Lociuro
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano; Segrate, Milan, Italy
| | - Oriola Gjana
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano; Segrate, Milan, Italy
| | - Luca Mollica
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano; Segrate, Milan, Italy
| | - Elena Pisanu
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano; Segrate, Milan, Italy
| | - Harcel Djaya Mbissam
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan, Italy
| | - Ornella Cappellari
- Department of Pharmacy - Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Chiara De Santis
- Department of Pharmacy - Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Alessandro Arcari
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan, Italy
| | - Anne Bigot
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | | | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Simona Del Quondam
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Annapaola Andolfo
- ProMeFa, Proteomics and Metabolomics Facility, Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Clarissa Braccia
- ProMeFa, Proteomics and Metabolomics Facility, Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Grazia Cattaneo
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano; Segrate, Milan, Italy
| | - Cristina Banfi
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Dario Brunetti
- Unità di Genetica Medica e Neurogenetica, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
- Dipartimento di Scienze Cliniche e di Comunità, Dipartimento di Eccellenza 2023-2027, Università degli Studi di Milano, Milan, Italy
| | - Emanuele Mocciaro
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan, Italy
| | - Annamaria De Luca
- Department of Pharmacy - Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences (DIBIC), Università degli Studi di Milano, Milan, Italy
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano; Segrate, Milan, Italy.
| |
Collapse
|
6
|
Chen L, Yu Y, Lin M, Liang M, Yang C, Kan X, Lin X, Qi J. Identification of components that increase NAD+ levels in oxygen-glucose deprived HUVEC s from Schisandra chinensis (Turcz.) Baill. Based on spectrum-effect correlation analysis and target cell extraction. Fitoterapia 2025; 181:106347. [PMID: 39701499 DOI: 10.1016/j.fitote.2024.106347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 12/05/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
Schisandra chinensis (Turcz.) Baill, a traditional Chinese medicine with significant nourishing functions, has a regulatory effect on the cardiovascular system, digestive system, central nervous system, and endocrine and immune systems. It can protect the cardiovascular system, improve immunity, and has anti-oxidant and anti-aging properties. This study aimed to identify the chisandra chinensis components that increased NAD+ levels, by using spectrum-effect analysis and experimental validations. First, the quality of the S. chinensis extract was analyzed by HPLC-MS. The extract of S. chinensis increased the NAD+ levels of HUVECs during oxygen and glucose deprivation injury, and protected HUVEC s from injury from superoxide dismutase (SOD) and aging. The relationships of spectral effects were studied by partial least square regression. Coupled with target cell extraction, the material basis for increasing NAD+ levels in S. chinensis was obtained. The pharmacological activity of S. chinensis was verified at the cellular level. The related enzymes of the NAD+ synthesis and decomposition pathways were identified, showing that S. chinensis increased the level of NAD+ by increasing the activity of related enzymes in the synthesis pathways of NMNAT1, NMNAT2, NMNAT3, and NAMPT, but had no effect on the decomposition pathway. Finally, four constituents were confirmed, in vitro, to be the basis of S. chinensis-induced increases in the levels of NAD+. The EC50 values of the four constituents were also determined.
Collapse
Affiliation(s)
- Liwenyu Chen
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yi Yu
- Infinitus (China) Company Limited, Guangzhou 510405, China
| | - Min Lin
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Ming Liang
- Infinitus (China) Company Limited, Guangzhou 510405, China
| | - Chen Yang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Xutian Kan
- Infinitus (China) Company Limited, Guangzhou 510405, China
| | - Xiaoliang Lin
- Infinitus (China) Company Limited, Guangzhou 510405, China
| | - Jin Qi
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
7
|
Liu YJ, Sulc J, Auwerx J. Mitochondrial genetics, signalling and stress responses. Nat Cell Biol 2025; 27:393-407. [PMID: 40065146 DOI: 10.1038/s41556-025-01625-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] [Received: 11/28/2023] [Accepted: 01/22/2025] [Indexed: 03/15/2025]
Abstract
Mitochondria are multifaceted organelles with crucial roles in energy generation, cellular signalling and a range of synthesis pathways. The study of mitochondrial biology is complicated by its own small genome, which is matrilineally inherited and not subject to recombination, and present in multiple, possibly different, copies. Recent methodological developments have enabled the analysis of mitochondrial DNA (mtDNA) in large-scale cohorts and highlight the far-reaching impact of mitochondrial genetic variation. Genome-editing techniques have been adapted to target mtDNA, further propelling the functional analysis of mitochondrial genes. Mitochondria are finely tuned signalling hubs, a concept that has been expanded by advances in methodologies for studying the function of mitochondrial proteins and protein complexes. Mitochondrial respiratory complexes are of dual genetic origin, requiring close coordination between mitochondrial and nuclear gene-expression systems (transcription and translation) for proper assembly and function, and recent findings highlight the importance of the mitochondria in this bidirectional signalling.
Collapse
Affiliation(s)
- Yasmine J Liu
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Jonathan Sulc
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
8
|
Fonseka O, Gare SR, Chen X, Zhang J, Alatawi NH, Ross C, Liu W. Molecular Mechanisms Underlying Heart Failure and Their Therapeutic Potential. Cells 2025; 14:324. [PMID: 40072053 PMCID: PMC11899429 DOI: 10.3390/cells14050324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Heart failure (HF) is a prominent fatal cardiovascular disorder afflicting 3.4% of the adult population despite the advancement of treatment options. Therefore, a better understanding of the pathogenesis of HF is essential for exploring novel therapeutic strategies. Hypertrophy and fibrosis are significant characteristics of pathological cardiac remodeling, contributing to HF. The mechanisms involved in the development of cardiac remodeling and consequent HF are multifactorial, and in this review, the key underlying mechanisms are discussed. These have been divided into the following categories thusly: (i) mitochondrial dysfunction, including defective dynamics, energy production, and oxidative stress; (ii) cardiac lipotoxicity; (iii) maladaptive endoplasmic reticulum (ER) stress; (iv) impaired autophagy; (v) cardiac inflammatory responses; (vi) programmed cell death, including apoptosis, pyroptosis, and ferroptosis; (vii) endothelial dysfunction; and (viii) defective cardiac contractility. Preclinical data suggest that there is merit in targeting the identified pathways; however, their clinical implications and outcomes regarding treating HF need further investigation in the future. Herein, we introduce the molecular mechanisms pivotal in the onset and progression of HF, as well as compounds targeting the related mechanisms and their therapeutic potential in preventing or rescuing HF. This, therefore, offers an avenue for the design and discovery of novel therapies for the treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (O.F.); (S.R.G.); (X.C.); (J.Z.); (N.H.A.)
| |
Collapse
|
9
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
10
|
Burtscher J, Denti V, Gostner JM, Weiss AK, Strasser B, Hüfner K, Burtscher M, Paglia G, Kopp M, Dünnwald T. The interplay of NAD and hypoxic stress and its relevance for ageing. Ageing Res Rev 2025; 104:102646. [PMID: 39710071 DOI: 10.1016/j.arr.2024.102646] [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: 08/12/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD) is an essential regulator of cellular metabolism and redox processes. NAD levels and the dynamics of NAD metabolism change with increasing age but can be modulated via the diet or medication. Because NAD metabolism is complex and its regulation still insufficiently understood, achieving specific outcomes without perturbing delicate balances through targeted pharmacological interventions remains challenging. NAD metabolism is also highly sensitive to environmental conditions and can be influenced behaviorally, e.g., by exercise. Changes in oxygen availability directly and indirectly affect NAD levels and may result from exposure to ambient hypoxia, increased oxygen demand during exercise, ageing or disease. Cellular responses to hypoxic stress involve rapid alterations in NAD metabolism and depend on many factors, including age, glucose status, the dose of the hypoxic stress and occurrence of reoxygenation phases, and exhibit complex time-courses. Here we summarize the known determinants of NAD-regulation by hypoxia and evaluate the role of NAD in hypoxic stress. We define the specific NAD responses to hypoxia and identify a great potential of the modulation of NAD metabolism regarding hypoxic injuries. In conclusion, NAD metabolism and cellular hypoxia responses are strongly intertwined and together mediate protective processes against hypoxic insults. Their interactions likely contribute to age-related changes and vulnerabilities. Targeting NAD homeostasis presents a promising avenue to prevent/treat hypoxic insults and - conversely - controlled hypoxia is a potential tool to regulate NAD homeostasis.
Collapse
Affiliation(s)
- Johannes Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria.
| | - Vanna Denti
- School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, MB, Italy
| | - Johanna M Gostner
- Medical University of Innsbruck, Biocenter, Institute of Medical Biochemistry, Innsbruck, Austria
| | - Alexander Kh Weiss
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Barbara Strasser
- Ludwig Boltzmann Institute for Rehabilitation Research, Vienna, Austria; Faculty of Medicine, Sigmund Freud Private University, Vienna, Austria
| | - Katharina Hüfner
- Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, University Hospital for Psychiatry II, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Giuseppe Paglia
- School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, MB, Italy
| | - Martin Kopp
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Tobias Dünnwald
- Institute for Sports Medicine, Alpine Medicine and Health Tourism (ISAG), UMIT TIROL - Private University for Health Sciences and Health Technology, Hall in Tirol, Austria
| |
Collapse
|
11
|
Chourasia S, Petucci C, Shoffler C, Abbasian D, Wang H, Han X, Sivan E, Brandis A, Mehlman T, Malitsky S, Itkin M, Sharp A, Rotkopf R, Dassa B, Regev L, Zaltsman Y, Gross A. MTCH2 controls energy demand and expenditure to fuel anabolism during adipogenesis. EMBO J 2025; 44:1007-1038. [PMID: 39753955 PMCID: PMC11832942 DOI: 10.1038/s44318-024-00335-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 02/19/2025] Open
Abstract
Mitochondrial carrier homolog 2 (MTCH2) is a regulator of apoptosis, mitochondrial dynamics, and metabolism. Loss of MTCH2 results in mitochondrial fragmentation, an increase in whole-body energy utilization, and protection against diet-induced obesity. In this study, we used temporal metabolomics on HeLa cells to show that MTCH2 deletion results in a high ATP demand, an oxidized cellular environment, and elevated utilization of lipids, amino acids, and carbohydrates, accompanied by a decrease in several metabolites. Lipidomics analysis revealed a strategic adaptive reduction in membrane lipids and an increase in storage lipids in MTCH2 knockout cells. Importantly, MTCH2 knockout cells showed an increase in mitochondrial oxidative function, which may explain the higher energy demand. Interestingly, this imbalance in energy metabolism and reductive potential triggered by MTCH2-deletion prevents NIH3T3L1 preadipocytes from differentiating into mature adipocytes, an energy consuming reductive biosynthetic process. In summary, the loss of MTCH2 leads to increased mitochondrial oxidative activity and energy demand, creating a catabolic and oxidative environment that fails to fuel the anabolic processes required for lipid accumulation and adipocyte differentiation.
