1
|
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 2024:156045. [PMID: 39393515 DOI: 10.1016/j.metabol.2024.156045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
2
|
Liu J, Wang S, Shen Y, Shi H, Han L. Lipid metabolites and sarcopenia-related traits: a Mendelian randomization study. Diabetol Metab Syndr 2024; 16:231. [PMID: 39285470 PMCID: PMC11406728 DOI: 10.1186/s13098-024-01465-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVE To explore the influence of lipid metabolism on the risk of sarcopenia. METHODS Two-sample Mendelian randomization (MR) analysis was used to determine causality. A total of 179 lipid metabolism data points were used for exposure, and the data were obtained from a plasma lipid metabolite study of 7174 participants. The total muscle mass and total muscle strength, as well as the muscle strength and muscle mass of different sex groups, were selected as the relevant traits of sarcopenia. Data for outcomes were obtained from the UK Biobank, and sample sizes ranged from 135 468 to 450 243. Inverse-variance weighted (IVW), as the main method for evaluating the causal relationship between lipid metabolites and sarcopenia, uses the false discovery rate (FDR) for multiple comparisons and conducts heterogeneity, pleiotropy, and reverse causality tests. RESULTS Twenty-seven lipid metabolites, mainly phosphatidylcholine, phosphatidylethanolamine, ceramide, triacylglycerol, sphingomyelin, and sterol ester, were found to be associated with the risk of sarcopenia. Ceramide (d40:1), ceramide (d40:2), and sterol ester are risk factors for decreased muscle mass and strength. There is a positive causal relationship between various phosphatidylcholine lipids and muscle mass and strength. Sphingomyelin (d42:2) is a protective factor for total muscle strength and female muscle strength. There are inconsistent effects between different lipid metabolites, triacylglycerol, and muscle strength and muscle mass. CONCLUSIONS There was a causal relationship between 27 lipid metabolites and sarcopenia traits, and targeting specific lipid metabolites may benefit sarcopenia diagnosis, disease assessment, and treatment.
Collapse
Affiliation(s)
- Jianping Liu
- Department of Neurology, Yancheng Third People's Hospital (The Sixth Affiliated Hospital of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, The affiliated hospital of Jiangsu Vocational College of Medicine), Yancheng, Jiangsu, China
| | - Sufang Wang
- Department of Neurology, Yancheng Third People's Hospital (The Sixth Affiliated Hospital of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, The affiliated hospital of Jiangsu Vocational College of Medicine), Yancheng, Jiangsu, China
| | - Yuan Shen
- Department of Neurology, Yancheng Third People's Hospital (The Sixth Affiliated Hospital of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, The affiliated hospital of Jiangsu Vocational College of Medicine), Yancheng, Jiangsu, China
| | - Haicun Shi
- Department of Neurology, Yancheng Third People's Hospital (The Sixth Affiliated Hospital of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, The affiliated hospital of Jiangsu Vocational College of Medicine), Yancheng, Jiangsu, China
| | - Lijian Han
- Department of Neurology, Yancheng Third People's Hospital (The Sixth Affiliated Hospital of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, The affiliated hospital of Jiangsu Vocational College of Medicine), Yancheng, Jiangsu, China.
| |
Collapse
|
3
|
Zhang J, Zhang F, Zhang L, Zhang M, Liu S, Ma Y. Screening and molecular docking verification of feature genes related to phospholipid metabolism in hepatocarcinoma caused by hepatitis B. Lipids Health Dis 2024; 23:268. [PMID: 39182089 PMCID: PMC11344459 DOI: 10.1186/s12944-024-02253-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND The progression of tumours is related to abnormal phospholipid metabolism. This study is anticipated to present a fresh perspective for disease therapy targets of hepatocarcinoma caused by hepatitis B virus in the future by screening feature genes related to phospholipid metabolism. METHODS This study analysed GSE121248 to pinpoint differentially expressed genes (DEGs). By examining the overlap between the metabolism-related genes and DEGs, the research focused on the genes involved in phospholipid metabolism. To find feature genes, functional enrichment studies were carried out and a network diagram was proposed. These findings were validated via data base of The Cancer Genome Atlas (TCGA). Further analyses included immune infiltration studies and metabolomics. Finally, the relationships between differentially abundant metabolites and feature genes were confirmed by molecular docking, providing a thorough comprehension of the molecular mechanisms. RESULTS The seven genes with the highest degree of connection (PTGS2, IGF1, SPP1, BCHE, NR1I2, NAMPT, and FABP1) were identified as feature genes. In the TCGA database, the seven feature genes also had certain diagnostic efficiency. Immune infiltration analysis revealed that feature genes regulate the infiltration of various immune cells. Metabolomics successfully identified the different metabolites of the phospholipid metabolism pathway between patients and normal individuals. The docking study indicated that different metabolites may play essential roles in causing disease by targeting feature genes. CONCLUSIONS In this study, for the first time, it reveals the possible involvement of genes linked to phospholipid metabolism-related genes using bioinformatics analysis. Identifying genes and probable therapeutic targets could provide clues for the further treatment of disease.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Clinical Laboratory, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Fengmei Zhang
- Department of Clinical Laboratory, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, The Third Central Hospital of Tianjin, Tianjin Institute of Hepatobiliary Disease, Tianjin, 300170, China
| | - Lei Zhang
- Department of Clinical Laboratory, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, The Third Central Hospital of Tianjin, Tianjin Institute of Hepatobiliary Disease, Tianjin, 300170, China.
| | - Meiling Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Shuye Liu
- Department of Clinical Laboratory, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, The Third Central Hospital of Tianjin, Tianjin Institute of Hepatobiliary Disease, Tianjin, 300170, China.
| | - Ying Ma
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
4
|
Yang Q, Zhang Z, He P, Mao X, Jing X, Hu Y, Jing L. LC/MS-Based Untargeted Lipidomics Reveals Lipid Signatures of Sarcopenia. Int J Mol Sci 2024; 25:8793. [PMID: 39201479 PMCID: PMC11354784 DOI: 10.3390/ijms25168793] [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: 07/10/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Sarcopenia, a multifactorial systemic disorder, has attracted extensive attention, yet its pathogenesis is not fully understood, partly due to limited research on the relationship between lipid metabolism abnormalities and sarcopenia. Lipidomics offers the possibility to explore this relationship. Our research utilized LC/MS-based nontargeted lipidomics to investigate the lipid profile changes as-sociated with sarcopenia, aiming to enhance understanding of its underlying mechanisms. The study included 40 sarcopenia patients and 40 control subjects matched 1:1 by sex and age. Plasma lipids were detected and quantified, with differential lipids identified through univariate and mul-tivariate statistical analyses. A weighted correlation network analysis (WGCNA) and MetaboAna-lyst were used to identify lipid modules related to the clinical traits of sarcopenia patients and to conduct pathway analysis, respectively. A total of 34 lipid subclasses and 1446 lipid molecules were detected. Orthogonal partial least squares discriminant analysis (OPLS-DA) identified 80 differen-tial lipid molecules, including 38 phospholipids. Network analysis revealed that the brown module (encompassing phosphatidylglycerol (PG) lipids) and the yellow module (containing phosphati-dylcholine (PC), phosphatidylserine (PS), and sphingomyelin (SM) lipids) were closely associated with the clinical traits such as maximum grip strength and skeletal muscle mass (SMI). Pathway analysis highlighted the potential role of the glycerophospholipid metabolic pathway in lipid me-tabolism within the context of sarcopenia. These findings suggest a correlation between sarcopenia and lipid metabolism disturbances, providing valuable insights into the disease's underlying mechanisms and indicating potential avenues for further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lipeng Jing
- Institute of Epidemiology and Statistics, School of Public Health, Lanzhou University, Lanzhou 730000, China; (Q.Y.); (Z.Z.); (P.H.); (X.M.); (X.J.); (Y.H.)
