1
|
Traoré M, Noviello C, Vergnol A, Gentil C, Halliez M, Saillard L, Gelin M, Forand A, Lemaitre M, Guesmia Z, Cadot B, Caldas de Almeida Araujo E, Marty B, Mougenot N, Messéant J, Strochlic L, Sadoine J, Slimani L, Jolly A, De la Grange P, Hogrel JY, Pietri-Rouxel F, Falcone S. GDF5 as a rejuvenating treatment for age-related neuromuscular failure. Brain 2024; 147:3834-3848. [PMID: 38584513 DOI: 10.1093/brain/awae107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/08/2024] [Accepted: 03/23/2024] [Indexed: 04/09/2024] Open
Abstract
Sarcopenia involves a progressive loss of skeletal muscle force, quality and mass during ageing, which results in increased inability and death; however, no cure has been established thus far. Growth differentiation factor 5 (GDF5) has been described to modulate muscle mass maintenance in various contexts. For our proof of concept, we overexpressed GDF5 by AAV vector injection in tibialis anterior muscle of adult aged (20 months) mice and performed molecular and functional analysis of skeletal muscle. We analysed human vastus lateralis muscle biopsies from adult young (21-42 years) and aged (77-80 years) donors, quantifying the molecular markers modified by GDF5 overexpression in mouse muscle. We validated the major effects of GDF5 overexpression using human immortalized myotubes and Schwann cells. We established a preclinical study by treating chronically (for 4 months) aged mice using recombinant GDF5 protein (rGDF5) in systemic administration and evaluated the long-term effect of this treatment on muscle mass and function. Here, we demonstrated that GDF5 overexpression in the old tibialis anterior muscle promoted an increase of 16.5% of muscle weight (P = 0.0471) associated with a higher percentage of 5000-6000 µm2 large fibres (P = 0.0211), without the induction of muscle regeneration. Muscle mass gain was associated with an amelioration of 26.8% of rate of force generation (P = 0.0330) and better neuromuscular connectivity (P = 0.0098). Moreover, GDF5 overexpression preserved neuromuscular junction morphology (38.5% of nerve terminal area increase, P < 0.0001) and stimulated the expression of reinnervation-related genes, in particular markers of Schwann cells (fold-change 3.19 for S100b gene expression, P = 0.0101). To characterize the molecular events induced by GDF5 overexpression during ageing, we performed a genome-wide transcriptomic analysis of treated muscles and showed that this factor leads to a 'rejuvenating' transcriptomic signature in aged mice, as 42% of the transcripts dysregulated by ageing reverted to youthful expression levels upon GDF5 overexpression (P < 0.05). Towards a preclinical approach, we performed a long-term systemic treatment using rGDF5 and showed its effectiveness in counteracting age-related muscle wasting, improving muscle function (17.8% of absolute maximal force increase, P = 0.0079), ensuring neuromuscular connectivity and preventing neuromuscular junction degeneration (7.96% of AchR area increase, P = 0.0125). In addition, in human muscle biopsies, we found the same age-related alterations than those observed in mice and improved by GDF5 and reproduced its major effects on human cells, suggesting this treatment as efficient in humans. Overall, these data provide a foundation to examine the curative potential of GDF5 drug in clinical trials for sarcopenia and, eventually, other neuromuscular diseases.