Collapse
Affiliation(s)
- Sabita Chourasia
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 76100, Rehovot, Israel.
| | - Christopher Petucci
- Metabolomics Core, Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Clarissa Shoffler
- Metabolomics Core, Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dina Abbasian
- Metabolomics Core, Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hu Wang
- Barshop Institute for Longevity and Aging Studies, and Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, and Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Ehud Sivan
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Alexander Brandis
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Tevie Mehlman
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Sergey Malitsky
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Maxim Itkin
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Ayala Sharp
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Ron Rotkopf
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Bareket Dassa
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Limor Regev
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Yehudit Zaltsman
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Atan Gross
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 76100, Rehovot, Israel.
| |
Collapse
|
12
|
Zhang L, Zhou Y, Yang Z, Jiang L, Yan X, Zhu W, Shen Y, Wang B, Li J, Song J. Lipid droplets in central nervous system and functional profiles of brain cells containing lipid droplets in various diseases. J Neuroinflammation 2025; 22:7. [PMID: 39806503 PMCID: PMC11730833 DOI: 10.1186/s12974-025-03334-5] [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: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Lipid droplets (LDs), serving as the convergence point of energy metabolism and multiple signaling pathways, have garnered increasing attention in recent years. Different cell types within the central nervous system (CNS) can regulate energy metabolism to generate or degrade LDs in response to diverse pathological stimuli. This article provides a comprehensive review on the composition of LDs in CNS, their generation and degradation processes, their interaction mechanisms with mitochondria, the distribution among different cell types, and the roles played by these cells-particularly microglia and astrocytes-in various prevalent neurological disorders. Additionally, we also emphasize the paradoxical role of LDs in post-cerebral ischemia inflammation and explore potential underlying mechanisms, aiming to identify novel therapeutic targets for this disease.
Collapse
Affiliation(s)
- Longxiao Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yunfei Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Zhongbo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Liangchao Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xinyang Yan
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Wenkai Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yi Shen
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Bolong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jiaxi Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Jinning Song
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
13
|
Serrano J, Kondo S, Link GM, Brown IS, Pratley RE, Baskin KK, Goodpaster BH, Coen PM, Kyriazis GA. A partial loss-of-function variant (Ile191Val) of the TAS1R2 glucose receptor is associated with enhanced responses to exercise training in older adults with obesity: A translational study. Metabolism 2025; 162:156045. [PMID: 39393515 PMCID: PMC11637915 DOI: 10.1016/j.metabol.2024.156045] [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: 06/14/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND The TAS1R2 receptor, known for its role in taste perception, has also emerged as a key regulator of muscle physiology. Previous studies have shown that genetic ablation of TAS1R2 in mice enhances muscle fitness mimicking responses to endurance exercise training. However, the translational relevance of these findings to humans remains uncertain. METHODS We explored responses to endurance exercise training in mice and humans with genetic deficiency of TAS1R2. First, we assessed the effects of muscle-specific deletion of TAS1R2 in mice (mKO) or wild type controls (mWT) following 4 weeks of voluntary wheel running (VWR). Next, we investigated the effects of the TAS1R2-Ile191Val (rs35874116) partial loss-of-function variant on responses to a 6-month diet-induced weight loss with exercise training (WLEX), weight loss alone (WL), or education control (CON) interventions in older individuals with obesity. Participants were retrospectively genotyped for the TAS1R2-Ile191Val polymorphism and classified as conventional function (Ile/Ile) or partial loss-of-function (Val carriers: Ile/Val and Val/Val). Body composition, cardiorespiratory fitness, and skeletal muscle mitochondrial function were assessed before and after the intervention. RESULTS In response to VWR, mKO mice demonstrated enhanced running endurance and mitochondrial protein content. Similarly, TAS1R2 Val carriers exhibited distinctive improvements in body composition, including increased muscle mass, along with enhanced cardiorespiratory fitness and mitochondrial function in skeletal muscle following the WLEX intervention compared to Ile/Ile counterparts. Notably, every Val carrier demonstrated substantial responses to exercise training and weight loss, surpassing all Ile/Ile participants in overall performance metrics. CONCLUSIONS Our findings suggest that TAS1R2 partial loss-of-function confers beneficial effects on muscle function and metabolism in humans in response to exercise training, akin to observations in TAS1R2 muscle-deficient mice. Targeting TAS1R2 may help enhancing exercise training adaptations in individuals with compromised exercise tolerance or metabolic disorders, presenting a potential avenue for personalized exercise interventions.
Collapse
Affiliation(s)
- Joan Serrano
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Saki Kondo
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Grace M Link
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Ian S Brown
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Kedryn K Baskin
- Physiology & Cell Biology College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Paul M Coen
- Translational Research Institute, Advent Health, Orlando, FL, USA.
| | - George A Kyriazis
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
14
|
Bo T, Fujii J. Primary Roles of Branched Chain Amino Acids (BCAAs) and Their Metabolism in Physiology and Metabolic Disorders. Molecules 2024; 30:56. [PMID: 39795113 PMCID: PMC11721030 DOI: 10.3390/molecules30010056] [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: 11/19/2024] [Revised: 12/25/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
Leucine, isoleucine, and valine are collectively known as branched chain amino acids (BCAAs) and are often discussed in the same physiological and pathological situations. The two consecutive initial reactions of BCAA catabolism are catalyzed by the common enzymes referred to as branched chain aminotransferase (BCAT) and branched chain α-keto acid dehydrogenase (BCKDH). BCAT transfers the amino group of BCAAs to 2-ketoglutarate, which results in corresponding branched chain 2-keto acids (BCKAs) and glutamate. BCKDH performs an oxidative decarboxylation of BCKAs, which produces their coenzyme A-conjugates and NADH. BCAT2 in skeletal muscle dominantly catalyzes the transamination of BCAAs. Low BCAT activity in the liver reduces the metabolization of BCAAs, but the abundant presence of BCKDH promotes the metabolism of muscle-derived BCKAs, which leads to the production of glucose and ketone bodies. While mutations in the genes responsible for BCAA catabolism are involved in rare inherited disorders, an aberrant regulation of their enzymatic activities is associated with major metabolic disorders such as diabetes, cardiovascular disease, and cancer. Therefore, an understanding of the regulatory process of metabolic enzymes, as well as the functions of the BCAAs and their metabolites, make a significant contribution to our health.
Collapse
Affiliation(s)
- Tomoki Bo
- Laboratory Animal Center, Institute for Promotion of Medical Science Research, Faculty of Medicine, Yamagata University, Yamagata 990-9585, Japan
| | - Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| |
Collapse
|
15
|
Samant V, Prabhu A. Exercise, exerkines and exercise mimetic drugs: Molecular mechanisms and therapeutics. Life Sci 2024; 359:123225. [PMID: 39522716 DOI: 10.1016/j.lfs.2024.123225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/09/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Chronic diseases linked with sedentary lifestyles and poor dietary habits are increasingly common in modern society. Exercise is widely acknowledged to have a plethora of health benefits, including its role in primary prevention of various chronic conditions like type 2 diabetes mellitus, obesity, cardiovascular disease, and several musculoskeletal as well as degenerative disorders. Regular physical activity induces numerous physiological adaptations that contribute to these positive effects, primarily observed in skeletal muscle but also impacting other tissues. There is a growing interest among researchers in developing pharmaceutical interventions that mimic the beneficial effects of exercise for therapeutic applications. Exercise mimetic medications have the potential to be helpful aids in enhancing functional outcomes for patients with metabolic dysfunction, neuromuscular and musculoskeletal disorders. Some of the potential targets for exercise mimetics include pathways involved in metabolism, mitochondrial function, inflammation, and tissue regeneration. The present review aims to provide an exhaustive overview of the current understanding of exercise physiology, the role of exerkines and biomolecular pathways, and the potential applications of exercise mimetic drugs for the treatment of several diseases.
Collapse
Affiliation(s)
- Vedant Samant
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Arati Prabhu
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India.
| |
Collapse
|
16
|
Bonato A, Raparelli G, Caruso M. Molecular pathways involved in the control of contractile and metabolic properties of skeletal muscle fibers as potential therapeutic targets for Duchenne muscular dystrophy. Front Physiol 2024; 15:1496870. [PMID: 39717824 PMCID: PMC11663947 DOI: 10.3389/fphys.2024.1496870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the gene encoding dystrophin, a subsarcolemmal protein whose absence results in increased susceptibility of the muscle fiber membrane to contraction-induced injury. This results in increased calcium influx, oxidative stress, and mitochondrial dysfunction, leading to chronic inflammation, myofiber degeneration, and reduced muscle regenerative capacity. Fast glycolytic muscle fibers have been shown to be more vulnerable to mechanical stress than slow oxidative fibers in both DMD patients and DMD mouse models. Therefore, remodeling skeletal muscle toward a slower, more oxidative phenotype may represent a relevant therapeutic approach to protect dystrophic muscles from deterioration and improve the effectiveness of gene and cell-based therapies. The resistance of slow, oxidative myofibers to DMD pathology is attributed, in part, to their higher expression of Utrophin; there are, however, other characteristics of slow, oxidative fibers that might contribute to their enhanced resistance to injury, including reduced contractile speed, resistance to fatigue, increased capillary density, higher mitochondrial activity, decreased cellular energy requirements. This review focuses on signaling pathways and regulatory factors whose genetic or pharmacologic modulation has been shown to ameliorate the dystrophic pathology in preclinical models of DMD while promoting skeletal muscle fiber transition towards a slower more oxidative phenotype.
Collapse
Affiliation(s)
| | | | - Maurizia Caruso
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), Monterotondo (RM), Italy
| |
Collapse
|
17
|
Ye F, Wei C, Wu A. The potential mechanism of mitochondrial homeostasis in postoperative neurocognitive disorders: an in-depth review. Ann Med 2024; 56:2411012. [PMID: 39450938 PMCID: PMC11514427 DOI: 10.1080/07853890.2024.2411012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 10/26/2024] Open
Abstract
Postoperative neurocognitive disorders (PND) are the most common neurological disorders following surgery and anaesthesia before and within 12 months after surgery, with a high prevalence in the geriatric population. PND can severely deteriorate the quality of life of patients, especially among the elderly, mainly manifested as memory loss, attention, decline and language comprehension disorders, mostly in elderly patients, with an incidence as high as 31%. Previous studies have also raised the possibility of accelerated cognitive decline and underlying neuropathological processes associated with diseases that affect cognitive performance (e.g. Alzheimer's dementia) for reasons related to anaesthesia and surgery. Currently, most research on PND has focused on various molecular pathways, especially in the geriatric population. The various hypotheses that have been proposed regarding the mechanisms imply peripheral neuroinflammation, oxidative stress, mitochondrial homeostasis, synaptic function, autophagy disorder, blood-brain barrier dysfunction, the microbiota-gut-brain axis and lack of neurotrophic support. However, the underlying pathogenesis and molecular mechanisms of PND have not yet been uncovered. Recent research has focused on mitochondrial homeostasis. In this paper, we present a review of various studies to better understand and characterize the mechanisms of associated cognitive dysfunction. As the biochemical basis of PND becomes more clearly defined, future treatments based on mitochondrial homeostasis modulation can prove to be very promising.