| |
Collapse
|
5
|
Han P, Chen X, Liang Z, Liu Y, Yu X, Song P, Zhao Y, Zhang H, Zhu S, Shi X, Guo Q. Metabolic signatures and risk of sarcopenia in suburb-dwelling older individuals by LC-MS-based untargeted metabonomics. Front Endocrinol (Lausanne) 2024; 15:1308841. [PMID: 38962681 PMCID: PMC11220188 DOI: 10.3389/fendo.2024.1308841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 06/04/2024] [Indexed: 07/05/2024] Open
Abstract
Background Untargeted metabonomics has provided new insight into the pathogenesis of sarcopenia. In this study, we explored plasma metabolic signatures linked to a heightened risk of sarcopenia in a cohort study by LC-MS-based untargeted metabonomics. Methods In this nested case-control study from the Adult Physical Fitness and Health Cohort Study (APFHCS), we collected blood plasma samples from 30 new-onset sarcopenia subjects (mean age 73.2 ± 5.6 years) and 30 healthy controls (mean age 74.2 ± 4.6 years) matched by age, sex, BMI, lifestyle, and comorbidities. An untargeted metabolomics methodology was employed to discern the metabolomic profile alterations present in individuals exhibiting newly diagnosed sarcopenia. Results In comparing individuals with new-onset sarcopenia to normal controls, a comprehensive analysis using liquid chromatography-mass spectrometry (LC-MS) identified a total of 62 metabolites, predominantly comprising lipids, lipid-like molecules, organic acids, and derivatives. Receiver operating characteristic (ROC) curve analysis indicated that the three metabolites hypoxanthine (AUC=0.819, 95% CI=0.711-0.927), L-2-amino-3-oxobutanoic acid (AUC=0.733, 95% CI=0.598-0.868) and PC(14:0/20:2(11Z,14Z)) (AUC= 0.717, 95% CI=0.587-0.846) had the highest areas under the curve. Then, these significant metabolites were observed to be notably enriched in four distinct metabolic pathways, namely, "purine metabolism"; "parathyroid hormone synthesis, secretion and action"; "choline metabolism in cancer"; and "tuberculosis". Conclusion The current investigation elucidates the metabolic perturbations observed in individuals diagnosed with sarcopenia. The identified metabolites hold promise as potential biomarkers, offering avenues for exploring the underlying pathological mechanisms associated with sarcopenia.
Collapse
Affiliation(s)
- Peipei Han
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
| | - Xiaoyu Chen
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Zhenwen Liang
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yuewen Liu
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xing Yu
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Peiyu Song
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
| | - Yinjiao Zhao
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
| | - Hui Zhang
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
| | - Shuyan Zhu
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xinyi Shi
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Qi Guo
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
| |
Collapse
|
6
|
Egawa T, Ogawa T, Yokokawa T, Kido K, Iyama R, Zhao H, Kurogi E, Goto K, Hayashi T. Glycative stress inhibits hypertrophy and impairs cell membrane integrity in overloaded mouse skeletal muscle. J Cachexia Sarcopenia Muscle 2024; 15:883-896. [PMID: 38575520 PMCID: PMC11154761 DOI: 10.1002/jcsm.13444] [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: 06/21/2023] [Revised: 01/04/2024] [Accepted: 01/21/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Glycative stress, characterized by the formation and accumulation of advanced glycation end products (AGEs) associated with protein glycation reactions, has been implicated in inducing a decline of muscle function. Although the inverse correlation between glycative stress and muscle mass and strength has been demonstrated, the underlying molecular mechanisms are not fully understood. This study aimed to elucidate how glycative stress affects the skeletal muscle, particularly the adaptive muscle response to hypertrophic stimuli and its molecular mechanism. METHODS Male C57BL/6NCr mice were randomly divided into the following two groups: the bovine serum albumin (BSA)-treated and AGE-treated groups. Mice in the AGE-treated group were intraperitoneally administered AGEs (0.5 mg/g) once daily, whereas those in the BSA-treated group received an equal amount of BSA (0.5 mg/g) as the vehicle control. After 7 days of continuous administration, the right leg plantaris muscle of mice in each group underwent functional overload treatment by synergist ablation for 7 days to induce muscle hypertrophy. In in vitro studies, cultured C2C12 myocytes were treated with AGEs (1 mg/mL) to examine cell adhesion and cell membrane permeability. RESULTS Continuous AGE administration increased the levels of fluorescent AGEs, Nε-(carboxymethyl) lysine, and methylglyoxal-derived hydroimidazolone-1 in both plasma and skeletal muscle. Plantaris muscle weight, muscle fibre cross-sectional area, protein synthesis rate, and the number of myonuclei increased with functional overload in both groups; however, the increase was significantly reduced by AGE treatment. Some muscles of AGE-treated mice were destroyed by functional overload. Proteomic analysis was performed to explore the mechanisms of muscle hypertrophy suppression and myofibre destruction by AGEs. When principal component analysis was performed on 4659 data obtained by proteomic analysis, AGE treatment was observed to affect protein expression only in functionally overloaded muscles. Enrichment analysis of the 436 proteins extracted using the K-means method further identified a group of proteins involved in cell adhesion. Consistent with this finding, dystrophin-glycoprotein complex proteins and cell adhesion-related proteins were confirmed to increase with functional overload; however, this was attenuated by AGE treatment. Additionally, the treatment of C2C12 muscle cells with AGEs inhibited their ability to adhere and increased cell membrane permeability. CONCLUSIONS This study indicates that glycative stress may be a novel pathogenic factor in skeletal muscle dysfunctions by causing loss of membrane integrity and preventing muscle mass gain.
Collapse
Affiliation(s)
- Tatsuro Egawa
- Laboratory of Health and Exercise Sciences, Graduate School of Human and Environmental StudiesKyoto UniversityKyotoJapan
| | - Takeshi Ogawa
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental StudiesKyoto UniversityKyotoJapan
| | - Takumi Yokokawa
- Division of Food Science and Biotechnology, Graduate School of AgricultureKyoto UniversityKyotoJapan
| | - Kohei Kido
- Health and Medical Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)KagawaJapan
- Institute for Physical ActivityFukuoka UniversityFukuokaJapan
| | - Ryota Iyama
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental StudiesKyoto UniversityKyotoJapan
| | - Haiyu Zhao
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental StudiesKyoto UniversityKyotoJapan
| | - Eriko Kurogi
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental StudiesKyoto UniversityKyotoJapan
| | - Katsumasa Goto
- Laboratory of Physiology, Graduate School of Health SciencesToyohashi SOZO UniversityToyohashiJapan
| | - Tatsuya Hayashi
- Laboratory of Sports and Exercise Medicine, Graduate School of Human and Environmental StudiesKyoto UniversityKyotoJapan
| |
Collapse
|
7
|
Davies TW, Watson N, Pilkington JJ, McClelland TJ, Azzopardi G, Pearse RM, Prowle J, Puthucheary Z. Creatine supplementation for optimization of physical function in the patient at risk of functional disability: A systematic review and meta-analysis. JPEN J Parenter Enteral Nutr 2024; 48:389-405. [PMID: 38417175 DOI: 10.1002/jpen.2607] [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: 12/12/2023] [Accepted: 01/22/2024] [Indexed: 03/01/2024]
Abstract
BACKGROUND The efficacy of creatine replacement through supplementation for the optimization of physical function in the population at risk of functional disability is unclear. METHODS We conducted a systematic literature search of MEDLINE, EMBASE, the Cochrane Library, and CINAHL from inception to November 2022. Studies included were randomized controlled trials (RCTs) comparing creatine supplementation with placebos in older adults and adults with chronic disease. The primary outcome was physical function measured by the sit-to-stand test after pooling data using random-effects modeling. We also performed a Bayesian meta-analysis to describe the treatment effect in probability terms. Secondary outcomes included other measures of physical function, muscle function, and body composition. The risk of bias was assessed using the Cochrane risk-of-bias tool. RESULTS We identified 33 RCTs, comprising 1076 participants. From six trials reporting the primary outcome, the pooled standardized mean difference (SMD) was 0.51 (95% confidence interval [CI]: 0.01-1.00; I2 = 62%; P = 0.04); using weakly informative priors, the posterior probability that creatine supplementation improves physical function was 66.7%. Upper-body muscle strength (SMD: 0.25; 95% CI: 0.06-0.44; I2 = 0%; P = 0.01), handgrip strength (SMD 0.23; 95% CI: 0.01-0.45; I2 = 0%; P = 0.04), and lean tissue mass (MD 1.08 kg; 95% CI: 0.77-1.38; I2 = 26%; P < 0.01) improved with creatine supplementation. The quality of evidence for all outcomes was low or very low because of a high risk of bias. CONCLUSION Creatine supplementation improves sit-to-stand performance, muscle function, and lean tissue mass. It is crucial to conduct high-quality prospective RCTs to confirm these hypotheses (PROSPERO number, CRD42023354929).