Collapse
Affiliation(s)
- Massiré Traoré
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Chiara Noviello
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Amélie Vergnol
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Christel Gentil
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Marius Halliez
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Lucile Saillard
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Maxime Gelin
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Anne Forand
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
- Inovarion, F-75005 Paris, France
| | - Mégane Lemaitre
- Sorbonne Université, INSERM UMS28, Phénotypage du Petit Animal, 75013 Paris, France
| | - Zoheir Guesmia
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Bruno Cadot
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | | | - Benjamin Marty
- Institut de Myologie, CEA, Laboratoire d'imagerie et de spectroscopie par RMN, F-75013 Paris, France
| | - Nathalie Mougenot
- Sorbonne Université, INSERM UMS28, Phénotypage du Petit Animal, 75013 Paris, France
| | - Julien Messéant
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Laure Strochlic
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Jeremy Sadoine
- Université de Paris, Plateforme d'Imagerie du Vivant (PIV), F-92120 Montrouge, France
| | - Lofti Slimani
- Université de Paris, Plateforme d'Imagerie du Vivant (PIV), F-92120 Montrouge, France
| | - Ariane Jolly
- GenoSplice, Paris Biotech Santé, F-75014 Paris, France
| | | | - Jean-Yves Hogrel
- Institut de Myologie, Laboratoire de physiologie et d'évaluation neuromusculaire, F-75013 Paris, France
| | - France Pietri-Rouxel
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Sestina Falcone
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| |
Collapse
|
2
|
Umamoto K, Bouchi R, Soeda K, Satake S, Hosoyama T, Ohsugi M, Ueki K, Kajio H. Association of biomarkers and Barthel Index with occurrence of age-related adverse health outcomes in individuals with diabetes. J Diabetes Investig 2024; 15:1675-1683. [PMID: 39140764 PMCID: PMC11527827 DOI: 10.1111/jdi.14286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/01/2024] [Accepted: 07/29/2024] [Indexed: 08/15/2024] Open
Abstract
AIMS/INTRODUCTION The clinical significance of age-related biomarkers in patients with diabetes has not been fully elucidated. In this study, we aimed to establish models to predict the progression of aging in patients with diabetes using biomarkers. MATERIALS AND METHODS This single-center, retrospective cohort study included 115 Japanese patients with diabetes aged ≥60 years. Age-related adverse health outcomes were defined as emergency hospitalization, any increase in the level of nursing care certification, admission to a nursing home or death. The associations of age-related biomarker levels (adiponectin, growth differentiation factor 15 [GDF15], C-X-C motif chemokine ligand 9 and apelin) and clinical indicators with age-related adverse health outcomes were evaluated. Factors that predominantly influenced the occurrence of age-related adverse health outcomes were explored using the Cox proportional hazards model. RESULTS The mean age of the 115 participants was 73 years, 50.6% were men, the mean body mass index and hemoglobin A1c level were 25.3 kg/m2 and 9.79%, respectively. There were 26 age-related adverse health outcomes during the study period (median 1.93, range 0-4.65 years). In a model combining clinical indicators and biomarkers, including the Barthel Index, GDF15 and adiponectin, the occurrence of age-related adverse health outcomes was found to be significantly associated with GDF15 and Barthel Index. The group with both GDF15 and adiponectin levels higher than the median proved to be significantly higher than the group with both lower. CONCLUSIONS The measurement of GDF15 and adiponectin levels and the Barthel Index might be useful for predicting age-related adverse health outcomes in patients with diabetes.
Collapse
Affiliation(s)
- Kotaro Umamoto
- Department of Diabetes, Endocrinology and Metabolism, Center HospitalNational Center for Global Health and MedicineTokyoJapan
| | - Ryotaro Bouchi
- Department of Diabetes, Endocrinology and Metabolism, Center HospitalNational Center for Global Health and MedicineTokyoJapan
- Diabetes and Metabolism Information Center, Research InstituteNational Center for Global Health and MedicineTokyoJapan
| | - Kotaro Soeda
- Diabetes Research Center, Research InstituteNational Center for Global Health and MedicineTokyoJapan
| | - Shosuke Satake
- Department of Geriatric MedicineNational Center for Geriatrics and GerontologyAichiJapan
| | - Tohru Hosoyama
- Department of Musculoskeletal Disease, The Geroscience Research CenterNational Center for Geriatrics and GerontologyAichiJapan
| | - Mitsuru Ohsugi