Collapse
Affiliation(s)
- Fan Ye
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
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
|
19
|
Hunter‐Manseau F, Cormier SB, Strang R, Pichaud N. Fasting as a precursor to high-fat diet enhances mitochondrial resilience in Drosophila melanogaster. INSECT SCIENCE 2024; 31:1770-1788. [PMID: 38514255 PMCID: PMC11632299 DOI: 10.1111/1744-7917.13355] [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: 12/13/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 03/23/2024]
Abstract
Changes in diet type and nutrient availability can impose significant environmental stress on organisms, potentially compromising physiological functions and reproductive success. In nature, dramatic fluctuations in dietary resources are often observed and adjustments to restore cellular homeostasis are crucial to survive this type of stress. In this study, we exposed male Drosophila melanogaster to two modulated dietary treatments: one without a fasting period before exposure to a high-fat diet and the other with a 24-h fasting period. We then investigated mitochondrial metabolism and molecular responses to these treatments. Exposure to a high-fat diet without a preceding fasting period resulted in disrupted mitochondrial respiration, notably at the level of complex I. On the other hand, a short fasting period before the high-fat diet maintained mitochondrial respiration. Generally, transcript abundance of genes associated with mitophagy, heat-shock proteins, mitochondrial biogenesis, and nutrient sensing pathways increased either slightly or significantly following a fasting period and remained stable when flies were subsequently put on a high-fat diet, whereas a drastic decrease of almost all transcript abundances was observed for all these pathways when flies were exposed directly to a high-fat diet. Moreover, mitochondrial enzymatic activities showed less variation after the fasting period than the treatment without a fasting period. Overall, our study sheds light on the mechanistic protective effects of fasting prior to a high-fat diet and highlights the metabolic flexibility of Drosophila mitochondria in response to abrupt dietary changes and have implication for adaptation of species to their changing environment.
Collapse
Affiliation(s)
- Florence Hunter‐Manseau
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNew BrunswickCanada
- New Brunswick Centre for Precision MedicineMonctonNew BrunswickCanada
| | - Simon B. Cormier
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNew BrunswickCanada
- New Brunswick Centre for Precision MedicineMonctonNew BrunswickCanada
| | - Rebekah Strang
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNew BrunswickCanada
- New Brunswick Centre for Precision MedicineMonctonNew BrunswickCanada
| | - Nicolas Pichaud
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNew BrunswickCanada
- New Brunswick Centre for Precision MedicineMonctonNew BrunswickCanada
| |
Collapse
|
20
|
Che X, Oh JH, Kang YJ, Kim DW, Kim SG, Choi JY, Garagiola U. 4-Hexylresorcinol Enhances Glut4 Expression and Glucose Homeostasis via AMPK Activation and Histone H3 Acetylation. Int J Mol Sci 2024; 25:12281. [PMID: 39596347 PMCID: PMC11594624 DOI: 10.3390/ijms252212281] [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: 10/05/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
This study investigates the potential of 4-hexylresorcinol (4HR) as a novel antidiabetic agent by assessing its effects on blood glucose levels, Glut4 expression, AMPK phosphorylation, and Histone H3 acetylation (Ac-H3) in the liver. In vitro experiments utilized Huh7 and HepG2 cells treated with varying concentrations of 4HR. Glut4, p-AMPK, and Ac-H3 expression levels were quantified via Western blotting. Additionally, GAPDH activity and glucose uptake were evaluated. In vivo experiments employed streptozotocin (STZ)-induced diabetic rats, with or without 4HR treatment, monitoring blood glucose, body weight, and hepatic levels of Glut4, p-AMPK, and Ac-H3. In vitro, 4HR treatment increased GAPDH activity and glucose uptake. Elevated Glut4, p-AMPK, and Ac-H3 levels were observed 8 h after 4HR administration. Inhibition of p-AMPK using compound C reduced 4HR-mediated Glut4 expression. In STZ-induced diabetic rats, 4HR significantly upregulated Glut4, p-AMPK, and Ac-H3 expression in the liver. Periodic 4HR injections mitigated weight loss and lowered blood glucose levels in STZ-injected animals. Histological analysis revealed increased glycogen storage in hepatocytes of the 4HR-treated group. Overall, 4HR enhanced Glut4 expression through upregulation of AMPK activity and histone H3 acetylation in vitro and in vivo, improving hepatic glucose homeostasis and suggesting potential as a candidate for diabetes treatment.
Collapse
Affiliation(s)
- Xiangguo Che
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Ji-Hyeon Oh
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea; (J.-H.O.); (Y.-J.K.)
| | - Yei-Jin Kang
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea; (J.-H.O.); (Y.-J.K.)
| | - Dae-Won Kim
- Department of Oral Biochemistry, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea;
| | - Seong-Gon Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea; (J.-H.O.); (Y.-J.K.)
| | - Je-Yong Choi
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Umberto Garagiola
- Maxillofacial and Dental Unit, Biomedical, Surgical and Oral Sciences Department, School of Dentistry, University of Milan, 20122 Milan, Italy;
| |
Collapse
|
21
|
Singh I, Anand S, Gowda DJ, Kamath A, Singh AK. Caloric restriction mimetics improve gut microbiota: a promising neurotherapeutics approach for managing age-related neurodegenerative disorders. Biogerontology 2024; 25:899-922. [PMID: 39177917 PMCID: PMC11486790 DOI: 10.1007/s10522-024-10128-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024]
Abstract
The gut microbiota (GM) produces various molecules that regulate the physiological functionality of the brain through the gut-brain axis (GBA). Studies suggest that alteration in GBA may lead to the onset and progression of various neurological dysfunctions. Moreover, aging is one of the prominent causes that contribute to the alteration of GBA. With age, GM undergoes a shift in population size and species of microflora leading to changes in their secreted metabolites. These changes also hamper communications among the HPA (hypothalamic-pituitary-adrenal), ENS (enteric nervous system), and ANS (autonomic nervous system). A therapeutic intervention that has recently gained attention in improving health and maintaining communication between the gut and the brain is calorie restriction (CR), which also plays a critical role in autophagy and neurogenesis processes. However, its strict regime and lifelong commitment pose challenges. The need is to produce similar beneficial effects of CR without having its rigorous compliance. This led to an exploration of calorie restriction mimetics (CRMs) which could mimic CR's functions without limiting diet, providing long-term health benefits. CRMs ensure the efficient functioning of the GBA through gut bacteria and their metabolites i.e., short-chain fatty acids, bile acids, and neurotransmitters. This is particularly beneficial for elderly individuals, as the GM deteriorates with age and the body's ability to digest the toxic accumulates declines. In this review, we have explored the beneficial effect of CRMs in extending lifespan by enhancing the beneficial bacteria and their effects on metabolite production, physiological conditions, and neurological dysfunctions including neurodegenerative disorders.
Collapse
Affiliation(s)
- Ishika Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Shashi Anand
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Deepashree J Gowda
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Amitha Kamath
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India.
| |
Collapse
|
22
|
Aragón-Vela J, Casuso RA, Aparisi AS, Plaza-Díaz J, Rueda-Robles A, Hidalgo-Gutiérrez A, López LC, Rodríguez-Carrillo A, Enriquez JA, Cogliati S, Huertas JR. Early heart and skeletal muscle mitochondrial response to a moderate hypobaric hypoxia environment. J Physiol 2024; 602:5631-5641. [PMID: 38630964 DOI: 10.1113/jp285516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
In eukaryotic cells, aerobic energy is produced by mitochondria through oxygen uptake. However, little is known about the early mitochondrial responses to moderate hypobaric hypoxia (MHH) in highly metabolic active tissues. Here, we describe the mitochondrial responses to acute MHH in the heart and skeletal muscle. Rats were randomly allocated into a normoxia control group (n = 10) and a hypoxia group (n = 30), divided into three groups (0, 6, and 24 h post-MHH). The normoxia situation was recapitulated at the University of Granada, at 662 m above sea level. The MHH situation was performed at the High-Performance Altitude Training Centre of Sierra Nevada located in Granada at 2320 m above sea level. We found a significant increase in mitochondrial supercomplex assembly in the heart as soon as the animals reached 2320 m above sea level and their levels are maintained 24 h post-exposure, but not in skeletal muscle. Furthermore, in skeletal muscle, at 0 and 6 h, there was increased dynamin-related protein 1 (Drp1) expression and a significant reduction in Mitofusin 2. In conclusion, mitochondria from the muscle and heart respond differently to MHH: mitochondrial supercomplexes increase in the heart, whereas, in skeletal muscle, the mitochondrial pro-fission response is trigged. Considering that skeletal muscle was not actively involved in the ascent when the heart was beating faster to compensate for the hypobaric, hypoxic conditions, we speculate that the different responses to MHH are a result of the different energetic requirements of the tissues upon MHH. KEY POINTS: The heart and the skeletal muscle showed different mitochondrial responses to moderate hypobaric hypoxia. Moderate hypobaric hypoxia increases the assembly of the electron transport chain complexes into supercomplexes in the heart. Skeletal muscle shows an early mitochondrial pro-fission response following exposure to moderate hypobaric hypoxia.
Collapse
Affiliation(s)
- Jerónimo Aragón-Vela
- Department of Health Sciences, Area of Physiology, University of Jaen, Jaen, Spain
| | - Rafael A Casuso
- Department of Health Sciences, Universidad Loyola Andalucía, Sevilla, Spain
| | - Ana Sagrera Aparisi
- Centro de Biologia Molecular Severo Ochoa (CBM), CSIC-UAM, Madrid, Spain
- Institute for Molecular Biology-IUBM (Universidad Autónoma de Madrid), Madrid, Spain
| | - Julio Plaza-Díaz
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada., Ottawa, ON, Canada
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Complejo Hospitalario Universitario de Granada, Granada, Spain
| | - Ascensión Rueda-Robles
- Institute of Nutrition and Food Technology 'José Mataix,' Biomedical Research Centre, Department of Physiology, Faculty of Sport Sciences, University of Granada, Granada, Spain
| | - Agustín Hidalgo-Gutiérrez
- Institute of Biotechnology, Biomedical Research Centre and Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Luis Carlos López
- Institute of Biotechnology, Biomedical Research Centre and Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Andrea Rodríguez-Carrillo
- Center for Biomedical Research (CIBM), University of Granada, Spain
- Department of Radiology and Physical Medicine, School of Medicine, University of Granada, Granada, Spain
| | - José Antonio Enriquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES)., Madrid, Spain
| | - Sara Cogliati
- Centro de Biologia Molecular Severo Ochoa (CBM), CSIC-UAM, Madrid, Spain
- Institute for Molecular Biology-IUBM (Universidad Autónoma de Madrid), Madrid, Spain
| | - Jesús R Huertas
- Institute of Nutrition and Food Technology 'José Mataix,' Biomedical Research Centre, Department of Physiology, Faculty of Sport Sciences, University of Granada, Granada, Spain
| |
Collapse
|
23
|
O’Brien J, Niehaus P, Chang K, Remark J, Barrett J, Dasgupta A, Adenegan M, Salimian M, Kevas Y, Chandrasekaran K, Kristian T, Chellappan R, Rubin S, Kiemen A, Lu CPJ, Russell JW, Ho CY. Skin keratinocyte-derived SIRT1 and BDNF modulate mechanical allodynia in mouse models of diabetic neuropathy. Brain 2024; 147:3471-3486. [PMID: 38554393 PMCID: PMC11449144 DOI: 10.1093/brain/awae100] [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: 10/05/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/01/2024] Open
Abstract
Diabetic neuropathy is a debilitating disorder characterized by spontaneous and mechanical allodynia. The role of skin mechanoreceptors in the development of mechanical allodynia is unclear. We discovered that mice with diabetic neuropathy had decreased sirtuin 1 (SIRT1) deacetylase activity in foot skin, leading to reduced expression of brain-derived neurotrophic factor (BDNF) and subsequent loss of innervation in Meissner corpuscles, a mechanoreceptor expressing the BDNF receptor TrkB. When SIRT1 was depleted from skin, the mechanical allodynia worsened in diabetic neuropathy mice, likely due to retrograde degeneration of the Meissner-corpuscle innervating Aβ axons and aberrant formation of Meissner corpuscles which may have increased the mechanosensitivity. The same phenomenon was also noted in skin-keratinocyte specific BDNF knockout mice. Furthermore, overexpression of SIRT1 in skin induced Meissner corpuscle reinnervation and regeneration, resulting in significant improvement of diabetic mechanical allodynia. Overall, the findings suggested that skin-derived SIRT1 and BDNF function in the same pathway in skin sensory apparatus regeneration and highlighted the potential of developing topical SIRT1-activating compounds as a novel treatment for diabetic mechanical allodynia.