Collapse
Affiliation(s)
- Thomas W Davies
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Naomi Watson
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Adult Critical Care Unit, Royal London Hospital, London, UK
| | - James J Pilkington
- Centre for Bioscience, Manchester Metropolitan University, John Dalton Building, Manchester, UK
| | - Thomas J McClelland
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Giada Azzopardi
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Rupert M Pearse
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Adult Critical Care Unit, Royal London Hospital, London, UK
| | - John Prowle
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Zudin Puthucheary
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
- Adult Critical Care Unit, Royal London Hospital, London, UK
| |
Collapse
|
8
|
Chen Y, Wu J. Aging-Related Sarcopenia: Metabolic Characteristics and Therapeutic Strategies. Aging Dis 2024:AD.2024.0407. [PMID: 38739945 DOI: 10.14336/ad.2024.0407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/07/2024] [Indexed: 05/16/2024] Open
Abstract
The proportion of the elderly population is gradually increasing as a result of medical care advances, leading to a subsequent surge in geriatric diseases that significantly impact quality of life and pose a substantial healthcare burden. Sarcopenia, characterized by age-related decline in skeletal muscle mass and quality, affects a considerable portion of older adults, particularly the elderly, and can result in adverse outcomes such as frailty, fractures, bedridden, hospitalization, and even mortality. Skeletal muscle aging is accompanied by underlying metabolic changes. Therefore, elucidating these metabolic profiles and specific mechanisms holds promise for informing prevention and treatment strategies for sarcopenia. This review provides a comprehensive overview of the key metabolites identified in current clinical studies on sarcopenia and their potential pathophysiological alterations in metabolic activity. Besides, we examine potential therapeutic strategies for sarcopenia from a perspective focused on metabolic regulation.
Collapse
|
9
|
Krššák M. Editorial for "Multi-Parametric Ageing Study Across Adulthood in the Leg Through Quantitative MR Imaging, 1H Spectroscopy and 31P Spectroscopy at 3T". J Magn Reson Imaging 2024. [PMID: 38593216 DOI: 10.1002/jmri.29380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024] Open
Affiliation(s)
- Martin Krššák
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- High Field MR Centre, Department of Biomedical Imaging and Image Guided Therapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
10
|
Han P, Yuan C, Chen X, Hu Y, Hu X, Xu Z, Guo Q. Metabolic signatures and potential biomarkers of sarcopenia in suburb-dwelling older Chinese: based on untargeted GC-MS and LC-MS. Skelet Muscle 2024; 14:4. [PMID: 38454497 PMCID: PMC10921582 DOI: 10.1186/s13395-024-00337-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Untargeted metabolomics can be used to expand our understanding of the pathogenesis of sarcopenia. However, the metabolic signatures of sarcopenia patients have not been thoroughly investigated. Herein, we explored metabolites associated with sarcopenia by untargeted gas chromatography (GC)/liquid chromatography (LC)-mass spectrometry (MS) and identified possible diagnostic markers. METHODS Forty-eight elderly subjects with sarcopenia were age and sex matched with 48 elderly subjects without sarcopenia. We first used untargeted GC/LC-MS to analyze the plasma of these participants and then combined it with a large number of multivariate statistical analyses to analyze the data. Finally, based on a multidimensional analysis of the metabolites, the most critical metabolites were considered to be biomarkers of sarcopenia. RESULTS According to variable importance in the project (VIP > 1) and the p-value of t-test (p < 0.05), a total of 55 metabolites by GC-MS and 85 metabolites by LC-MS were identified between sarcopenia subjects and normal controls, and these were mostly lipids and lipid-like molecules. Among the top 20 metabolites, seven phosphatidylcholines, seven lysophosphatidylcholines (LysoPCs), phosphatidylinositol, sphingomyelin, palmitamide, L-2-amino-3-oxobutanoic acid, and palmitic acid were downregulated in the sarcopenia group; only ethylamine was upregulated. Among that, three metabolites of LysoPC(17:0), L-2-amino-3-oxobutanoic acid, and palmitic acid showed very good prediction capacity with AUCs of 0.887 (95% CI = 0.817-0.957), 0.836 (95% CI = 0.751-0.921), and 0.805 (95% CI = 0.717-0.893), respectively. CONCLUSIONS These findings show that metabonomic analysis has great potential to be applied to sarcopenia. The identified metabolites could be potential biomarkers and could be used to study sarcopenia pathomechanisms.
Collapse
Affiliation(s)
- Peipei Han
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- College of Rehabilitation Sciences, Pudong New Area, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, China
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
| | - Chunhua Yuan
- Comprehensive Surgical Rehabilitation Ward, Shanghai Health Rehabilitation Hospital, Shanghai, China
| | - Xiaoyu Chen
- College of Rehabilitation Sciences, Pudong New Area, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, China
| | - Yuanqing Hu
- College of Rehabilitation Sciences, Pudong New Area, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, China
| | - Xiaodan Hu
- College of Rehabilitation Sciences, Pudong New Area, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, China
| | - Zhangtao Xu
- College of Rehabilitation Sciences, Pudong New Area, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, China
| | - Qi Guo
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.
- College of Rehabilitation Sciences, Pudong New Area, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, China.
| |
Collapse
|
11
|
Vasilevska Nikodinovska V, Ivanoski S. Sarcopenia, More Than Just Muscle Atrophy: Imaging Methods for the Assessment of Muscle Quantity and Quality. ROFO-FORTSCHR RONTG 2023; 195:777-789. [PMID: 37160148 DOI: 10.1055/a-2057-0205] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
BACKGROUND Sarcopenia, a progressive reduction of muscle mass and function, is associated with adverse outcomes in the elderly. Sarcopenia and muscle atrophy are not equal processes. Low muscle strength in association with muscle quantity/quality reduction is currently the optimal method for assessing sarcopenia. There is a practical need for indirect measurement of muscle strength using state-of-the-art imaging techniques. METHODS The following provides a narrative, broad review of all current imaging techniques for evaluating muscles and identifying sarcopenia, including DEXA, CT, MRI, and high-resolution ultrasound, their main strengths, weaknesses, and possible solutions to problems regarding each technique. RESULTS AND CONCLUSION Well-recognized imaging methods for the assessment of muscle mass are explained, including evaluation with DEXA, CT, and MRI muscle quantity assessment, ultrasound evaluation of muscle thickness and CSA, and their correlations with established muscle mass calculation methods. A special focus is on imaging methods for muscle quality evaluation. Several innovative and promising techniques that are still in the research phase but show potential in the assessment of different properties of muscle quality, including MRI DIXON sequences, MRI spectroscopy, Diffusion Tensor Imaging, ultrasound echo intensity, ultrasound elastography, and speed-of-sound ultrasound imaging are briefly mentioned. KEY POINTS · Sarcopenia definition includes low muscle strength and low muscle quantity/quality.. · DEXA is a low-radiation method for whole-body composition measurement in a single image.. · CT has established cut-off values for muscle quality/quantity evaluation and sarcopenia diagnosis.. · MRI is the most sophisticated muscle quality assessment method capable of evaluating myosteatosis, myofibrosis, and microstructure.. · Ultrasound can evaluate muscle quality, including tissue architecture, and elasticity with excellent spatial resolution.. CITATION FORMAT · Vasilevska Nikodinovska V, Ivanoski S, . Sarcopenia, More Than Just Muscle Atrophy: Imaging Methods for the Assessment of Muscle Quantity and Quality. Fortschr Röntgenstr 2023; 195: 777 - 789.