- Department of Diabetes, Endocrinology and Metabolism, Center HospitalNational Center for Global Health and MedicineTokyoJapan
- Diabetes and Metabolism Information Center, Research InstituteNational Center for Global Health and MedicineTokyoJapan
| | - Kohjiro Ueki
- Department of Diabetes, Endocrinology and Metabolism, Center HospitalNational Center for Global Health and MedicineTokyoJapan
- Diabetes Research Center, Research InstituteNational Center for Global Health and MedicineTokyoJapan
| | - Hiroshi Kajio
- Department of Diabetes, Endocrinology and Metabolism, Center HospitalNational Center for Global Health and MedicineTokyoJapan
| |
Collapse
|
3
|
Lee YJ, Kim GH, Lee DS, Jeong HJ, Lim JH. Activation of the apelin/APJ system by vitamin D attenuates age-related muscle atrophy. Life Sci 2024; 359:123205. [PMID: 39488262 DOI: 10.1016/j.lfs.2024.123205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/20/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024]
Abstract
AIMS Age-related frailty and reduced physical activity contribute to a degenerative loss of muscle mass, function, and strength, which is known as sarcopenia. Increasing evidence has shown that vitamin D has beneficial effects on the muscle health. However, the molecular mechanisms of vitamin D have not been fully elucidated. In this study, we aimed to demonstrate whether vitamin D can overcome muscle atrophy due to aging, especially with respect to the regulation of myokines. MAIN METHODS Young (3-month-old) and aged (18-month-old) C57BL/6 mice were assigned to the following 3 groups: normal diet (1000 IU/kg), vitamin D3-supplemented diet (20,000 IU/kg), and normal diet plus exercise for 4 months. KEY FINDINGS We found that the reduction in muscle strength and mass due to aging was reversed by vitamin D3 supplementation. The levels of markers involved in muscle atrophy and cellular senescence in the muscle of the aged mice were substantially decreased by vitamin D3. Interestingly, we observed that the expression of apelin and its receptor (APJ), which is known to be secreted after exercise, significantly increased in aged muscles with a vitamin D3-supplemented diet but not in the young mice. Moreover, circulating interleukin-6 (IL-6) and growth differentiation factor 8 (GDF8) levels were significantly increased in the aged mice but were restored by vitamin D3 treatment. SIGNIFICANCE Our present data indicate that vitamin D3 supplementation ameliorates aging-induced muscle atrophy and senescence, similar to the effects of exercise, suggesting the positive impact of vitamin D as an intervention strategy to prevent aging-induced metabolic diseases.
Collapse
Affiliation(s)
- Yoo Jeong Lee
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Cheongju 28159, Republic of Korea.
| | - Gyu Hee Kim
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Cheongju 28159, Republic of Korea
| | - Da Som Lee
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Cheongju 28159, Republic of Korea
| | - Hyeon-Ju Jeong
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Cheongju 28159, Republic of Korea
| | - Joo Hyun Lim
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Cheongju 28159, Republic of Korea.
| |
Collapse
|
4
|
Gustafsson T, Ulfhake B. Aging Skeletal Muscles: What Are the Mechanisms of Age-Related Loss of Strength and Muscle Mass, and Can We Impede Its Development and Progression? Int J Mol Sci 2024; 25:10932. [PMID: 39456714 PMCID: PMC11507513 DOI: 10.3390/ijms252010932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
As we age, we lose muscle strength and power, a condition commonly referred to as sarcopenia (ICD-10-CM code (M62.84)). The prevalence of sarcopenia is about 5-10% of the elderly population, resulting in varying degrees of disability. In this review we emphasise that sarcopenia does not occur suddenly. It is an aging-induced deterioration that occurs over time and is only recognised as a disease when it manifests clinically in the 6th-7th decade of life. Evidence from animal studies, elite athletes and longitudinal population studies all confirms that the underlying process has been ongoing for decades once sarcopenia has manifested. We present hypotheses about the mechanism(s) underlying this process and their supporting evidence. We briefly review various proposals to impede sarcopenia, including cell therapy, reducing senescent cells and their secretome, utilising targets revealed by the skeletal muscle secretome, and muscle innervation. We conclude that although there are potential candidates and ongoing preclinical and clinical trials with drug treatments, the only evidence-based intervention today for humans is exercise. We present different exercise programmes and discuss to what extent the interindividual susceptibility to developing sarcopenia is due to our genetic predisposition or lifestyle factors.