Collapse
Affiliation(s)
- Jennifer O’Brien
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Peter Niehaus
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Koping Chang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, National Taiwan University, Taipei, 100, Taiwan
| | - Juliana Remark
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Joy Barrett
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Abhishikta Dasgupta
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Morayo Adenegan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Mohammad Salimian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yanni Kevas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Tibor Kristian
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21021, USA
| | - Rajeshwari Chellappan
- Department of Pathology, University of Alabama Birmingham, Birmingham, AL 35233, USA
| | - Samuel Rubin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Chemistry, College of William and Mary, Williamsburg, VA 23187, USA
| | - Ashley Kiemen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Catherine Pei-Ju Lu
- Hansjörg Wyss Department of Plastic Surgery, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - James W Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Cheng-Ying Ho
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
24
|
Thomas ACQ, Stead CA, Burniston JG, Phillips SM. Exercise-specific adaptations in human skeletal muscle: Molecular mechanisms of making muscles fit and mighty. Free Radic Biol Med 2024; 223:341-356. [PMID: 39147070 DOI: 10.1016/j.freeradbiomed.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
The mechanisms leading to a predominantly hypertrophied phenotype versus a predominantly oxidative phenotype, the hallmarks of resistance training (RT) or aerobic training (AT), respectively, are being unraveled. In humans, exposure of naïve persons to either AT or RT results in their skeletal muscle exhibiting generic 'exercise stress-related' signaling, transcription, and translation responses. However, with increasing engagement in AT or RT, the responses become refined, and the phenotype typically associated with each form of exercise emerges. Here, we review some of the mechanisms underpinning the adaptations of how muscles become, through AT, 'fit' and RT, 'mighty.' Much of our understanding of molecular exercise physiology has arisen from targeted analysis of post-translational modifications and measures of protein synthesis. Phosphorylation of specific residue sites has been a dominant focus, with canonical signaling pathways (AMPK and mTOR) studied extensively in the context of AT and RT, respectively. These alone, along with protein synthesis, have only begun to elucidate key differences in AT and RT signaling. Still, key yet uncharacterized differences exist in signaling and regulation of protein synthesis that drive unique adaptation to AT and RT. Omic studies are required to better understand the divergent relationship between exercise and phenotypic outcomes of training.
Collapse
Affiliation(s)
- Aaron C Q Thomas
- Protein Metabolism Research Lab, Department of Kinesiology, McMaster University, Hamilton, ON, Canada; Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Connor A Stead
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Jatin G Burniston
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Stuart M Phillips
- Protein Metabolism Research Lab, Department of Kinesiology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
25
|
Ha SE, Singh R, Jin B, Baek G, Jorgensen BG, Zogg H, Debnath S, Park HS, Cho H, Watkins CM, Cho S, Kim MS, Lee MY, Yu TY, Jeong JW, Ro S. miR-10a/b-5p-NCOR2 Regulates Insulin-Resistant Diabetes in Female Mice. Int J Mol Sci 2024; 25:10147. [PMID: 39337631 PMCID: PMC11432729 DOI: 10.3390/ijms251810147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Gender and biological sex have distinct impacts on the pathogenesis of type 2 diabetes (T2D). Estrogen deficiency is known to predispose female mice to T2D. In our previous study, we found that a high-fat, high-sucrose diet (HFHSD) induces T2D in male mice through the miR-10b-5p/KLF11/KIT pathway, but not in females, highlighting hormonal disparities in T2D susceptibility. However, the underlying molecular mechanisms of this hormonal protection in females remain elusive. To address this knowledge gap, we utilized ovariectomized, estrogen-deficient female mice, fed them a HFHSD to induce T2D, and investigated the molecular mechanisms involved in estrogen-deficient diabetic female mice, relevant cell lines, and female T2D patients. Initially, female mice fed a HFHSD exhibited a delayed onset of T2D, but ovariectomy-induced estrogen deficiency promptly precipitated T2D without delay. Intriguingly, insulin (INS) was upregulated, while insulin receptor (INSR) and protein kinase B (AKT) were downregulated in these estrogen-deficient diabetic female mice, indicating insulin-resistant T2D. These dysregulations of INS, INSR, and AKT were mediated by a miR-10a/b-5p-NCOR2 axis. Treatment with miR-10a/b-5p effectively alleviated hyperglycemia in estrogen-deficient T2D female mice, while β-estradiol temporarily reduced hyperglycemia. Consistent with the murine findings, plasma samples from female T2D patients exhibited significant reductions in miR-10a/b-5p, estrogen, and INSR, but increased insulin levels. Our findings suggest that estrogen protects against insulin-resistant T2D in females through miR-10a/b-5p/NCOR2 pathway, indicating the potential therapeutic benefits of miR-10a/b-5p restoration in female T2D management.
Collapse
Affiliation(s)
- Se Eun Ha
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Rajan Singh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Byungchang Jin
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Gain Baek
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Brian G. Jorgensen
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Hannah Zogg
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Sushmita Debnath
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Hahn Sung Park
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Hayeong Cho
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Claudia Marie Watkins
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Sumin Cho
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
| | - Min-Seob Kim
- Department of Physiology, Wonkwang Digestive Disease Research Institute & Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (M.-S.K.); (M.Y.L.)
| | - Moon Young Lee
- Department of Physiology, Wonkwang Digestive Disease Research Institute & Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Republic of Korea; (M.-S.K.); (M.Y.L.)
| | - Tae Yang Yu
- Division of Endocrinology and Metabolism, Department of Medicine, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea; (T.Y.Y.); (J.W.J.)
| | - Jin Woo Jeong
- Division of Endocrinology and Metabolism, Department of Medicine, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea; (T.Y.Y.); (J.W.J.)
| | - Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.J.); (G.B.); (H.Z.); (H.S.P.); (S.C.)
- RosVivo Therapeutics, Applied Research Facility, 1664 N. Virginia St., Reno, NV 89557, USA
| |
Collapse
|
26
|
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
|
27
|
Mirza Z, Karim S. Unraveling the Mystery of Energy-Sensing Enzymes and Signaling Pathways in Tumorigenesis and Their Potential as Therapeutic Targets for Cancer. Cells 2024; 13:1474. [PMID: 39273044 PMCID: PMC11394487 DOI: 10.3390/cells13171474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 09/15/2024] Open
Abstract
Cancer research has advanced tremendously with the identification of causative genes, proteins, and signaling pathways. Numerous antitumor drugs have been designed and screened for cancer therapeutics; however, designing target-specific drugs for malignant cells with minimal side effects is challenging. Recently, energy-sensing- and homeostasis-associated molecules and signaling pathways playing a role in proliferation, apoptosis, autophagy, and angiogenesis have received increasing attention. Energy-metabolism-based studies have shown the contribution of energetics to cancer development, where tumor cells show increased glycolytic activity and decreased oxidative phosphorylation (the Warburg effect) in order to obtain the required additional energy for rapid division. The role of energy homeostasis in the survival of normal as well as malignant cells is critical; therefore, fuel intake and expenditure must be balanced within acceptable limits. Thus, energy-sensing enzymes detecting the disruption of glycolysis, AMP, ATP, or GTP levels are promising anticancer therapeutic targets. Here, we review the common energy mediators and energy sensors and their metabolic properties, mechanisms, and associated signaling pathways involved in carcinogenesis, and explore the possibility of identifying drugs for inhibiting the energy metabolism of tumor cells. Furthermore, to corroborate our hypothesis, we performed meta-analysis based on transcriptomic profiling to search for energy-associated biomarkers and canonical pathways.
Collapse
Affiliation(s)
- Zeenat Mirza
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21587, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21587, Saudi Arabia;
| | - Sajjad Karim
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21587, Saudi Arabia;
- Center of Excellence in Genomic Medicine Research, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21587, Saudi Arabia
| |
Collapse
|
28
|
Al‐Ibraheem AMT, Hameed AAZ, Marsool MDM, Jain H, Prajjwal P, Khazmi I, Nazzal RS, AL‐Najati HMH, Al‐Zuhairi BHYK, Razzaq M, Abd ZB, Marsool ADM, wahedaldin AI, Amir O. Exercise-Induced cytokines, diet, and inflammation and their role in adipose tissue metabolism. Health Sci Rep 2024; 7:e70034. [PMID: 39221051 PMCID: PMC11365580 DOI: 10.1002/hsr2.70034] [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: 11/12/2023] [Revised: 04/23/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Obesity poses a significant global health challenge, necessitating effective prevention and treatment strategies. Exercise and diet are recognized as pivotal interventions in combating obesity. This study reviews the literature concerning the impact of exercise-induced cytokines, dietary factors, and inflammation on adipose tissue metabolism, shedding light on potential pathways for therapeutic intervention. METHODOLOGY A comprehensive review of relevant literature was conducted to elucidate the role of exercise-induced cytokines, including interleukin-6 (IL-6), interleukin-15 (IL-15), brain-derived neurotrophic factor (BDNF), irisin, myostatin, fibroblast growth factor 21 (FGF21), follistatin (FST), and angiopoietin-like 4 (ANGPTL4), in adipose tissue metabolism. Various databases were systematically searched using predefined search terms to identify relevant studies. Articles selected for inclusion underwent thorough analysis to extract pertinent data on the mechanisms underlying the influence of these cytokines on adipose tissue metabolism. RESULTS AND DISCUSSION Exercise-induced cytokines exert profound effects on adipose tissue metabolism, influencing energy expenditure (EE), thermogenesis, fat loss, and adipogenesis. For instance, IL-6 activates AMP-activated protein kinase (AMPK), promoting fatty acid oxidation and reducing lipogenesis. IL-15 upregulates peroxisome proliferator-activated receptor delta (PPARδ), stimulating fatty acid catabolism and suppressing lipogenesis. BDNF enhances AMPK-dependent fat oxidation, while irisin induces the browning of white adipose tissue (WAT), augmenting thermogenesis. Moreover, myostatin, FGF21, FST, and ANGPTL4 each play distinct roles in modulating adipose tissue metabolism, impacting factors such as fatty acid oxidation, adipogenesis, and lipid uptake. The elucidation of these pathways offers valuable insights into the complex interplay between exercise, cytokines, and adipose tissue metabolism, thereby informing the development of targeted obesity management strategies. CONCLUSION Understanding the mechanisms by which exercise-induced cytokines regulate adipose tissue metabolism is critical for devising effective obesity prevention and treatment modalities. Harnessing the therapeutic potential of exercise-induced cytokines, in conjunction with dietary interventions, holds promise for mitigating the global burden of obesity. Further research is warranted to delineate the precise mechanisms underlying the interactions between exercise, cytokines, and adipose tissue metabolism.