Collapse
Affiliation(s)
| | - Slavcho Ivanoski
- Diagnostic Radiology, St. Erasmo Hospital, Ohrid, North Macedonia
| |
Collapse
|
12
|
Hinkley JM, Yu G, Standley RA, Distefano G, Tolstikov V, Narain NR, Greenwood BP, Karmacharya S, Kiebish MA, Carnero EA, Yi F, Vega RB, Goodpaster BH, Gardell SJ, Coen PM. Exercise and ageing impact the kynurenine/tryptophan pathway and acylcarnitine metabolite pools in skeletal muscle of older adults. J Physiol 2023; 601:2165-2188. [PMID: 36814134 PMCID: PMC10278663 DOI: 10.1113/jp284142] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
Exercise-induced perturbation of skeletal muscle metabolites is a probable mediator of long-term health benefits in older adults. Although specific metabolites have been identified to be impacted by age, physical activity and exercise, the depth of coverage of the muscle metabolome is still limited. Here, we investigated resting and exercise-induced metabolite distribution in muscle from well-phenotyped older adults who were active or sedentary, and a group of active young adults. Percutaneous biopsies of the vastus lateralis were obtained before, immediately after and 3 h following a bout of endurance cycling. Metabolite profile in muscle biopsies was determined by tandem mass spectrometry. Mitochondrial energetics in permeabilized fibre bundles was assessed by high resolution respirometry and fibre type proportion was assessed by immunohistology. We found that metabolites of the kynurenine/tryptophan pathway were impacted by age and activity. Specifically, kynurenine was elevated in muscle from older adults, whereas downstream metabolites of kynurenine (kynurenic acid and NAD+ ) were elevated in muscle from active adults and associated with cardiorespiratory fitness and muscle oxidative capacity. Acylcarnitines, a potential marker of impaired metabolic health, were elevated in muscle from physically active participants. Surprisingly, despite baseline group difference, acute exercise-induced alterations in whole-body substrate utilization, as well as muscle acylcarnitines and ketone bodies, were remarkably similar between groups. Our data identified novel muscle metabolite signatures that associate with the healthy ageing phenotype provoked by physical activity and reveal that the metabolic responsiveness of muscle to acute endurance exercise is retained [NB]:AUTHOR: Please ensure that the appropriate material has been provide for Table S2, as well as for Figures S1 to S7, as also cited in the text with age regardless of activity levels. KEY POINTS: Kynurenine/tryptophan pathway metabolites were impacted by age and physical activity in human muscle, with kynurenine elevated in older muscle, whereas downstream products kynurenic acid and NAD+ were elevated in exercise-trained muscle regardless of age. Acylcarnitines, a marker of impaired metabolic health when heightened in circulation, were elevated in exercise-trained muscle of young and older adults, suggesting that muscle act as a metabolic sink to reduce the circulating acylcarnitines observed with unhealthy ageing. Despite the phenotypic differences, the exercise-induced response of various muscle metabolite pools, including acylcarnitine and ketone bodies, was similar amongst the groups, suggesting that older adults can achieve the metabolic benefits of exercise seen in young counterparts.
Collapse
Affiliation(s)
- J. Matthew Hinkley
- AdventHealth Translational Research Institute, AdventHealth Orlando, Orlando, FL, 32804, USA
| | - GongXin Yu
- AdventHealth Translational Research Institute, AdventHealth Orlando, Orlando, FL, 32804, USA
| | - Robert A. Standley
- AdventHealth Translational Research Institute, AdventHealth Orlando, Orlando, FL, 32804, USA
| | - Giovanna Distefano
- AdventHealth Translational Research Institute, AdventHealth Orlando, Orlando, FL, 32804, USA
| | | | | | | | | | | | - Elvis Alvarez Carnero
- AdventHealth Translational Research Institute, AdventHealth Orlando, Orlando, FL, 32804, USA
| | - Fanchao Yi
- AdventHealth Translational Research Institute, AdventHealth Orlando, Orlando, FL, 32804, USA
| | - Rick B. Vega
- AdventHealth Translational Research Institute, AdventHealth Orlando, Orlando, FL, 32804, USA
| | - Bret H. Goodpaster
- AdventHealth Translational Research Institute, AdventHealth Orlando, Orlando, FL, 32804, USA
| | - Stephen J. Gardell
- AdventHealth Translational Research Institute, AdventHealth Orlando, Orlando, FL, 32804, USA
| | - Paul M. Coen
- AdventHealth Translational Research Institute, AdventHealth Orlando, Orlando, FL, 32804, USA
| |
Collapse
|
13
|
Lin Z, Li H, He C, Yang M, Chen H, Yang X, Zhuo J, Shen W, Hu Z, Pan L, Wei X, Lu D, Zheng S, Xu X. Metabolomic biomarkers for the diagnosis and post-transplant outcomes of AFP negative hepatocellular carcinoma. Front Oncol 2023; 13:1072775. [PMID: 36845695 PMCID: PMC9947281 DOI: 10.3389/fonc.2023.1072775] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
Background Early diagnosis for α-fetoprotein (AFP) negative hepatocellular carcinoma (HCC) remains a critical problem. Metabolomics is prevalently involved in the identification of novel biomarkers. This study aims to identify new and effective markers for AFP negative HCC. Methods In total, 147 patients undergoing liver transplantation were enrolled from our hospital, including liver cirrhosis patients (LC, n=25), AFP negative HCC patients (NEG, n=44) and HCC patients with AFP over 20 ng/mL (POS, n=78). 52 Healthy volunteers (HC) were also recruited in this study. Metabolomic profiling was performed on the plasma of those patients and healthy volunteers to select candidate metabolomic biomarkers. A novel diagnostic model for AFP negative HCC was established based on Random forest analysis, and prognostic biomarkers were also identified. Results 15 differential metabolites were identified being able to distinguish NEG group from both LC and HC group. Random forest analysis and subsequent Logistic regression analysis showed that PC(16:0/16:0), PC(18:2/18:2) and SM(d18:1/18:1) are independent risk factor for AFP negative HCC. A three-marker model of Metabolites-Score was established for the diagnosis of AFP negative HCC patients with an area under the time-dependent receiver operating characteristic curve (AUROC) of 0.913, and a nomogram was then established as well. When the cut-off value of the score was set at 1.2895, the sensitivity and specificity for the model were 0.727 and 0.92, respectively. This model was also applicable to distinguish HCC from cirrhosis. Notably, the Metabolites-Score was not correlated to tumor or body nutrition parameters, but difference of the score was statistically significant between different neutrophil-lymphocyte ratio (NLR) groups (≤5 vs. >5, P=0.012). Moreover, MG(18:2/0:0/0:0) was the only prognostic biomarker among 15 metabolites, which is significantly associated with tumor-free survival of AFP negative HCC patients (HR=1.160, 95%CI 1.012-1.330, P=0.033). Conclusion The established three-marker model and nomogram based on metabolomic profiling can be potential non-invasive tool for the diagnosis of AFP negative HCC. The level of MG(18:2/0:0/0:0) exhibits good prognosis prediction performance for AFP negative HCC.
Collapse
Affiliation(s)
- Zuyuan Lin
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China,Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Health Commission Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Huigang Li
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Health Commission Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Chiyu He
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Health Commission Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Modan Yang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China,Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Health Commission Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Hao Chen
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Health Commission Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Xinyu Yang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China,Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Health Commission Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Jianyong Zhuo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Institute of Organ Transplantation, Zhejiang University, Hangzhou, China
| | - Wei Shen
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Health Commission Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Zhihang Hu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Health Commission Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Linhui Pan
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Institute of Organ Transplantation, Zhejiang University, Hangzhou, China
| | - Xuyong Wei
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Health Commission Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Di Lu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Institute of Organ Transplantation, Zhejiang University, Hangzhou, China
| | - Shusen Zheng
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Health Commission Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China,Institute of Organ Transplantation, Zhejiang University, Hangzhou, China,Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, China
| | - Xiao Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,National Health Commission Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China,Institute of Organ Transplantation, Zhejiang University, Hangzhou, China,Zhejiang University School of Medicine, Hangzhou, China,*Correspondence: Xiao Xu,
| |
Collapse
|
14
|
Jones RL, Paul L, Steultjens MPM, Smith SL. Biomarkers associated with lower limb muscle function in individuals with sarcopenia: a systematic review. J Cachexia Sarcopenia Muscle 2022; 13:2791-2806. [PMID: 35977879 PMCID: PMC9745467 DOI: 10.1002/jcsm.13064] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/10/2022] [Accepted: 07/04/2022] [Indexed: 12/15/2022] Open
Abstract
Lower limb muscle dysfunction is a key driver for impaired physical capacity and frailty status, both characteristics of sarcopenia. Sarcopenia is the key pathway between frailty and disability. Identifying biological markers for early diagnosis, treatment, and prevention may be key to early intervention and prevention of disability particularly mobility issues. To identify biological markers associated with lower limb muscle (dys)function in adults with sarcopenia, a systematic literature search was conducted in AMED, CINAHL, Cochrane Library, EMBASE, Medline, PubMed, Scopus, SPORTDiscus, and Web of Science databases from inception to 17 November 2021. Title, abstract, and full-text screening, data extraction, and methodological quality assessment were performed by two reviewers independently and verified by a third reviewer. Depending on available data, associations are reported as either Pearson's correlations, regression R2 or partial R2 , P value, and sample size (n). Twenty eligible studies including 3306 participants were included (females: 79%, males: 15%, unreported: 6%; mean age ranged from 53 to 92 years) with 36% in a distinct sarcopenic subgroup (females: 73%, males: 19%, unreported: 8%; mean age range 55-92 years). A total of 119 biomarkers were reported, categorized into: genetic and microRNAs (n = 64), oxidative stress (n = 10), energy metabolism (n = 18), inflammation (n = 7), enzyme (n = 4), hormone (n = 7), bone (n = 3), vitamin (n = 2), and cytokine (n = 4) markers) and seven lower limb muscle measures predominately focused on strength. Seven studies reported associations between lower limb muscle measures including (e.g. power, force, and torque) and biomarkers. In individuals with sarcopenia, muscle strength was positively associated with free testosterone (r = 0.40, P = 0.01; n = 46). In analysis with combined sarcopenic and non-sarcopenic individuals, muscle strength was positively associated with combined genetic and methylation score (partial R2 = 0.122, P = 0.03; n = 48) and negatively associated with sarcopenia-driven methylation score (partial R2 = 0.401, P < 0.01; n = 48). Biomarkers related to genetics (R2 = 0.001-0.014, partial R2 = 0.013-0.122, P > 0.05; n = 48), oxidative stress (r = 0.061, P > 0.05; n ≥ 77), hormone (r = 0.01, ρ = 0.052 p > 0.05, n ≥ 46) and combined protein, oxidative stress, muscle performance, and hormones (R2 = 22.0, P > 0.05; n ≥ 82) did not report significant associations with lower limb muscle strength. Several biomarkers demonstrated associations with lower limb muscle dysfunction. The current literature remains difficult to draw clear conclusions on the relationship between biomarkers and lower limb muscle dysfunction in adults with sarcopenia. Heterogeneity of biomarkers and lower limb muscle function precluded direct comparison. Use of international classification of sarcopenia and a set of core standardized outcome measures should be adopted to aid future investigation and recommendations to be made.