Collapse
Affiliation(s)
| | - Brun Ulfhake
- Department of Laboratory Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden;
| |
Collapse
|
5
|
Deng Z, Song C, Chen L, Zhang R, Yang L, Zhang P, Xiu Y, Su Y, Luo F, Luo J, Dai H, Xu J. Inhibition of CILP2 Improves Glucose Metabolism and Mitochondrial Dysfunction in Sarcopenia via the Wnt Signalling Pathway. J Cachexia Sarcopenia Muscle 2024. [PMID: 39385717 DOI: 10.1002/jcsm.13597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Skeletal muscle is the primary organ involved in insulin-mediated glucose metabolism. Elevated levels of CILP2 are a significant indicator of impaired glucose tolerance and are predominantly expressed in skeletal muscle. It remains unclear whether CILP2 contributes to age-related muscle atrophy through regulating the glucose homeostasis and insulin sensitivity. METHODS Initially, the expression levels of CILP2 were assessed in elderly mice and patients with sarcopenia. Lentiviral vectors were used to induce either silencing or overexpression of CILP2 in C2C12 myoblast cells. The effects of CILP2 on proliferation, myogenic differentiation, insulin sensitivity and glucose uptake were evaluated using immunofluorescence, western blotting, real-time quantitative polymerase chain reaction, RNA sequencing, glucose uptake experiments, dual-luciferase reporter assays and co-immunoprecipitation (CO-IP). An adeno-associated virus-9 containing a muscle-specific promoter was injected into SAMP8 senile mice to observe the efficacy of CILP2 knockout. RESULTS We found that there was more CLIP2 expressed in the skeletal muscle of ageing mice (+1.1-fold, p < 0.01) and in patients with sarcopenia (+2.5-fold, p < 0.01) compared to the control group. Following the overexpression of CILP2, Ki67 (-65%, p < 0.01), PCNA (-32%, p < 0.05), MyoD1 (-89%, p < 0.001), MyoG (-31%, p < 0.05) and MyHC (-85%, p < 0.001), which indicate proliferation and differentiation potential, were significantly reduced. In contrast, MuRF-1 (+59%, p < 0.05), atrogin-1 (+43%, p < 0.05) and myostatin (+31%, p < 0.05), the markers of muscular atrophy, were significantly increased. Overexpression of CILP2 decreased insulin sensitivity, glucose uptake (-18%, p < 0.001), GLUT4 translocation to the membrane and the maximum respiratory capacity of mitochondria. Canonical Wnt signalling was identified through RNA sequencing as a potential pathway for CILP2 regulation in C2C12, and Wnt3a was confirmed as an interacting protein of CILP2 in the CO-IP assay. The addition of recombinant Wnt3a protein reversed the inhibitory effects on myogenesis and glucose metabolism caused by CILP2 overexpression. Conversely, CILP2 knockdown promoted myogenesis and glucose metabolism. CILP2 knockdown improved muscle atrophy in mice, characterized by significant increases in time to exhaustion (+42%, p < 0.001), grip strength (+19%, p < 0.01), muscle mass (+15%, p < 0.001) and mean muscle cross-sectional area (+37%, p < 0.01). CILP2 knockdown enhanced glycogen synthesis (+83%, p < 0.001) and the regeneration of oxidative and glycolytic muscle fibres in SAMP8 ageing mice via the Wnt/β-catenin signalling pathway. CONCLUSIONS Our results indicate that CILP2 interacts with Wnt3a to suppress the Wnt/β-catenin signalling pathway and its downstream cascade, leading to impaired insulin sensitivity and glucose metabolism in skeletal muscle. Targeting CILP2 inhibition could offer potential therapeutic benefits for sarcopenia.
Collapse
Affiliation(s)
- Zhibo Deng
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, People's Republic of China
- Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Chao Song
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, People's Republic of China
- Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Long Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, People's Republic of China
- Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Rongsheng Zhang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, People's Republic of China
- Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Linhai Yang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, People's Republic of China
- Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Peng Zhang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, People's Republic of China
- Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Yu Xiu
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, People's Republic of China
| | - Yibin Su
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, People's Republic of China
| | - Fenqi Luo
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, People's Republic of China
- Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jun Luo
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, People's Republic of China
- Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Hanhao Dai
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, People's Republic of China
- Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jie Xu
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, People's Republic of China
- Department of Orthopedics, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| |
Collapse
|
6
|