Collapse
Affiliation(s)
| | | | | | - Hritvik Jain
- All India Institute of Medical SciencesJodhpurIndia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Gao Y, Kim K, Vitrac H, Salazar RL, Gould BD, Soedkamp D, Spivia W, Raedschelders K, Dinh AQ, Guzman AG, Tan L, Azinas S, Taylor DJR, Schiffer W, McNavish D, Burks HB, Gottlieb RA, Lorenzi PL, Hanson BM, Van Eyk JE, Taegtmeyer H, Karlstaedt A. Autophagic signaling promotes systems-wide remodeling in skeletal muscle upon oncometabolic stress by D2-HG. Mol Metab 2024; 86:101969. [PMID: 38908793 PMCID: PMC11278897 DOI: 10.1016/j.molmet.2024.101969] [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: 05/20/2024] [Accepted: 06/11/2024] [Indexed: 06/24/2024] Open
Abstract
OBJECTIVES Cachexia is a metabolic disorder and comorbidity with cancer and heart failure. The syndrome impacts more than thirty million people worldwide, accounting for 20% of all cancer deaths. In acute myeloid leukemia, somatic mutations of the metabolic enzyme isocitrate dehydrogenase 1 and 2 cause the production of the oncometabolite D2-hydroxyglutarate (D2-HG). Increased production of D2-HG is associated with heart and skeletal muscle atrophy, but the mechanistic links between metabolic and proteomic remodeling remain poorly understood. Therefore, we assessed how oncometabolic stress by D2-HG activates autophagy and drives skeletal muscle loss. METHODS We quantified genomic, metabolomic, and proteomic changes in cultured skeletal muscle cells and mouse models of IDH-mutant leukemia using RNA sequencing, mass spectrometry, and computational modeling. RESULTS D2-HG impairs NADH redox homeostasis in myotubes. Increased NAD+ levels drive activation of nuclear deacetylase Sirt1, which causes deacetylation and activation of LC3, a key regulator of autophagy. Using LC3 mutants, we confirm that deacetylation of LC3 by Sirt1 shifts its distribution from the nucleus into the cytosol, where it can undergo lipidation at pre-autophagic membranes. Sirt1 silencing or p300 overexpression attenuated autophagy activation in myotubes. In vivo, we identified increased muscle atrophy and reduced grip strength in response to D2-HG in male vs. female mice. In male mice, glycolytic intermediates accumulated, and protein expression of oxidative phosphorylation machinery was reduced. In contrast, female animals upregulated the same proteins, attenuating the phenotype in vivo. Network modeling and machine learning algorithms allowed us to identify candidate proteins essential for regulating oncometabolic adaptation in mouse skeletal muscle. CONCLUSIONS Our multi-omics approach exposes new metabolic vulnerabilities in response to D2-HG in skeletal muscle and provides a conceptual framework for identifying therapeutic targets in cachexia.
Collapse
Affiliation(s)
- Yaqi Gao
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kyoungmin Kim
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Heidi Vitrac
- Department of Biochemistry, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Bruker Daltonics, Billerica, MA, USA
| | - Rebecca L Salazar
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Benjamin D Gould
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Daniel Soedkamp
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Weston Spivia
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Koen Raedschelders
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - An Q Dinh
- Center for Infectious Diseases, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Anna G Guzman
- Center for Stem Cell and Regeneration, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Stavros Azinas
- Department of Biochemistry, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Cell and Molecular Biology, Uppsala University, Sweden
| | - David J R Taylor
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Walter Schiffer
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Daniel McNavish
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Helen B Burks
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Roberta A Gottlieb
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Blake M Hanson
- Center for Infectious Diseases, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Heinrich Taegtmeyer
- Department of Biochemistry, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
30
|
Russo C, Santangelo R, Malaguarnera L, Valle MS. The "Sunshine Vitamin" and Its Antioxidant Benefits for Enhancing Muscle Function. Nutrients 2024; 16:2195. [PMID: 39064638 PMCID: PMC11279438 DOI: 10.3390/nu16142195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Pathological states marked by oxidative stress and systemic inflammation frequently compromise the functional capacity of muscular cells. This progressive decline in muscle mass and tone can significantly hamper the patient's motor abilities, impeding even the most basic physical tasks. Muscle dysfunction can lead to metabolic disorders and severe muscle wasting, which, in turn, can potentially progress to sarcopenia. The functionality of skeletal muscle is profoundly influenced by factors such as environmental, nutritional, physical, and genetic components. A well-balanced diet, rich in proteins and vitamins, alongside an active lifestyle, plays a crucial role in fortifying tissues and mitigating general weakness and pathological conditions. Vitamin D, exerting antioxidant effects, is essential for skeletal muscle. Epidemiological evidence underscores a global prevalence of vitamin D deficiency, which induces oxidative harm, mitochondrial dysfunction, reduced adenosine triphosphate production, and impaired muscle function. This review explores the intricate molecular mechanisms through which vitamin D modulates oxidative stress and its consequent effects on muscle function. The aim is to evaluate if vitamin D supplementation in conditions involving oxidative stress and inflammation could prevent decline and promote or maintain muscle function effectively.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy;
| | - Rosa Santangelo
- Department of Medicine and Health Sciences, University of Catania, Via Santa Sofia, 97, 95124 Catania, Italy;
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy;
| | - Maria Stella Valle
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| |
Collapse
|
31
|
He J, Feng L, Yang H, Gao S, Dong J, Lu G, Liu L, Zhang X, Zhong K, Guo S, Zha G, Han L, Li H, Wang Y. Sirtuin 5 alleviates apoptosis and autophagy stimulated by ammonium chloride in bovine mammary epithelial cells. Exp Ther Med 2024; 28:295. [PMID: 38827477 PMCID: PMC11140291 DOI: 10.3892/etm.2024.12584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/28/2024] [Indexed: 06/04/2024] Open
Abstract
Ammonia (NH3) is an irritating and harmful gas that affects cell apoptosis and autophagy. Sirtuin 5 (SIRT5) has multiple enzymatic activities and regulates NH3-induced autophagy in tumor cells. In order to determine whether SIRT5 regulates NH3-induced bovine mammary epithelial cell apoptosis and autophagy, cells with SIRT5 overexpression or knockdown were generated and in addition, bovine mammary epithelial cells were treated with SIRT5 inhibitors. The results showed that SIRT5 overexpression reduced the content of NH3 and glutamate in cells by inhibiting glutaminase activity in glutamine metabolism, and reduced the ratio of ADP/ATP. The results in the SIRT5 knockdown and inhibitor groups were comparable, including increased content of NH3 and glutamate in cells by activating glutaminase activity, and an elevated ratio of ADP/ATP. It was further confirmed that SIRT5 inhibited the apoptosis and autophagy of bovine mammary epithelial cells through reverse transcription-quantitative PCR, western blot, flow cytometry with Annexin V FITC/PI staining and transmission electron microscopy. In addition, it was also found that the addition of LY294002 or Rapamycin inhibited the PI3K/Akt or mTOR kinase signal, decreasing the apoptosis and autophagy activities of bovine mammary epithelial cells induced by SIRT5-inhibited NH3. In summary, the PI3K/Akt/mTOR signal involved in NH3-induced cell autophagy and apoptosis relies on the regulation of SIRT5. This study provides a new theory for the use of NH3 to regulate bovine mammary epithelial cell apoptosis and autophagy, and provides guidance for improving the health and production performance of dairy cows.
Collapse
Affiliation(s)
- Junhui He
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Luping Feng
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Hanlin Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Shikai Gao
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Jinru Dong
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Guangyang Lu
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Luya Liu
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Xinyi Zhang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Kai Zhong
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Shuang Guo
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Guangming Zha
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Liqiang Han
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Heping Li
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Yueying Wang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| |
Collapse
|
32
|
Wang Y, Ruan L, Zhu J, Zhang X, Chang ACC, Tomaszewski A, Li R. Metabolic regulation of misfolded protein import into mitochondria. eLife 2024; 12:RP87518. [PMID: 38900507 PMCID: PMC11189628 DOI: 10.7554/elife.87518] [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] [Indexed: 06/21/2024] Open
Abstract
Mitochondria are the cellular energy hub and central target of metabolic regulation. Mitochondria also facilitate proteostasis through pathways such as the 'mitochondria as guardian in cytosol' (MAGIC) whereby cytosolic misfolded proteins (MPs) are imported into and degraded inside mitochondria. In this study, a genome-wide screen in Saccharomyces cerevisiae uncovered that Snf1, the yeast AMP-activated protein kinase (AMPK), inhibits the import of MPs into mitochondria while promoting mitochondrial biogenesis under glucose starvation. We show that this inhibition requires a downstream transcription factor regulating mitochondrial gene expression and is likely to be conferred through substrate competition and mitochondrial import channel selectivity. We further show that Snf1/AMPK activation protects mitochondrial fitness in yeast and human cells under stress induced by MPs such as those associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuhao Wang
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Biochemistry, Cellular and Molecular Biology (BCMB) Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Linhao Ruan
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Jin Zhu
- Mechanobiology Institute and Department of Biological Sciences, National University of SingaporeSingaporeSingapore
| | - Xi Zhang
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Alexander Chih-Chieh Chang
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Alexis Tomaszewski
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Biochemistry, Cellular and Molecular Biology (BCMB) Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Rong Li
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Mechanobiology Institute and Department of Biological Sciences, National University of SingaporeSingaporeSingapore
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins UniversityBaltimoreUnited States
| |
Collapse
|
33
|
Serrano J, Boyd J, Brown IS, Mason C, Smith KR, Karolyi K, Maurya SK, Meshram NN, Serna V, Link GM, Gardell SJ, Kyriazis GA. The TAS1R2 G-protein-coupled receptor is an ambient glucose sensor in skeletal muscle that regulates NAD homeostasis and mitochondrial capacity. Nat Commun 2024; 15:4915. [PMID: 38851747 PMCID: PMC11162498 DOI: 10.1038/s41467-024-49100-8] [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: 12/14/2023] [Accepted: 05/21/2024] [Indexed: 06/10/2024] Open
Abstract
The bioavailability of nicotinamide adenine dinucleotide (NAD) is vital for skeletal muscle health, yet the mechanisms or signals regulating NAD homeostasis remain unclear. Here, we uncover a pathway connecting peripheral glucose sensing to the modulation of muscle NAD through TAS1R2, the sugar-sensing G protein-coupled receptor (GPCR) initially identified in taste perception. Muscle TAS1R2 receptor stimulation by glucose and other agonists induces ERK1/2-dependent phosphorylation and activation of poly(ADP-ribose) polymerase1 (PARP1), a major NAD consumer in skeletal muscle. Consequently, muscle-specific deletion of TAS1R2 (mKO) in male mice suppresses PARP1 activity, elevating NAD levels and enhancing mitochondrial capacity and running endurance. Plasma glucose levels negatively correlate with muscle NAD, and TAS1R2 receptor deficiency enhances NAD responses across the glycemic range, implicating TAS1R2 as a peripheral energy surveyor. These findings underscore the role of GPCR signaling in NAD regulation and propose TAS1R2 as a potential therapeutic target for maintaining muscle health.