Collapse
Affiliation(s)
- Rebecca Louise Jones
- Institute for Sport and Physical Activity Research, School of Sport Science and Physical Activity, University of Bedfordshire, Bedford, UK.,Health Advancement Research Team (HART), School of Sport and Exercise Science, University of Lincoln, Lincoln, UK
| | - Lorna Paul
- Centre for Living, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Martijn P M Steultjens
- Centre for Living, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Stephanie Louise Smith
- Centre for Living, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, UK.,Academic Rheumatology, Division of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham, UK.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
15
|
Al Saedi A, Debruin DA, Hayes A, Hamrick M. Lipid metabolism in sarcopenia. Bone 2022; 164:116539. [PMID: 36007811 DOI: 10.1016/j.bone.2022.116539] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/10/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022]
Abstract
Sarcopenia is an age-related disease associated with loss of muscle mass and strength. This geriatric syndrome predisposes elderly individuals to a disability, falls, fractures, and death. Fat infiltration in muscle is one of the hallmarks of sarcopenia and aging. Alterations in fatty acid (FA) metabolism are evident in aging, type 2 diabetes, and obesity, with the accumulation of lipids inside muscle cells contributing to muscle insulin resistance and ceramide accumulation. These lipids include diacylglycerol, lipid droplets, intramyocellular lipids, intramuscular triglycerides, and polyunsaturated fatty acids (PUFAs). In this review, we examine the regulation of lipid metabolism in skeletal muscle, including lipid metabolization and storage, intervention, and the types of lipases expressed in skeletal muscle responsible for the breakdown of adipose triglyceride fats. In addition, we address the role of FAs in sarcopenia and the potential benefits of PUFAs.
Collapse
Affiliation(s)
- Ahmed Al Saedi
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, VIC, Australia; Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St. Albans, VIC, Australia; Institute of Health and Sport (IHeS), Victoria University, Melbourne, VIC, Australia.
| | - Danielle A Debruin
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, VIC, Australia; Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St. Albans, VIC, Australia; Institute of Health and Sport (IHeS), Victoria University, Melbourne, VIC, Australia
| | - Alan Hayes
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, VIC, Australia; Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St. Albans, VIC, Australia; Institute of Health and Sport (IHeS), Victoria University, Melbourne, VIC, Australia
| | - Mark Hamrick
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University, Laney Walker Blvd. CB2915, Augusta, GA 30912, USA
| |
Collapse
|
16
|
Stephenson MC, Krishna L, Pannir Selvan RM, Tai YK, Kit Wong CJ, Yin JN, Toh SJ, Torta F, Triebl A, Fröhlich J, Beyer C, Li JZ, Tan SS, Wong CK, Chinnasamy D, Pakkiri LS, Lee Drum C, Wenk MR, Totman JJ, Franco-Obregón A. Magnetic field therapy enhances muscle mitochondrial bioenergetics and attenuates systemic ceramide levels following ACL reconstruction: Southeast Asian randomized-controlled pilot trial. J Orthop Translat 2022; 35:99-112. [PMID: 36262374 PMCID: PMC9574347 DOI: 10.1016/j.jot.2022.09.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/06/2022] Open
Abstract
Background Metabolic disruption commonly follows Anterior Cruciate Ligament Reconstruction (ACLR) surgery. Brief exposure to low amplitude and frequency pulsed electromagnetic fields (PEMFs) has been shown to promote in vitro and in vivo murine myogeneses via the activation of a calcium–mitochondrial axis conferring systemic metabolic adaptations. This randomized-controlled pilot trial sought to detect local changes in muscle structure and function using MRI, and systemic changes in metabolism using plasma biomarker analyses resulting from ACLR, with or without accompanying PEMF therapy. Methods 20 patients requiring ACLR were randomized into two groups either undergoing PEMF or sham exposure for 16 weeks following surgery. The operated thighs of 10 patients were exposed weekly to PEMFs (1 mT for 10 min) for 4 months following surgery. Another 10 patients were subjected to sham exposure and served as controls to allow assessment of the metabolic repercussions of ACLR and PEMF therapy. Blood samples were collected prior to surgery and at 16 weeks for plasma analyses. Magnetic resonance data were acquired at 1 and 16 weeks post-surgery using a Siemens 3T Tim Trio system. Phosphorus (31P) Magnetic Resonance Spectroscopy (MRS) was utilized to monitor changes in high-energy phosphate metabolism (inorganic phosphate (Pi), adenosine triphosphate (ATP) and phosphocreatine (PCr)) as well as markers of membrane synthesis and breakdown (phosphomonoesters (PME) and phosphodiester (PDE)). Quantitative Magnetization Transfer (qMT) imaging was used to elucidate changes in the underlying tissue structure, with T1-weighted and 2-point Dixon imaging used to calculate muscle volumes and muscle fat content. Results Improvements in markers of high-energy phosphate metabolism including reductions in ΔPi/ATP, Pi/PCr and (Pi + PCr)/ATP, and membrane kinetics, including reductions in PDE/ATP were detected in the PEMF-treated cohort relative to the control cohort at study termination. These were associated with reductions in the plasma levels of certain ceramides and lysophosphatidylcholine species. The plasma levels of biomarkers predictive of muscle regeneration and degeneration, including osteopontin and TNNT1, respectively, were improved, whilst changes in follistatin failed to achieve statistical significance. Liquid chromatography with tandem mass spectrometry revealed reductions in small molecule biomarkers of metabolic disruption, including cysteine, homocysteine, and methionine in the PEMF-treated cohort relative to the control cohort at study termination. Differences in measurements of force, muscle and fat volumes did not achieve statistical significance between the cohorts after 16 weeks post-ACLR. Conclusion The detected changes suggest improvements in systemic metabolism in the post-surgical PEMF-treated cohort that accords with previous preclinical murine studies. PEMF-based therapies may potentially serve as a manner to ameliorate post-surgery metabolic disruptions and warrant future examination in more adequately powered clinical trials. The Translational Potential of this Article Some degree of physical immobilisation must inevitably follow orthopaedic surgical intervention. The clinical paradox of such a scenario is that the regenerative potential of the muscle mitochondrial pool is silenced. The unmet need was hence a manner to maintain mitochondrial activation when movement is restricted and without producing potentially damaging mechanical stress. PEMF-based therapies may satisfy the requirement of non-invasively activating the requisite mitochondrial respiration when mobility is restricted for improved metabolic and regenerative recovery.