Tarum J, Ball G, Gustafsson T, Altun M, Santos L. Artificial neural network inference analysis identified novel genes and gene interactions associated with skeletal muscle aging. J Cachexia Sarcopenia Muscle 2024; 15:2143-2155. [PMID: 39210538 PMCID: PMC11446686 DOI: 10.1002/jcsm.13562] [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: 11/27/2023] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Sarcopenia is an age-related muscle disease that increases the risk of falls, disabilities, and death. It is associated with increased muscle protein degradation driven by molecular signalling pathways including Akt and FOXO1. This study aims to identify genes, gene interactions, and molecular pathways and processes associated with muscle aging and exercise in older adults that remained undiscovered until now leveraging on an artificial intelligence approach called artificial neural network inference (ANNi). METHODS Four datasets reporting the profile of muscle transcriptome obtained by RNA-seq of young (21-43 years) and older adults (63-79 years) were selected and retrieved from the Gene Expression Omnibus (GEO) data repository. Two datasets contained the transcriptome profiles associated to muscle aging and two the transcriptome linked to resistant exercise in older adults, the latter before and after 6 months of exercise training. Each dataset was individually analysed by ANNi based on a swarm neural network approach integrated into a deep learning model (Intelligent Omics). This allowed us to identify top 200 genes influencing (drivers) or being influenced (targets) by aging or exercise and the strongest interactions between such genes. Downstream gene ontology (GO) analysis of these 200 genes was performed using Metacore (Clarivate™) and the open-source software, Metascape. To confirm the differential expression of the genes showing the strongest interactions, real-time quantitative PCR (RT-qPCR) was employed on human muscle biopsies obtained from eight young (25 ± 4 years) and eight older men (78 ± 7.6 years), partaking in a 6-month resistance exercise training programme. RESULTS CHAD, ZDBF2, USP54, and JAK2 were identified as the genes with the strongest interactions predicting aging, while SCFD1, KDM5D, EIF4A2, and NIPAL3 were the main interacting genes associated with long-term exercise in older adults. RT-qPCR confirmed significant upregulation of USP54 (P = 0.005), CHAD (P = 0.03), and ZDBF2 (P = 0.008) in the aging muscle, while exercise-related genes were not differentially expressed (EIF4A2 P = 0.99, NIPAL3 P = 0.94, SCFD1 P = 0.94, and KDM5D P = 0.64). GO analysis related to skeletal muscle aging suggests enrichment of pathways linked to bone development (adj P-value 0.006), immune response (adj P-value <0.001), and apoptosis (adj P-value 0.01). In older exercising adults, these were ECM remodelling (adj P-value <0.001), protein folding (adj P-value <0.001), and proteolysis (adj P-value <0.001). CONCLUSIONS Using ANNi and RT-qPCR, we identified three strongly interacting genes predicting muscle aging, ZDBF2, USP54, and CHAD. These findings can help to inform the design of nonpharmacological and pharmacological interventions that prevent or mitigate sarcopenia.
Collapse
Affiliation(s)
- Janelle Tarum
- Department of Sport Science, Sport, Health and Performance Enhancement Research Centre (SHAPE), School of Science and TechnologyNottingham Trent UniversityNottinghamUK
| | - Graham Ball
- Medical Technology Research CentreAnglia Ruskin UniversityEssexUK
| | - Thomas Gustafsson
- Department of Laboratory Medicine, Section of Clinical PhysiologyKarolinska Institutet HuddingeHuddingeSweden
- Department of Clinical PhysiologyKarolinska University HospitalHuddingeSweden
| | - Mikael Altun
- Department of Laboratory Medicine, Section of Clinical PhysiologyKarolinska Institutet HuddingeHuddingeSweden
- Department of Clinical PhysiologyKarolinska University HospitalHuddingeSweden
| | - Lívia Santos
- Department of Sport Science, Sport, Health and Performance Enhancement Research Centre (SHAPE), School of Science and TechnologyNottingham Trent UniversityNottinghamUK
| |
Collapse
|
7
|
Kim SJ, Jo Y, Park SJ, Ji E, Lee JY, Choi E, Baek JY, Jang IY, Jung HW, Kim K, Ryu D, Yoo HJ, Kim BJ. Metabolomic profiles of ovariectomized mice and their associations with body composition and frailty-related parameters in postmenopausal women. J Endocrinol Invest 2024; 47:2551-2563. [PMID: 38493245 DOI: 10.1007/s40618-024-02338-x] [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: 11/21/2023] [Accepted: 02/12/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Menopause, a dramatical estrogen-deficient condition, is considered the most significant milestone in women's health. PURPOSE To investigate the metabolite changes attributed to estrogen deficiency using random forest (RF)-based machine learning (ML) modeling strategy in ovariectomized (OVX) mice as well as determine the clinical relevance of selected metabolites in older women. METHODS AND RESULTS Untargeted and targeted metabolomic analyses revealed that metabolites related to TCA cycle, sphingolipids, phospholipids, fatty acids, and amino acids, were significantly changed in the plasma and/or muscle of OVX mice. Subsequent ML classifiers based on RF algorithm selected alpha-ketoglutarate (AKG), arginine, carnosine, ceramide C24, phosphatidylcholine (PC) aa C36:6, and PC ae C42:3 in plasma as well as PC aa 34:1, PC aa C34:3, PC aa C36:5, PC aa C32:1, PC aa C36:2, and sphingosine in muscle as top featured metabolites that differentiate the OVX mice from the sham-operated group. When circulating levels of AKG, arginine, and carnosine, which showed the most significant changes in OVX mice blood, were measured in postmenopausal women, higher plasma AKG levels were associated with lower bone mass, weak grip strength, poor physical performance, and increased frailty risk. CONCLUSIONS Metabolomics- and ML-based methods identified the key metabolites of blood and muscle that were significantly changed after ovariectomy in mice, and the clinical implication of several metabolites was investigated by looking at their correlation with body composition and frailty-related parameters in postmenopausal women. These findings provide crucial context for understanding the diverse physiological alterations caused by estrogen deficiency in women.