Collapse
Affiliation(s)
- Joan Serrano
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University; Columbus, Columbus, 43210, USA
| | - Jordan Boyd
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University; Columbus, Columbus, 43210, USA
| | - Ian S Brown
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University; Columbus, Columbus, 43210, USA
| | - Carter Mason
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University; Columbus, Columbus, 43210, USA
| | - Kathleen R Smith
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University; Columbus, Columbus, 43210, USA
| | - Katalin Karolyi
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University; Columbus, Columbus, 43210, USA
| | - Santosh K Maurya
- Physiology and Cell Biology, College of Medicine, The Ohio State University; Columbus, Columbus, 43210, USA
| | - Nishita N Meshram
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University; Columbus, Columbus, 43210, USA
| | - Vanida Serna
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University; Columbus, Columbus, 43210, USA
| | - Grace M Link
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University; Columbus, Columbus, 43210, USA
| | - Stephen J Gardell
- Translational Research Institute, Advent Health, Orlando, 32804, USA
| | - George A Kyriazis
- Biological Chemistry & Pharmacology, College of Medicine, The Ohio State University; Columbus, Columbus, 43210, USA.
| |
Collapse
|
34
|
Poljšak B, Milisav I. Decreasing Intracellular Entropy by Increasing Mitochondrial Efficiency and Reducing ROS Formation-The Effect on the Ageing Process and Age-Related Damage. Int J Mol Sci 2024; 25:6321. [PMID: 38928027 PMCID: PMC11203720 DOI: 10.3390/ijms25126321] [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: 04/23/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
A hypothesis is presented to explain how the ageing process might be influenced by optimizing mitochondrial efficiency to reduce intracellular entropy. Research-based quantifications of entropy are scarce. Non-equilibrium metabolic reactions and compartmentalization were found to contribute most to lowering entropy in the cells. Like the cells, mitochondria are thermodynamically open systems exchanging matter and energy with their surroundings-the rest of the cell. Based on the calculations from cancer cells, glycolysis was reported to produce less entropy than mitochondrial oxidative phosphorylation. However, these estimations depended on the CO2 concentration so that at slightly increased CO2, it was oxidative phosphorylation that produced less entropy. Also, the thermodynamic efficiency of mitochondrial respiratory complexes varies depending on the respiratory state and oxidant/antioxidant balance. Therefore, in spite of long-standing theoretical and practical efforts, more measurements, also in isolated mitochondria, with intact and suboptimal respiration, are needed to resolve the issue. Entropy increases in ageing while mitochondrial efficiency of energy conversion, quality control, and turnover mechanisms deteriorate. Optimally functioning mitochondria are necessary to meet energy demands for cellular defence and repair processes to attenuate ageing. The intuitive approach of simply supplying more metabolic fuels (more nutrients) often has the opposite effect, namely a decrease in energy production in the case of nutrient overload. Excessive nutrient intake and obesity accelerate ageing, while calorie restriction without malnutrition can prolong life. Balanced nutrient intake adapted to needs/activity-based high ATP requirement increases mitochondrial respiratory efficiency and leads to multiple alterations in gene expression and metabolic adaptations. Therefore, rather than overfeeding, it is necessary to fine-tune energy production by optimizing mitochondrial function and reducing oxidative stress; the evidence is discussed in this paper.
Collapse
Affiliation(s)
- Borut Poljšak
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia;
| | - Irina Milisav
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia;
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
35
|
Gibril BAA, Xiong X, Chai X, Xu Q, Gong J, Xu J. Unlocking the Nexus of Sirtuins: A Comprehensive Review of Their Role in Skeletal Muscle Metabolism, Development, and Disorders. Int J Biol Sci 2024; 20:3219-3235. [PMID: 38904020 PMCID: PMC11186354 DOI: 10.7150/ijbs.96885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024] Open
Abstract
The sirtuins constitute a group of histone deacetylases reliant on NAD+ for their activity that have gained recognition for their critical roles as regulators of numerous biological processes. These enzymes have various functions in skeletal muscle biology, including development, metabolism, and the body's response to disease. This comprehensive review seeks to clarify sirtuins' complex role in skeletal muscle metabolism, including glucose uptake, fatty acid oxidation, mitochondrial dynamics, autophagy regulation, and exercise adaptations. It also examines their critical roles in developing skeletal muscle, including myogenesis, the determination of muscle fiber type, regeneration, and hypertrophic responses. Moreover, it sheds light on the therapeutic potential of sirtuins by examining their impact on a range of skeletal muscle disorders. By integrating findings from various studies, this review outlines the context of sirtuin-mediated regulation in skeletal muscle, highlighting their importance and possible consequences for health and disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiguo Xu
- Jiangxi Provincial Key Laboratory of Poultry Genetic Improvement, Institute of Biological Technology, Nanchang Normal University, Nanchang, 330032, China
| |
Collapse
|
36
|
Jiang Z, Huang B, Cui Z, Lu Z, Ma H. Synergistic effect of genistein and adiponectin reduces fat deposition in chicken hepatocytes by activating the ERβ-mediated SIRT1-AMPK signaling pathway. Poult Sci 2024; 103:103734. [PMID: 38636201 PMCID: PMC11040169 DOI: 10.1016/j.psj.2024.103734] [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: 02/10/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024] Open
Abstract
Dietary supplementation with bioactive substances that can regulate lipid metabolism is an effective approach for reducing excessive fat deposition in chickens. Genistein (GEN) has the potential to alleviate fat deposition; however, the underlying mechanism of GEN's fat-reduction action in chickens remains unclear. Therefore, the present study aimed to explore the underlying mechanism of GEN on the reduction of fat deposition from a novel perspective: intercellular transmission of adipokine between adipocytes and hepatocytes. The findings showed that GEN enhanced the secretion of adiponectin (APN) in chicken adipocytes, and the enhancement effect of GEN was completely blocked when the cells were pretreated with inhibitors targeting estrogen receptor β (ERβ) or proliferator-activated receptor γ (PPARγ) signals, respectively. Furthermore, the results demonstrated that both co-treatment with GEN and APN or treatment with the medium supernatant (Med SUP) derived from chicken adipocytes treated with GEN significantly decreased the content of triglyceride and increased the protein levels of ERβ, Sirtuin 1 (SIRT1) and phosphor-AMP-activated protein kinase (p-AMPK) in chicken hepatocytes compared to the cells treated with GEN or APN alone. Moreover, the increase in the protein levels of SIRT1 and p-AMPK induced by GEN and APN co-treatment or Med SUP treatment were blocked in chicken hepatocytes pretreated with the inhibitor of ERβ signals. Importantly, the up-regulatory effect of GEN and APN co-treatment or Med SUP treatment on the protein level of p-AMPK was also blocked in chicken hepatocytes pretreated with a SIRT1 inhibitor; however, the increase in the protein level of SIRT1 induced by GEN and APN co-treatment or Med SUP treatment was not reversed when the hepatocytes were pretreated with an AMPK inhibitor. In conclusion, the present study demonstrated that GEN enhanced APN secretion by activating the ERβ-Erk-PPARγ signaling pathway in chicken adipocytes. Subsequently, adipocyte-derived APN synergized with GEN to activate the ERβ-mediated SIRT1-AMPK signaling pathway in chicken hepatocytes, ultimately reducing fat deposition. These findings provide substantial evidence from a novel perspective, supporting the potential use of GEN as a dietary supplement to prevent excessive fat deposition in poultry.
Collapse
Affiliation(s)
- Zhihao Jiang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Benzeng Huang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ziyi Cui
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ze Lu
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Haitian Ma
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
37
|
Pugh CF, Paton CD, Ferguson RA, Driller MW, Martyn Beaven C. Acute physiological responses of blood flow restriction between high-intensity interval repetitions in trained cyclists. Eur J Sport Sci 2024; 24:777-787. [PMID: 38874956 PMCID: PMC11235839 DOI: 10.1002/ejsc.12107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 06/15/2024]
Abstract
Blood flow restriction (BFR) is increasingly being used to enhance aerobic performance in endurance athletes. This study examined physiological responses to BFR applied in recovery phases within a high-intensity interval training (HIIT) session in trained cyclists. Eleven competitive road cyclists (mean ± SD, age: 28 ± 7 years, body mass: 69 ± 6 kg, peak oxygen uptake: 65 ± 9 mL · kg-1 · min-1) completed two randomised crossover conditions: HIIT with (BFR) and without (CON) BFR applied during recovery phases. HIIT consisted of six 30-s cycling bouts at an intensity equivalent to 85% of maximal 30-s power (523 ± 93 W), interspersed with 4.5-min recovery. BFR (200 mmHg, 12 cm cuff width) was applied for 2-min in the early recovery phase between each interval. Pulmonary gas exchange (V̇O2, V̇CO2, and V̇E), tissue oxygen saturation index (TSI), heart rate (HR), and serum vascular endothelial growth factor concentration (VEGF) were measured. Compared to CON, BFR increased V̇CO2 and V̇E during work bouts (both p < 0.05, dz < 0.5), but there was no effect on V̇O2, TSI, or HR (p > 0.05). In early recovery, BFR decreased TSI, V̇O2, V̇CO2, and V̇E (all p < 0.05, dz > 0.8) versus CON, with no change in HR (p > 0.05). In late recovery, when BFR was released, V̇O2, V̇CO2, V̇E, and HR increased, but TSI decreased versus CON (all p < 0.05, dz > 0.8). There was a greater increase in VEGF at 3-h post-exercise in BFR compared to CON (p < 0.05, dz > 0.8). Incorporating BFR into HIIT recovery phases altered physiological responses compared to exercise alone.
Collapse
Affiliation(s)
- Charles F. Pugh
- Te Huataki Waiora School of HealthUniversity of WaikatoHamiltonNew Zealand
| | - Carl D. Paton
- School of Health and Sport ScienceTe PukengaThe Eastern Institute of TechnologyNapierNew Zealand
| | - Richard A. Ferguson
- School of Sport, Exercise and Health SciencesLoughborough UniversityLoughboroughUK
| | - Matthew W. Driller
- Sport, Performance and Nutrition Research GroupSchool of Allied Health, Human Services and SportLa Trobe UniversityMelbourneVictoriaAustralia
| | - C. Martyn Beaven
- Te Huataki Waiora School of HealthUniversity of WaikatoHamiltonNew Zealand
| |
Collapse
|
38
|
Chen M, Tan J, Jin Z, Jiang T, Wu J, Yu X. Research progress on Sirtuins (SIRTs) family modulators. Biomed Pharmacother 2024; 174:116481. [PMID: 38522239 DOI: 10.1016/j.biopha.2024.116481] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024] Open
Abstract
Sirtuins (SIRTs) represent a class of nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylases that exert a crucial role in cellular signal transduction and various biological processes. The mammalian sirtuins family encompasses SIRT1 to SIRT7, exhibiting therapeutic potential in counteracting cellular aging, modulating metabolism, responding to oxidative stress, inhibiting tumors, and improving cellular microenvironment. These enzymes are intricately linked to the occurrence and treatment of diverse pathological conditions, including cancer, autoimmune diseases, and cardiovascular disorders. Given the significance of histone modification in gene expression and chromatin structure, maintaining the equilibrium of the sirtuins family is imperative for disease prevention and health restoration. Mounting evidence suggests that modulators of SIRTs play a crucial role in treating various diseases and maintaining physiological balance. This review delves into the molecular structure and regulatory functions of the sirtuins family, reviews the classification and historical evolution of SIRTs modulators, offers a systematic overview of existing SIRTs modulation strategies, and elucidates the regulatory mechanisms of SIRTs modulators (agonists and inhibitors) and their clinical applications. The article concludes by summarizing the challenges encountered in SIRTs modulator research and offering insights into future research directions.