Collapse
Affiliation(s)
- Mary C. Stephenson
- Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,Corresponding author. Centre for Translational MR Research, Yong Loo Lin School of Medicine, Tahir Foundation Building, 13-03, MD1, National University of Singapore, Singapore, 117549.
| | - Lingaraj Krishna
- Division of Sports Medicine and Surgery, Department of Orthopaedic Surgery, National University Hospital, National University Health System, Singapore
| | - Rina Malathi Pannir Selvan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, Singapore
| | - Yee Kit Tai
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, Singapore,Corresponding author. Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block Level 8, 1E Kent Ridge Road, Singapore, 119228.
| | - Craig Jun Kit Wong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, Singapore
| | - Jocelyn Naixin Yin
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, Singapore
| | - Shi-Jie Toh
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore,Precision Medicine Translational Research Program, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Alexander Triebl
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore
| | | | - Christian Beyer
- Centre Suisse d'électronique et de Microtechnique, CSEM SA, Neuchatel, Switzerland
| | - Jing Ze Li
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sara S. Tan
- Division of Sports Medicine and Surgery, Department of Orthopaedic Surgery, National University Hospital, National University Health System, Singapore
| | - Chun-Kit Wong
- Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Duraimurugan Chinnasamy
- National University Hospital, Department of Rehabilitation Centre, National University Health System, Singapore
| | - Leroy Sivappiragasam Pakkiri
- Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS), Singapore,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chester Lee Drum
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS), Singapore,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Markus R. Wenk
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore,Precision Medicine Translational Research Program, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - John J. Totman
- Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,Academic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, Singapore,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,Institute for Health Innovation & Technology, iHealthtech, National University of Singapore, Singapore,NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore,Competence Center for Applied Biotechnology and Molecular Medicine, University of Zürich, Zürich, Switzerland,Corresponding author. Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block Level 8, 1E Kent Ridge Road, Singapore, 119228.
| |
Collapse
|
17
|
Mirzai S, Eck BL, Chen PH, Estep JD, Tang WHW. Current Approach to the Diagnosis of Sarcopenia in Heart Failure: A Narrative Review on the Role of Clinical and Imaging Assessments. Circ Heart Fail 2022; 15:e009322. [PMID: 35924562 PMCID: PMC9588634 DOI: 10.1161/circheartfailure.121.009322] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sarcopenia has been established as a predictor of poor outcomes in various clinical settings. It is particularly prevalent in heart failure, a clinical syndrome that poses significant challenges to health care worldwide. Despite this, sarcopenia remains overlooked and undertreated in cardiology practice. Understanding the currently proposed diagnostic process is paramount for the early detection and treatment of sarcopenia to mitigate downstream adverse health outcomes.
Collapse
Affiliation(s)
- Saeid Mirzai
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH
| | - Brendan L. Eck
- Section of Musculoskeletal Imaging, Imaging Institute, Cleveland Clinic, Cleveland, OH
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH
| | - Po-Hao Chen
- Section of Musculoskeletal Imaging, Imaging Institute, Cleveland Clinic, Cleveland, OH
| | - Jerry D. Estep
- Department of Cardiology, Cleveland Clinic Florida, Weston, FL
| | - W. H. Wilson Tang
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
18
|
Molecular Mechanisms of Inflammation in Sarcopenia: Diagnosis and Therapeutic Update. Cells 2022; 11:cells11152359. [PMID: 35954203 PMCID: PMC9367570 DOI: 10.3390/cells11152359] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 01/10/2023] Open
Abstract
Sarcopenia is generally an age-related condition that directly impacts the quality of life. It is also related to chronic diseases such as metabolic dysfunction associated with diabetes and obesity. This means that everyone will be vulnerable to sarcopenia at some point in their life. Research to find the precise molecular mechanisms implicated in this condition can increase knowledge for the better prevention, diagnosis, and treatment of sarcopenia. Our work gathered the most recent research regarding inflammation in sarcopenia and new therapeutic agents proposed to target its consequences in pyroptosis and cellular senescence. Finally, we compared dual X-ray absorptiometry (DXA), magnetic resonance imaging (MRI), and ultrasound (US) as imaging techniques to diagnose and follow up on sarcopenia, indicating their respective advantages and disadvantages. Our goal is for the scientific evidence presented here to help guide future research to understand the molecular mechanisms involved in sarcopenia, new treatment strategies, and their translation into clinical practice.
Collapse
|
19
|
Ubaida-Mohien C, Spendiff S, Lyashkov A, Moaddel R, MacMillan NJ, Filion ME, Morais JA, Taivassalo T, Ferrucci L, Hepple RT. Unbiased proteomics, histochemistry, and mitochondrial DNA copy number reveal better mitochondrial health in muscle of high-functioning octogenarians. eLife 2022; 11:e74335. [PMID: 35404238 PMCID: PMC9090325 DOI: 10.7554/elife.74335] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Master athletes (MAs) prove that preserving a high level of physical function up to very late in life is possible, but the mechanisms responsible for their high function remain unclear. Methods We performed muscle biopsies in 15 octogenarian world-class track and field MAs and 14 non-athlete age/sex-matched controls (NA) to provide insights into mechanisms for preserving function in advanced age. Muscle samples were assessed for respiratory compromised fibers, mitochondrial DNA (mtDNA) copy number, and proteomics by liquid-chromatography mass spectrometry. Results MA exhibited markedly better performance on clinical function tests and greater cross-sectional area of the vastus lateralis muscle. Proteomics analysis revealed marked differences, where most of the ~800 differentially represented proteins in MA versus NA pertained to mitochondria structure/function such as electron transport capacity (ETC), cristae formation, mitochondrial biogenesis, and mtDNA-encoded proteins. In contrast, proteins from the spliceosome complex and nuclear pore were downregulated in MA. Consistent with proteomics data, MA had fewer respiratory compromised fibers, higher mtDNA copy number, and an increased protein ratio of the cristae-bound ETC subunits relative to the outer mitochondrial membrane protein voltage-dependent anion channel. There was a substantial overlap of proteins overrepresented in MA versus NA with proteins that decline with aging and that are higher in physically active than sedentary individuals. However, we also found 176 proteins related to mitochondria that are uniquely differentially expressed in MA. Conclusions We conclude that high function in advanced age is associated with preserving mitochondrial structure/function proteins, with underrepresentation of proteins involved in the spliceosome and nuclear pore complex. Whereas many of these differences in MA appear related to their physical activity habits, others may reflect unique biological (e.g., gene, environment) mechanisms that preserve muscle integrity and function with aging. Funding Funding for this study was provided by operating grants from the Canadian Institutes of Health Research (MOP 84408 to TT and MOP 125986 to RTH). This work was supported in part by the Intramural Research Program of the National Institute on Aging, NIH, Baltimore, MD, USA.