Collapse
Affiliation(s)
- S J Kim
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center,, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, South Korea
| | - Y Jo
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - S J Park
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - E Ji
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - J Y Lee
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - E Choi
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - J-Y Baek
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - I Y Jang
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - H-W Jung
- Division of Geriatrics, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - K Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - D Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea.
| | - H J Yoo
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center,, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, South Korea.
| | - B-J Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, South Korea.
| |
Collapse
|
8
|
Li Q, Li C, Zhang X. Research Progress on the Effects of Different Exercise Modes on the Secretion of Exerkines After Spinal Cord Injury. Cell Mol Neurobiol 2024; 44:62. [PMID: 39352588 PMCID: PMC11445308 DOI: 10.1007/s10571-024-01497-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
Exercise training is a conventional treatment strategy throughout the entire treatment process for patients with spinal cord injury (SCI). Currently, exercise modalities for SCI patients primarily include aerobic exercise, endurance training, strength training, high-intensity interval training, and mind-body exercises. These exercises play a positive role in enhancing skeletal muscle function, inducing neuroprotection and regeneration, thereby influencing neural plasticity, reducing limb spasticity, and improving motor function and daily living abilities in SCI patients. However, the mechanism by which exercise training promotes functional recovery after SCI is still unclear, and there is no consensus on a unified and standardized exercise treatment plan. Different exercise methods may bring different benefits. After SCI, patients' physical activity levels decrease significantly due to factors such as motor dysfunction, which may be a key factor affecting changes in exerkines. The changes in exerkines of SCI patients caused by exercise training are an important and highly relevant and visual evaluation index, which may provide a new research direction for revealing the intrinsic mechanism by which exercise promotes functional recovery after SCI. Therefore, this article summarizes the changes in the expression of common exerkines (neurotrophic factors, inflammatory factors, myokines, bioactive peptides) after SCI, and intends to analyze the impact and role of different exercise methods on functional recovery after SCI from the perspective of exerkines mechanism. We hope to provide theoretical basis and data support for scientific exercise treatment programs after SCI.
Collapse
Affiliation(s)
- Qianxi Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Chenyu Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Xin Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| |
Collapse
|
9
|
Selvais CM, Davis-López de Carrizosa MA, Versele R, Dubuisson N, Noel L, Brichard SM, Abou-Samra M. Challenging Sarcopenia: Exploring AdipoRon in Aging Skeletal Muscle as a Healthspan-Extending Shield. Antioxidants (Basel) 2024; 13:1073. [PMID: 39334732 PMCID: PMC11428238 DOI: 10.3390/antiox13091073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Sarcopenia, characterized by loss of muscle mass, quality, and function, poses significant risks in aging. We previously demonstrated that long-term treatment with AdipoRon (AR), an adiponectin receptor agonist, alleviated myosteatosis and muscle degeneration in middle-aged obese mice. This study aimed to determine if a shorter AR treatment could effectively offset sarcopenia in older mice. Two groups of old mice (20-23 months) were studied, one untreated (O) and one orally-treated with AR (O-AR) at 50 mg/kg/day for three months, compared with control 3-month-old young mice (Y) or 10-month-old young-adult mice (C-10). Results showed that AR remarkably inversed the loss of muscle mass by restoring the sarcopenia index and fiber count, which were greatly diminished with age. Additionally, AR successfully saved muscle quality of O mice by halving the accumulation of tubular aggregates and aberrant mitochondria, through AMPK pathway activation and enhanced autophagy. AR also bolstered muscle function by rescuing mitochondrial activity and improving exercise endurance. Finally, AR markedly curbed muscle fibrosis and mitigated local/systemic inflammation. Thus, a late three-month AR treatment successfully opposed sarcopenia and counteracted various hallmarks of aging, suggesting AR as a promising anti-aging therapy for skeletal muscles, potentially extending healthspan.