Collapse
Affiliation(s)
- Mingkai Chen
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China; School of Medicine Jiangsu University, Zhenjiang, Jiangsu, China
| | - Junfei Tan
- School of Medicine Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zihan Jin
- Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou City, China
| | - Tingting Jiang
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
| | - Jiabiao Wu
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
| | - Xiaolong Yu
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China; The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China.
| |
Collapse
|
39
|
Jiang YZ, Huang XR, Chang J, Zhou Y, Huang XT. SIRT1: An Intermediator of Key Pathways Regulating Pulmonary Diseases. J Transl Med 2024; 104:102044. [PMID: 38452903 DOI: 10.1016/j.labinv.2024.102044] [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: 08/31/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024] Open
Abstract
Silent information regulator type-1 (SIRT1), a nicotinamide adenine dinucleotide+-dependent deacetylase, is a member of the sirtuins family and has unique protein deacetylase activity. SIRT1 participates in physiological as well as pathophysiological processes by targeting a wide range of protein substrates and signalings. In this review, we described the latest progress of SIRT1 in pulmonary diseases. We have introduced the basic information and summarized the prominent role of SIRT1 in several lung diseases, such as acute lung injury, acute respiratory distress syndrome, chronic obstructive pulmonary disease, lung cancer, and aging-related diseases.
Collapse
Affiliation(s)
- Yi-Zhu Jiang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xin-Ran Huang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jing Chang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, China.
| |
Collapse
|
40
|
Jevtovic F, Zheng D, Claiborne A, Biagioni EM, Wisseman BL, Krassovskaia PM, Collier DN, Isler C, DeVente JE, Neufer PD, Houmard JA, May LE. Effects of maternal exercise on infant mesenchymal stem cell mitochondrial function, insulin action, and body composition in infancy. Physiol Rep 2024; 12:e16028. [PMID: 38684442 PMCID: PMC11058002 DOI: 10.14814/phy2.16028] [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: 03/05/2024] [Revised: 03/30/2024] [Accepted: 04/12/2024] [Indexed: 05/02/2024] Open
Abstract
Maternal exercise (ME) has been established as a useful non-pharmacological intervention to improve infant metabolic health; however, mechanistic insight behind these adaptations remains mostly confined to animal models. Infant mesenchymal stem cells (MSCs) give rise to infant tissues (e.g., skeletal muscle), and remain involved in mature tissue maintenance. Importantly, these cells maintain metabolic characteristics of an offspring donor and provide a model for the investigation of mechanisms behind infant metabolic health improvements. We used undifferentiated MSC to investigate if ME affects infant MSC mitochondrial function and insulin action, and if these adaptations are associated with lower infant adiposity. We found that infants from exercising mothers have improvements in MSC insulin signaling related to higher MSC respiration and fat oxidation, and expression and activation of energy-sensing and redox-sensitive proteins. Further, we found that infants exposed to exercise in utero were leaner at 1 month of age, with a significant inverse correlation between infant MSC respiration and infant adiposity at 6 months of age. These data suggest that infants from exercising mothers are relatively leaner, and this is associated with higher infant MSC mitochondrial respiration, fat use, and insulin action.
Collapse
Affiliation(s)
- Filip Jevtovic
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Donghai Zheng
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Alex Claiborne
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Ericka M. Biagioni
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Breanna L. Wisseman
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Polina M. Krassovskaia
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - David N. Collier
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Department of Pediatrics, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Christy Isler
- Department of Obstetrics and Gynecology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - James E. DeVente
- Department of Obstetrics and Gynecology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - P. Darrell Neufer
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Department of Physiology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Department of Biochemistry & Molecular Biology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Joseph A. Houmard
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Linda E. May
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Department of Obstetrics and Gynecology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| |
Collapse
|
41
|
Zhao C, Gong Y, Zheng L, Zhao M. Untargeted metabolomic reveals the changes in muscle metabolites of mice during exercise recovery and the mechanisms of whey protein and whey protein hydrolysate in promoting muscle repair. Food Res Int 2024; 184:114261. [PMID: 38609238 DOI: 10.1016/j.foodres.2024.114261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024]
Abstract
Our previous study indicated that whey protein hydrolysate (WPH) showed effective anti-fatigue properties, but its regulatory mechanism on recovery from exercise in mice is unclear. In the present study, we divided the mice into control, WP, and WPH groups and allowed them to rest for 1 h and 24 h after exercise, respectively. The changes in muscle metabolites of mice in the recovery period were investigated using metabolomics techniques. The results showed that the WPH group significantly up-regulated 94 muscle metabolites within 1 h of rest, which was 1.96 and 2.61 times more than the control and WP groups, respectively. In detail, significant decreases in TCA cycle intermediates, lipid metabolites, and carbohydrate metabolites were observed in the control group during exercise recovery. In contrast, administration with WP and WPH enriched more amino acid metabolites within 1 h of rest, which might provide a more comprehensive metabolic environment for muscle repair. Moreover, the WPH group remarkably stimulated the enhancement of lipid, carbohydrate, and vitamin metabolites in the recovery period which might provide raw materials and energy for anabolic reactions. The result of the western blot further demonstrated that WPH could promote muscle repair via activating the Sestrin2/Akt/mTOR/S6K signaling pathway within 1 h of rest. These findings deepen our understanding of the regulatory mechanisms by WPH to promote muscle recovery and may serve as a reference for comprehensive assessments of protein supplements on exercise.
Collapse
Affiliation(s)
- Chaoya Zhao
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| | - Yurong Gong
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China
| | - Lin Zheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Mouming Zhao
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Food Laboratory of Zhongyuan, Luohe 462300, China; Guangdong Food Green Processing and Nutrition Regulation Technologies Research Center, Guangzhou 510650, China.
| |
Collapse
|
42
|
Rahman SU, Qadeer A, Wu Z. Role and Potential Mechanisms of Nicotinamide Mononucleotide in Aging. Aging Dis 2024; 15:565-583. [PMID: 37548938 PMCID: PMC10917541 DOI: 10.14336/ad.2023.0519-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/19/2023] [Indexed: 08/08/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) has recently attracted much attention due to its role in aging and lifespan extension. NAD+ directly and indirectly affects many cellular processes, including metabolic pathways, DNA repair, and immune cell activities. These mechanisms are critical for maintaining cellular homeostasis. However, the decline in NAD+ levels with aging impairs tissue function, which has been associated with several age-related diseases. In fact, the aging population has been steadily increasing worldwide, and it is important to restore NAD+ levels and reverse or delay these age-related disorders. Therefore, there is an increasing demand for healthy products that can mitigate aging, extend lifespan, and halt age-related consequences. In this case, several studies in humans and animals have targeted NAD+ metabolism with NAD+ intermediates. Among them, nicotinamide mononucleotide (NMN), a precursor in the biosynthesis of NAD+, has recently received much attention from the scientific community for its anti-aging properties. In model organisms, ingestion of NMN has been shown to improve age-related diseases and probably delay death. Here, we review aspects of NMN biosynthesis and the mechanism of its absorption, as well as potential anti-aging mechanisms of NMN, including recent preclinical and clinical tests, adverse effects, limitations, and perceived challenges.
Collapse
Affiliation(s)
- Sajid Ur Rahman
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Abdul Qadeer
- Institute for Infectious Diseases and Vaccine Development, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Ziyun Wu
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
43
|
Baghdassarian HM, Lewis NE. Resource allocation in mammalian systems. Biotechnol Adv 2024; 71:108305. [PMID: 38215956 PMCID: PMC11182366 DOI: 10.1016/j.biotechadv.2023.108305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/14/2024]
Abstract
Cells execute biological functions to support phenotypes such as growth, migration, and secretion. Complementarily, each function of a cell has resource costs that constrain phenotype. Resource allocation by a cell allows it to manage these costs and optimize their phenotypes. In fact, the management of resource constraints (e.g., nutrient availability, bioenergetic capacity, and macromolecular machinery production) shape activity and ultimately impact phenotype. In mammalian systems, quantification of resource allocation provides important insights into higher-order multicellular functions; it shapes intercellular interactions and relays environmental cues for tissues to coordinate individual cells to overcome resource constraints and achieve population-level behavior. Furthermore, these constraints, objectives, and phenotypes are context-dependent, with cells adapting their behavior according to their microenvironment, resulting in distinct steady-states. This review will highlight the biological insights gained from probing resource allocation in mammalian cells and tissues.
Collapse
Affiliation(s)
- Hratch M Baghdassarian
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
44
|
Qian L, Zhu Y, Deng C, Liang Z, Chen J, Chen Y, Wang X, Liu Y, Tian Y, Yang Y. Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family in physiological and pathophysiological process and diseases. Signal Transduct Target Ther 2024; 9:50. [PMID: 38424050 PMCID: PMC10904817 DOI: 10.1038/s41392-024-01756-w] [Citation(s) in RCA: 76] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/13/2024] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family (PGC-1s), consisting of three members encompassing PGC-1α, PGC-1β, and PGC-1-related coactivator (PRC), was discovered more than a quarter-century ago. PGC-1s are essential coordinators of many vital cellular events, including mitochondrial functions, oxidative stress, endoplasmic reticulum homeostasis, and inflammation. Accumulating evidence has shown that PGC-1s are implicated in many diseases, such as cancers, cardiac diseases and cardiovascular diseases, neurological disorders, kidney diseases, motor system diseases, and metabolic disorders. Examining the upstream modulators and co-activated partners of PGC-1s and identifying critical biological events modulated by downstream effectors of PGC-1s contribute to the presentation of the elaborate network of PGC-1s. Furthermore, discussing the correlation between PGC-1s and diseases as well as summarizing the therapy targeting PGC-1s helps make individualized and precise intervention methods. In this review, we summarize basic knowledge regarding the PGC-1s family as well as the molecular regulatory network, discuss the physio-pathological roles of PGC-1s in human diseases, review the application of PGC-1s, including the diagnostic and prognostic value of PGC-1s and several therapies in pre-clinical studies, and suggest several directions for future investigations. This review presents the immense potential of targeting PGC-1s in the treatment of diseases and hopefully facilitates the promotion of PGC-1s as new therapeutic targets.
Collapse
Affiliation(s)
- Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yanli Zhu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou, 450052, China
| | - Junmin Chen
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xue Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yanqing Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ye Tian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yang Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China.