Collapse
Affiliation(s)
- Ceereena Ubaida-Mohien
- Intramural Research Program, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Sally Spendiff
- Research Institute, Children's Hospital of Eastern OntarioOttawaCanada
| | - Alexey Lyashkov
- Intramural Research Program, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Ruin Moaddel
- Intramural Research Program, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Norah J MacMillan
- Research Institute of the McGill University Health Centre, McGill UniversityMontrealCanada
| | - Marie-Eve Filion
- Research Institute of the McGill University Health Centre, McGill UniversityMontrealCanada
| | - Jose A Morais
- Research Institute of the McGill University Health Centre, McGill UniversityMontrealCanada
| | - Tanja Taivassalo
- Department of Physical Therapy, University of FloridaGainesvilleUnited States
| | - Luigi Ferrucci
- Intramural Research Program, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Russell T Hepple
- Department of Physical Therapy, University of FloridaGainesvilleUnited States
- Department of Physiology and Functional Genomics, University of FloridaGainesvilleUnited States
| |
Collapse
|
20
|
Tsai J, Wang S, Chang C, Chen C, Wen C, Chen G, Kuo C, Tseng YJ, Chen C. Identification of traumatic acid as a potential plasma biomarker for sarcopenia using a metabolomics-based approach. J Cachexia Sarcopenia Muscle 2022; 13:276-286. [PMID: 34939349 PMCID: PMC8818620 DOI: 10.1002/jcsm.12895] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 08/30/2021] [Accepted: 11/21/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The pathogenesis of sarcopenia is complex and has not been well explored. Identifying biomarkers is a promising strategy for exploring the mechanism of sarcopenia. This study aimed to identify potential biomarkers of sarcopenia through a metabolomic analysis of plasma metabolites in elderly subjects (≥65 years of age) vs. younger adults (<65 years of age). METHODS Of the 168 candidates in the Comprehensive Geriatric Assessment and Frailty Study of Elderly Outpatients, 24 elderly subjects (≥65 years of age) with sarcopenia were age and sex matched with 24 elderly subjects without sarcopenia. In addition, 24 younger adults were recruited for comparison. Muscle strength, gait speed, and metabolic and inflammatory parameters, including plasma tumour necrosis factor-α, C-reactive protein, irisin, and growth differentiation factor 15 (GDF-15) levels were assessed. Metabolomic analysis was carried out using the plasma metabolites. RESULTS Seventy-two participants were enrolled, including 10 (41.6%) men and 14 (58.3%) women in both groups of elderly subjects. The median ages of elderly subjects with and without sarcopenia were 82 (range: 67-88) and 81.5 (range: 67-87) years, respectively. Among the 242 plasma metabolic peaks analysed among these three groups, traumatic acid was considered as a sarcopenia-related metabolite. The plasma traumatic acid signal intensity level was significantly higher in elderly subjects with sarcopenia than in elderly subjects without sarcopenia [591.5 (inter-quartile range, IQR: 491.5-664.5) vs. 430.0 (IQR: 261.0-599.5), P = 0.0063]. The plasma concentrations of traumatic acid were 15.8 (IQR: 11.5-21.7), 21.1 (IQR: 16.0-25.8), and 24.3 (IQR: 18.0-29.5) ppb in younger adults [age range: 23-37 years, 12 (50%) men], elderly subjects without sarcopenia, and elderly subjects with sarcopenia, respectively, thereby depicting an increasing tendency (P for trend = 0.034). This pattern was similar to that of GDF-15, a recognized sarcopenia-related factor. Plasma traumatic acid concentrations were also positively correlated with the presence of hypertension (r = 0.25, P = 0.034), glucose AC (r = 0.34, P = 0.0035), creatinine (r = 0.40, P = 0.0006), and GDF-15 levels (r = 0.25, P = 0.0376), but negatively correlated with the Modification of Diet in Renal Disease-simplify-glomerular filtration rate (r = -0.50, P < 0.0001). Similarly, plasma GDF-15 concentrations were associated with these factors. CONCLUSIONS Traumatic acid might represent a potential plasma biomarker of sarcopenia. However, further studies are needed to validate the results and investigate the underlying mechanisms.
Collapse
Affiliation(s)
- Jaw‐Shiun Tsai
- Department of Family MedicineNational Taiwan University Hospital, National Taiwan UniversityTaipeiTaiwan
- Department of Family Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - San‐Yuan Wang
- Master Program in Clinical Genomics and Proteomics, College of PharmacyTaipei Medical UniversityTaipeiTaiwan
| | - Chin‐Hao Chang
- Department of Medical ResearchNational Taiwan University HospitalTaipeiTaiwan
| | - Chin‐Ying Chen
- Department of Family MedicineNational Taiwan University Hospital, National Taiwan UniversityTaipeiTaiwan
- Department of Family Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Chiung‐Jung Wen
- Department of Family Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of Geriatrics and GerontologyNational Taiwan University HospitalTaipeiTaiwan
| | - Guan‐Yuan Chen
- Department and Graduate Institute of Forensic Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Ching‐Hua Kuo
- The Metabolomics Core Laboratory, Center of Genomic MedicineNational Taiwan UniversityTaipeiTaiwan
- School of Pharmacy, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of PharmacyNational Taiwan University Hospital, National Taiwan UniversityTaipeiTaiwan
| | - Y. Jane Tseng
- The Metabolomics Core Laboratory, Center of Genomic MedicineNational Taiwan UniversityTaipeiTaiwan
- School of Pharmacy, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of Computer Science and Information EngineeringNational Taiwan UniversityTaipeiTaiwan
- Graduate Institute of Biomedical Electronics and BioinformaticsNational Taiwan UniversityTaipeiTaiwan
| | - Ching‐Yu Chen
- Department of Family MedicineNational Taiwan University Hospital, National Taiwan UniversityTaipeiTaiwan
- Department of Family Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
21
|
Metabolomics as an Important Tool for Determining the Mechanisms of Human Skeletal Muscle Deconditioning. Int J Mol Sci 2021; 22:ijms222413575. [PMID: 34948370 PMCID: PMC8706620 DOI: 10.3390/ijms222413575] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/28/2022] Open
Abstract
Muscle deconditioning impairs both locomotor function and metabolic health, and is associated with reduced quality life and increased mortality rates. Despite an appreciation of the existence of phenomena such as muscle anabolic resistance, mitophagy, and insulin resistance with age and disease in humans, little is known about the mechanisms responsible for these negative traits. With the complexities surrounding these unknowns and the lack of progress to date in development of effective interventions, there is a need for alternative approaches. Metabolomics is the study of the full array of metabolites within cells or tissues, which collectively constitute the metabolome. As metabolomics allows for the assessment of the cellular metabolic state in response to physiological stimuli, any chronic change in the metabolome is likely to reflect adaptation in the physiological phenotype of an organism. This, therefore, provides a holistic and unbiased approach that could be applied to potentially uncover important novel facets in the pathophysiology of muscle decline in ageing and disease, as well as identifying prognostic markers of those at risk of decline. This review will aim to highlight the current knowledge and potential impact of metabolomics in the study of muscle mass loss and deconditioning in humans and will highlight key areas for future research.
Collapse
|
22
|
Ferrara PJ, Verkerke ARP, Maschek JA, Shahtout JL, Siripoksup P, Eshima H, Johnson JM, Petrocelli JJ, Mahmassani ZS, Green TD, McClung JM, Cox JE, Drummond MJ, Funai K. Low lysophosphatidylcholine induces skeletal muscle myopathy that is aggravated by high-fat diet feeding. FASEB J 2021; 35:e21867. [PMID: 34499764 DOI: 10.1096/fj.202101104r] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/27/2021] [Accepted: 08/09/2021] [Indexed: 12/25/2022]
Abstract
Obesity alters skeletal muscle lipidome and promotes myopathy, but it is unknown whether aberrant muscle lipidome contributes to the reduction in skeletal muscle contractile force-generating capacity. Comprehensive lipidomic analyses of mouse skeletal muscle revealed a very strong positive correlation between the abundance of lysophosphatidylcholine (lyso-PC), a class of lipids that is known to be downregulated with obesity, with maximal tetanic force production. The level of lyso-PC is regulated primarily by lyso-PC acyltransferase 3 (LPCAT3), which acylates lyso-PC to form phosphatidylcholine. Tamoxifen-inducible skeletal muscle-specific overexpression of LPCAT3 (LPCAT3-MKI) was sufficient to reduce muscle lyso-PC content in both standard chow diet- and high-fat diet (HFD)-fed conditions. Strikingly, the assessment of skeletal muscle force-generating capacity ex vivo revealed that muscles from LPCAT3-MKI mice were weaker regardless of diet. Defects in force production were more apparent in HFD-fed condition, where tetanic force production was 40% lower in muscles from LPCAT3-MKI compared to that of control mice. These observations were partly explained by reductions in the cross-sectional area in type IIa and IIx fibers, and signs of muscle edema in the absence of fibrosis. Future studies will pursue the mechanism by which LPCAT3 may alter protein turnover to promote myopathy.
Collapse
Affiliation(s)
- Patrick J Ferrara
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Anthony R P Verkerke
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - J Alan Maschek
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Metabolomics, Mass Spectrometry, and Proteomics Core, University of Utah, Salt Lake City, Utah, USA
| | - Justin L Shahtout
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Piyarat Siripoksup
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Hiroaki Eshima
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of International Tourism, Nagasaki International University, Sasebo, Japan
| | - Jordan M Johnson
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - Jonathan J Petrocelli
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Ziad S Mahmassani
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Thomas D Green
- East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - Joseph M McClung
- East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - James E Cox
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Metabolomics, Mass Spectrometry, and Proteomics Core, University of Utah, Salt Lake City, Utah, USA.,Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| | - Micah J Drummond
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Katsuhiko Funai
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
23
|
Oikawa SY, Brisbois TD, van Loon LJC, Rollo I. Eat like an athlete: insights of sports nutrition science to support active aging in healthy older adults. GeroScience 2021; 43:2485-2495. [PMID: 34283389 PMCID: PMC8599603 DOI: 10.1007/s11357-021-00419-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/06/2021] [Indexed: 11/29/2022] Open
Abstract
Skeletal muscle mass losses with age are associated with negative health consequences, including an increased risk of developing metabolic disease and the loss of independence. Athletes adopt numerous nutritional strategies to maximize the benefits of exercise training and enhance recovery in pursuit of improving skeletal muscle quality, mass, or function. Importantly, many of the principles applied to enhance skeletal muscle health in athletes may be applicable to support active aging and prevent sarcopenia in the healthy (non-clinical) aging population. Here, we discuss the anabolic properties of protein supplementation in addition to ingredients that may enhance the anabolic effects of protein (e.g. omega 3 s, creatine, inorganic nitrate) in older persons. We conclude that nutritional strategies used in pursuit of performance enhancement in athletes are often applicable to improve skeletal muscle health in the healthy older population when implemented as part of a healthy active lifestyle. Further research is required to elucidate the mechanisms by which these nutrients may induce favourable changes in skeletal muscle and to determine the appropriate dosing and timing of nutrient intakes to support active aging.