Collapse
Affiliation(s)
- Camille M Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, 1200 Brussels, Belgium
| | - Maria A Davis-López de Carrizosa
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, 1200 Brussels, Belgium
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41004 Seville, Spain
| | - Romain Versele
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, 1200 Brussels, Belgium
| | - Nicolas Dubuisson
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, 1200 Brussels, Belgium
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, 1200 Brussels, Belgium
| | - Sonia M Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, 1200 Brussels, Belgium
| | - Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, 1200 Brussels, Belgium
| |
Collapse
|
10
|
Rathor R, Suryakumar G. Myokines: A central point in managing redox homeostasis and quality of life. Biofactors 2024; 50:885-909. [PMID: 38572958 DOI: 10.1002/biof.2054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 03/15/2024] [Indexed: 04/05/2024]
Abstract
Redox homeostasis is a crucial phenomenon that is obligatory for maintaining the healthy status of cells. However, the loss of redox homeostasis may lead to numerous diseases that ultimately result in a compromised quality of life. Skeletal muscle is an endocrine organ that secretes hundreds of myokines. Myokines are peptides and cytokines produced and released by muscle fibers. Skeletal muscle secreted myokines act as a robust modulator for regulating cellular metabolism and redox homeostasis which play a prime role in managing and improving metabolic function in multiple organs. Further, the secretory myokines maintain redox homeostasis not only in muscles but also in other organs of the body via stabilizing oxidants and antioxidant levels. Myokines are also engaged in maintaining mitochondrial dynamics as mitochondria is a central point for the generation of reactive oxygen species (ROS). Ergo, myokines also act as a central player in communicating signals to other organs, including the pancreas, gut, liver, bone, adipose tissue, brain, and skin via their autocrine, paracrine, or endocrine effects. The present review provides a comprehensive overview of skeletal muscle-secreted myokines in managing redox homeostasis and quality of life. Additionally, probable strategies will be discussed that provide a solution for a better quality of life.
Collapse
Affiliation(s)
- Richa Rathor
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Ministry of Defence, Delhi, India
| | - Geetha Suryakumar
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Ministry of Defence, Delhi, India
| |
Collapse
|
11
|
Wu S, Li L, Xi H, Wu X, He Y, Sun X, Wu L. Bibliometrics and knowledge mapping of the pathogenesis of hepatic encephalopathy in patients with liver cirrhosis. Heliyon 2024; 10:e34330. [PMID: 39145014 PMCID: PMC11320160 DOI: 10.1016/j.heliyon.2024.e34330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024] Open
Abstract
Background Hepatic encephalopathy is a common and serious complication of decompensated cirrhosis. It can considerably contribute to economic burden and impaired quality of life. However, its pathogenesis remains unclear. Method In this study, we aimed to visually analyse the research status and development trends in hepatic encephalopathy pathogenesis using bibliometrics and knowledge mapping. Information regarding publications between 1978 and 2022 were obtained from the Web of Science Core Collection. CiteSpace was used to analyse and present data by year, author, institution, country, journal, reference, and keyword. Results A total of 1578 publications on hepatic encephalopathy pathogenesis in patients with cirrhosis were retrieved from Web of Science Core Collection. A gradual increasing trend in annual publications has occurred. The collaborative network analysis results suggest the United States of America, the University of London, and Bajaj, Jasmohan S as the most influential country, institution, and author, respectively, in this research field. Notably, China appeariiuis to be the most promising country. Research on 'hepatology' garners the most significant papers in the field. Combined with reference co-citation and keyword co-occurrence analyses, we found that ammonia metabolism, gut microbiota, sarcopenia, and trace elements will become future research frontiers that are likely to be explored for a considerable length of time. Conclusion Future research directions in HE pathogenesis may target modulating the ammonia metabolism, the gut microbiota, sarcopenia, and trace elements.