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
45
|
Swargiary D, Kashyap B, Sarma P, Ahmed SA, Gurumayum S, Barge SR, Basumatary D, Borah JC. Free radical scavenging polyphenols isolated from Phyllanthus niruri L. ameliorates hyperglycemia via SIRT1 induction and GLUT4 translocation in in vitro and in vivo models. Fitoterapia 2024; 173:105803. [PMID: 38171388 DOI: 10.1016/j.fitote.2023.105803] [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: 09/29/2023] [Revised: 12/12/2023] [Accepted: 12/28/2023] [Indexed: 01/05/2024]
Abstract
Type 2 diabetes milletus (T2DM) is a complex multifaceted disorder characterized by insulin resistance in skeletal muscle. Phyllanthus niruri L. is well reported sub-tropical therapeutically beneficial ayurvedic medicinal plant from Euphorbiaceae family used in various body ailments such as metabolic disorder including diabetes. The present study emphasizes on the therapeutic potential of Phyllanthus niruri L. and its phytochemical(s) against insulin resistance conditions and impaired antioxidant activity thereby aiding as an anti-hyperglycemic agent in targeting T2DM. Three compounds were isolated from the most active ethyl acetate fraction namely compound 1 as 1-O-galloyl-6-O-luteoyl-β-D-glucoside, compound 2 as brevifolincarboxylic acid and compound 3 as ricinoleic acid. Compounds 1 and 2, the two polyphenols enhanced the uptake of glucose and inhibited ROS levels in palmitate induced C2C12 myotubes. PNEAF showed the potent enhancement of glucose uptake in palmitate-induced insulin resistance condition in C2C12 myotubes and significant ROS inhibition was observed in skeletal muscle cell line. PNEAF treated IR C2C12 myotubes and STZ induced Wistar rats elevated SIRT1, PGC1-α signaling cascade through phosphorylation of AMPK and GLUT4 translocation resulting in insulin sensitization. Our study revealed an insight into the efficacy of marker compounds isolated from P. niruri and its enriched ethyl acetate fraction as ROS scavenging agent and helps in attenuating insulin resistance condition in C2C12 myotubes as well as in STZ induced Wistar rat by restoring glucose metabolism. Overall, this study can provide prospects for the marker-assisted development of P. niruri as a phytopharmaceutical drug for the insulin resistance related diabetic complications.
Collapse
Affiliation(s)
- Deepsikha Swargiary
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P, India
| | - Bhaswati Kashyap
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Pranamika Sarma
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Semim Akhtar Ahmed
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P, India
| | - Shalini Gurumayum
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Sagar Ramrao Barge
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Devi Basumatary
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Jagat C Borah
- Chemical Biology Lab-I, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P, India.
| |
Collapse
|
46
|
Storoschuk KL, Lesiuk D, Nuttall J, LeBouedec M, Khansari A, Islam H, Gurd BJ. Impact of fasting on the AMPK and PGC-1α axis in rodent and human skeletal muscle: A systematic review. Metabolism 2024; 152:155768. [PMID: 38154612 DOI: 10.1016/j.metabol.2023.155768] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 12/30/2023]
Abstract
Based primarily on evidence from rodent models fasting is currently believed to improve metabolic health via activation of the AMPK-PGC-1α axis in skeletal muscle. However, it is unclear whether the skeletal muscle AMPK-PGC-1α axis is activated by fasting in humans. The current systematic review examined the fasting response in skeletal muscle from 34 selected studies (7 human, 21 mouse, and 6 rat). From these studies, we gathered 38 unique data points related to AMPK and 47 related to PGC-1α. In human studies, fasting mediated activation of the AMPK-PGC-1α axis is largely absent. Although evidence does support fasting-induced activation of the AMPK-PGC-1α axis in rodent skeletal muscle, the evidence is less robust than anticipated. Our findings question the ability of fasting to activate the AMPK-PGC-1α axis in human skeletal muscle and suggest that the metabolic benefits of fasting in humans are associated with caloric restriction rather than the induction of mitochondrial biogenesis. Registration: https://doi.org/10.17605/OSF.IO/KWNQY.
Collapse
Affiliation(s)
- K L Storoschuk
- School of Kinesiology and Health Studies, Queen's University, Kingston, Ontario, Canada
| | - D Lesiuk
- School of Kinesiology and Health Studies, Queen's University, Kingston, Ontario, Canada
| | - J Nuttall
- School of Kinesiology and Health Studies, Queen's University, Kingston, Ontario, Canada
| | - M LeBouedec
- School of Kinesiology and Health Studies, Queen's University, Kingston, Ontario, Canada
| | - A Khansari
- School of Kinesiology and Health Studies, Queen's University, Kingston, Ontario, Canada
| | - H Islam
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, Canada
| | - B J Gurd
- School of Kinesiology and Health Studies, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
47
|
Kondoh H, Kameda M. Metabolites in aging and aging-relevant diseases: Frailty, sarcopenia and cognitive decline. Geriatr Gerontol Int 2024; 24 Suppl 1:44-48. [PMID: 37837183 PMCID: PMC11503595 DOI: 10.1111/ggi.14684] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 10/15/2023]
Abstract
Aging shows biologically complex features with high individual variability, which reflects the exposure to several stimuli and the adaptation to them. Among them, metabolic changes are well observed as consequences or possible causes of aging. Calorie restriction extends organismal life span in experimental models. Several metabolites; for example, resveratrol or nicotinamide mononucleotide, are reported to mimic calorie restriction effects in vivo. Metabolomic research would be useful to evaluate metabolites as biomarkers in aging-relevant events and to identify metabolic regulation of aging. We recently developed the metabolomic approach for whole blood analysis, which functions as strong tool for this purpose. We review the update findings in aging-relevant metabolites detected by this method. Geriatr Gerontol Int 2024; 24: 44-48.
Collapse
Affiliation(s)
- Hiroshi Kondoh
- Geriatric Unit, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Masahiro Kameda
- Geriatric Unit, Graduate School of MedicineKyoto UniversityKyotoJapan
| |
Collapse
|
48
|
Yin X, Guo Z, Song C. AMPK, a key molecule regulating aging-related myocardial ischemia-reperfusion injury. Mol Biol Rep 2024; 51:257. [PMID: 38302614 DOI: 10.1007/s11033-023-09050-8] [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: 05/17/2023] [Accepted: 10/10/2023] [Indexed: 02/03/2024]
Abstract
Aging leads to the threat of more diseases to the biological anatomical structure and the decline of disease resistance, increasing the incidence and mortality of myocardial ischemia-reperfusion injury (MI/RI). Moreover, MI/RI promotes damage to an aging heart. Notably, 5'-adenosine monophosphate-activated protein kinase (AMPK) regulates cellular energy metabolism, stress response, and protein metabolism, participates in aging-related signaling pathways, and plays an essential role in ischemia-reperfusion (I/R) injury diseases. This study aims to introduce the aging theory, summarize the interaction between aging and MI/RI, and describe the crosstalk of AMPK in aging and MI/RI. We show how AMPK can offer protective effects against age-related stressors, lifestyle factors such as alcohol consumption and smoking, and hypertension. We also review some of the clinical prospects for the development of interventions that harness the effect of AMPK to treat MI/RI and other age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaorui Yin
- Department of Cardiology, Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun, 130041, China
| | - Ziyuan Guo
- Department of Cardiology, Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun, 130041, China
| | - Chunli Song
- Department of Cardiology, Second Hospital of Jilin University, No.218 Ziqiang Street, Changchun, 130041, China.
| |
Collapse
|
49
|
Wei X, Tan Y, Huang J, Dong X, Feng W, Liu T, Yang Z, Yang G, Luo X. N1-methylnicotinamide impairs gestational glucose tolerance in mice. J Mol Endocrinol 2024; 72:e230126. [PMID: 38029302 PMCID: PMC10831565 DOI: 10.1530/jme-23-0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 11/29/2023] [Indexed: 12/01/2023]
Abstract
N1-methylnicotinamide (MNAM), a product of methylation of nicotinamide through nicotinamide N-methyltransferase, displays antidiabetic effects in male rodents. This study aimed to evaluate the ameliorative potential of MNAM on glucose metabolism in a gestational diabetes mellitus (GDM) model. C57BL/6N mice were fed with a high-fat diet (HFD) for 6 weeks before pregnancy and throughout gestation to establish the GDM model. Pregnant mice were treated with 0.3% or 1% MNAM during gestation. MNAM supplementation in CHOW diet and HFD both impaired glucose tolerance at gestational day 14.5 without changes in insulin tolerance. However, MNAM supplementation reduced hepatic lipid accumulation as well as mass and inflammation in visceral adipose tissue. MNAM treatment decreased GLUT4 mRNA and protein expression in skeletal muscle, where NAD+ salvage synthesis and antioxidant defenses were dampened. The NAD+/sirtuin system was enhanced in liver, which subsequently boosted hepatic gluconeogenesis. GLUT1 protein was diminished in placenta by MNAM. In addition, weight of placenta, fetus weight, and litter size were not affected by MNAM treatment. The decreased GLUT4 in skeletal muscle, boosted hepatic gluconeogenesis and dampened GLUT1 in placenta jointly contribute to the impairment of glucose tolerance tests by MNAM. Our data provide evidence for the careful usage of MNAM in treatment of GDM.
Collapse
Affiliation(s)
- Xiaojing Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yutian Tan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Jiaqi Huang
- Institute of Basic Medicine, School of Medicine, Tsinghua University, Beijing, China
| | - Ximing Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Weijie Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Tanglin Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Zhao Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Guiying Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi’an Jiaotong University, Xi’an, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, China
| |
Collapse
|
50
|
Engin A. Misalignment of Circadian Rhythms in Diet-Induced Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:27-71. [PMID: 39287848 DOI: 10.1007/978-3-031-63657-8_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The biological clocks of the circadian timing system coordinate cellular and physiological processes and synchronize them with daily cycles. While the central clock in the suprachiasmatic nucleus (SCN) is mainly synchronized by the light/dark cycles, the peripheral clocks react to other stimuli, including the feeding/fasting state, nutrients, sleep-wake cycles, and physical activity. During the disruption of circadian rhythms due to genetic mutations or social and occupational obligations, incorrect arrangement between the internal clock system and environmental rhythms leads to the development of obesity. Desynchronization between the central and peripheral clocks by altered timing of food intake and diet composition leads to uncoupling of the peripheral clocks from the central pacemaker and to the development of metabolic disorders. The strong coupling of the SCN to the light-dark cycle creates a situation of misalignment when food is ingested during the "wrong" time of day. Food-anticipatory activity is mediated by a self-sustained circadian timing, and its principal component is a food-entrainable oscillator. Modifying the time of feeding alone greatly affects body weight, whereas ketogenic diet (KD) influences circadian biology, through the modulation of clock gene expression. Night-eating behavior is one of the causes of circadian disruption, and night eaters have compulsive and uncontrolled eating with severe obesity. By contrast, time-restricted eating (TRE) restores circadian rhythms through maintaining an appropriate daily rhythm of the eating-fasting cycle. The hypothalamus has a crucial role in the regulation of energy balance rather than food intake. While circadian locomotor output cycles kaput (CLOCK) expression levels increase with high-fat diet-induced obesity, peroxisome proliferator-activated receptor-alpha (PPARα) increases the transcriptional level of brain and muscle aryl hydrocarbon receptor nuclear translocator (ARNT)-like 1 (BMAL1) in obese subjects. In this context, effective timing of chronotherapies aiming to correct SCN-driven rhythms depends on an accurate assessment of the SCN phase. In fact, in a multi-oscillator system, local rhythmicity and its disruption reflects the disruption of either local clocks or central clocks, thus imposing rhythmicity on those local tissues, whereas misalignment of peripheral oscillators is due to exosome-based intercellular communication.Consequently, disruption of clock genes results in dyslipidemia, insulin resistance, and obesity, while light exposure during the daytime, food intake during the daytime, and sleeping during the biological night promote circadian alignment between the central and peripheral clocks. Thus, shift work is associated with an increased risk of obesity, diabetes, and cardiovascular diseases because of unusual eating times as well as unusual light exposure and disruption of the circadian rhythm.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|