Collapse
Affiliation(s)
- Sara Y Oikawa
- Gatorade Sports Science Institute, PepsiCo Life Sciences, Global R&D, 5500 34th Street West, Bradenton, FL, 34210, USA.
| | | | - Luc J C van Loon
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University, Maastricht, Netherlands
| | - Ian Rollo
- Gatorade Sports Science Institute, PepsiCo Life Sciences, Global R&D, 5500 34th Street West, Bradenton, FL, 34210, USA.,School of Sports Exercise and Health Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
24
|
Dao T, Green AE, Kim YA, Bae SJ, Ha KT, Gariani K, Lee MR, Menzies KJ, Ryu D. Sarcopenia and Muscle Aging: A Brief Overview. Endocrinol Metab (Seoul) 2020; 35:716-732. [PMID: 33397034 PMCID: PMC7803599 DOI: 10.3803/enm.2020.405] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
The world is facing the new challenges of an aging population, and understanding the process of aging has therefore become one of the most important global concerns. Sarcopenia is a condition which is defined by the gradual loss of skeletal muscle mass and function with age. In research and clinical practice, sarcopenia is recognized as a component of geriatric disease and is a current target for drug development. In this review we define this condition and provide an overview of current therapeutic approaches. We further highlight recent findings that describe key pathophysiological phenotypes of this condition, including alterations in muscle fiber types, mitochondrial function, nicotinamide adenine dinucleotide (NAD+) metabolism, myokines, and gut microbiota, in aged muscle compared to young muscle or healthy aged muscle. The last part of this review examines new therapeutic avenues for promising treatment targets. There is still no accepted therapy for sarcopenia in humans. Here we provide a brief review of the current state of research derived from various mouse models or human samples that provide novel routes for the development of effective therapeutics to maintain muscle health during aging.
Collapse
Affiliation(s)
- Tam Dao
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon,
Korea
| | - Alexander E. Green
- University of Ottawa Eric Poulin Centre for Neuromuscular Disease, Ottawa, ON,
Canada
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences University of Ottawa, Ottawa, ON,
Canada
| | - Yun A Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon,
Korea
| | - Sung-Jin Bae
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan,
Korea
| | - Ki-Tae Ha
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan,
Korea
- Department of Korean Medical Science, Pusan National University School of Korean Medicine, Yangsan,
Korea
| | - Karim Gariani
- Service of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Geneva University Hospitals, Geneva,
Switzerland
- Faculty of Medicine, University of Geneva, Geneva,
Switzerland
| | - Mi-ra Lee
- Department of Social Welfare, Division of Public Service, Dong-Eui University, Busan,
Korea
- Mi-ra Lee, Department of Public Service, Dong-Eui University, 176 Eomgwang-ro, Busanjin-gu, Busan 47340, Korea, Tel: +82-51-890-2038, E-mail:
| | - Keir J. Menzies
- University of Ottawa Eric Poulin Centre for Neuromuscular Disease, Ottawa, ON,
Canada
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences University of Ottawa, Ottawa, ON,
Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON,
Canada
- Keir J. Menzies, Eric Poulin Centre for Neuromuscular Disease, Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada, Tel: +1-613-562-5800, E-mail:
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon,
Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon,
Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul,
Korea
- Corresponding authors: Dongryeol Ryu, Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Korea, Tel: +82-31-299-6138, E-mail:
| |
Collapse
|
25
|
Seo DY, Hwang BG. Effects of exercise training on the biochemical pathways associated with sarcopenia. Phys Act Nutr 2020; 24:32-38. [PMID: 33108716 PMCID: PMC7669465 DOI: 10.20463/pan.2020.0019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
[Purpose] Sarcopenia is considered one of the major causes of disability in the elderly population and is highly associated with aging. Exercise is an essential strategy for improving muscle health while aging and involves multiple metabolic and transcriptional adaptations. Although the beneficial effects of exercise modalities on skeletal muscle structure and function in aging are well recognized, the exact cellular and molecular mechanisms underlying the influence of exercise have not been fully elucidated. [Methods] We summarize the biochemical pathways involved in the progression and pathogenesis of sarcopenia and describe the beneficial effects of exercise training on the relevant signaling pathways associated with sarcopenia. [Results] This study briefly introduces current knowledge on the signaling pathways involved in the development of sarcopenia, effects of aerobic exercise on mitochondria-related parameters and mitochondrial function, and role of resistance exercise in the regulation of muscle protein synthesis against sarcopenia. [Conclusion] This review suggested that the beneficial effects of exercise are still under-explored, and accelerated research will help develop better modalities for the prevention, management, and treatment of sarcopenia.
Collapse
Affiliation(s)
- Dae Yun Seo
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea
| | - Boo Geun Hwang
- Department of Sport Rehabilitation, Tong Myong University, Busan, Republic of Korea
| |
Collapse
|
26
|
Hinkley JM, Coen PM. Muscle phosphorus metabolites in sarcopenia. Aging (Albany NY) 2020; 12:15880-15881. [PMID: 32860671 PMCID: PMC7485693 DOI: 10.18632/aging.104032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 08/21/2020] [Indexed: 12/21/2022]
Affiliation(s)
- J Matthew Hinkley
- AdventHealth Translational Research Institute, Orlando, FL 32804, USA
| | - Paul M Coen
- AdventHealth Translational Research Institute, Orlando, FL 32804, USA
| |
Collapse
|
27
|
Hinkley JM, Cornnell HH, Standley RA, Chen EY, Narain NR, Greenwood BP, Bussberg V, Tolstikov VV, Kiebish MA, Yi F, Vega RB, Goodpaster BH, Coen PM. Older adults with sarcopenia have distinct skeletal muscle phosphodiester, phosphocreatine, and phospholipid profiles. Aging Cell 2020; 19:e13135. [PMID: 32468656 PMCID: PMC7294783 DOI: 10.1111/acel.13135] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/04/2020] [Accepted: 02/23/2020] [Indexed: 12/12/2022] Open
Abstract
The loss of skeletal muscle mass and function with age (sarcopenia) is a critical healthcare challenge for older adults. 31‐phosphorus magnetic resonance spectroscopy (31P‐MRS) is a powerful tool used to evaluate phosphorus metabolite levels in muscle. Here, we sought to determine which phosphorus metabolites were linked with reduced muscle mass and function in older adults. This investigation was conducted across two separate studies. Resting phosphorus metabolites in skeletal muscle were examined by 31P‐MRS. In the first study, fifty‐five older adults with obesity were enrolled and we found that resting phosphocreatine (PCr) was positively associated with muscle volume and knee extensor peak power, while a phosphodiester peak (PDE2) was negatively related to these variables. In the second study, we examined well‐phenotyped older adults that were classified as nonsarcopenic or sarcopenic based on sex‐specific criteria described by the European Working Group on Sarcopenia in Older People. PCr content was lower in muscle from older adults with sarcopenia compared to controls, while PDE2 was elevated. Percutaneous biopsy specimens of the vastus lateralis were obtained for metabolomic and lipidomic analyses. Lower PCr was related to higher muscle creatine. PDE2 was associated with glycerol‐phosphoethanolamine levels, a putative marker of phospholipid membrane damage. Lipidomic analyses revealed that the major phospholipids, (phosphatidylcholine, phosphatidylethanolamine, and phosphatidylglycerol) were elevated in sarcopenic muscle and were inversely related to muscle volume and peak power. These data suggest phosphorus metabolites and phospholipids are associated with the loss of skeletal muscle mass and function in older adults.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Fanchao Yi
- AdventHealth Translational Research Institute Orlando FL USA
| | - Rick B. Vega
- AdventHealth Translational Research Institute Orlando FL USA
| | | | - Paul M. Coen
- AdventHealth Translational Research Institute Orlando FL USA
| |
Collapse
|