Collapse
Affiliation(s)
- Shiyan Wu
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Department of Gastroenterology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, Sichuan Province, China
| | - Lu Li
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Department of Gastroenterology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, Sichuan Province, China
| | - Heng Xi
- Department of Pharmacy, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, Sichuan Province, China
| | - Xiaoping Wu
- Department of Gastroenterology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, Sichuan Province, China
| | - Yumei He
- North Sichuan Medical College, Nanchong, 623300, Sichuan Province, China
| | - Xiaobin Sun
- Department of Gastroenterology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, Sichuan Province, China
| | - Liping Wu
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Department of Gastroenterology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, Sichuan Province, China
| |
Collapse
|
12
|
Briand M, Raffin J, Gonzalez-Bautista E, Ritz P, Abellan Van Kan G, Pillard F, Faruch-Bilfeld M, Guyonnet S, Dray C, Vellas B, de Souto Barreto P, Rolland Y. Body composition and aging: cross-sectional results from the INSPIRE study in people 20 to 93 years old. GeroScience 2024:10.1007/s11357-024-01245-6. [PMID: 39028455 DOI: 10.1007/s11357-024-01245-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 06/06/2024] [Indexed: 07/20/2024] Open
Abstract
Aging is characterized by several major changes, including altered body composition, which is associated with numerous negative clinical consequences such as sarcopenia, osteoporosis, and frailty. The study is to evaluate body composition parameters depending on age and sex in a population ranging from the young adult to the very old, and to identify break points in the association between body composition and age. In this cross-sectional study, we included the enrolment population of the French INSPIRE-T prospective cohort, accounting for 915 subjects (62% female). Age ranged from 20 to 93 years, median age (years) was 63 (IQR 27). Body composition (lean mass, fat mass, and bone mineral content) was assessed with dual-X-ray absorptiometry (DXA). Different break points in the relationship between age and body composition variables in males and females were identified using a segmented regression analysis adjusted on physical activity, nutritional status, educational level, and comorbidities. Lean mass decreased from the age of 55 years for males (CI 95% 44-66) and 31 years for females (CI 95% 23-39). For fat mass, we observed a trend towards an increase with age for males. For females, we observed an increase with age up to age 75 (CI 95% 62-86), followed by a decreasing trend. In this study, we described the relationship between body composition and age as a function of sex, establishing a foundation for further studies on predictive biomarkers of age-related body composition alteration.
Collapse
Affiliation(s)
- Marguerite Briand
- IHU HealthAge, Toulouse, France.
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France.
- Institut RESTORE, UMR 1301, University of Toulouse III, Inserm, UPS, CNRS, Toulouse, France.
| | - Jeremy Raffin
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
| | - Emmanuel Gonzalez-Bautista
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
| | - Patrick Ritz
- CERPOP UMR1295, University of Toulouse III, Inserm, UPS, Toulouse, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Toulouse University Hospital, Toulouse, France
| | - Gabor Abellan Van Kan
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
- CERPOP UMR1295, University of Toulouse III, Inserm, UPS, Toulouse, France
| | - Fabien Pillard
- Department of Sport Medicine, Hospital and University of Toulouse, Toulouse, France
- Institut RESTORE, UMR 1301, University of Toulouse III, Inserm, UPS, CNRS, Toulouse, France
| | | | - Sophie Guyonnet
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
- CERPOP UMR1295, University of Toulouse III, Inserm, UPS, Toulouse, France
| | - Cédric Dray
- Institut RESTORE, UMR 1301, University of Toulouse III, Inserm, UPS, CNRS, Toulouse, France
| | - Bruno Vellas
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
- CERPOP UMR1295, University of Toulouse III, Inserm, UPS, Toulouse, France
| | - Philipe de Souto Barreto
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
- CERPOP UMR1295, University of Toulouse III, Inserm, UPS, Toulouse, France
| | - Yves Rolland
- IHU HealthAge, Toulouse, France
- Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, Toulouse, France
- CERPOP UMR1295, University of Toulouse III, Inserm, UPS, Toulouse, France
| |
Collapse
|
13
|
Zhang H, Gu W, Wu G, Yu Y. Aging and Autophagy: Roles in Musculoskeletal System Injury. Aging Dis 2024:AD.2024.0362. [PMID: 38913046 DOI: 10.14336/ad.2024.0362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024] Open
Abstract
Aging is a multifactorial process that ultimately leads to a decline in physiological function and a consequent reduction in the health span, and quality of life in elderly population. In musculoskeletal diseases, aging is often associated with a gradual loss of skeletal muscle mass and strength, resulting in reduced functional capacity and an increased risk of chronic metabolic diseases, leading to impaired function and increased mortality. Autophagy is a highly conserved physiological process by which cells, under the regulation of autophagy-related genes, degrade their own organelles and large molecules by lysosomal degradation. This process is unique to eukaryotic cells and is a strict regulator of homeostasis, the maintenance of energy and substance balance. Autophagy plays an important role in a wide range of physiological and pathological processes such as cell homeostasis, aging, immunity, tumorigenesis and neurodegenerative diseases. On the one hand, under mild stress conditions, autophagy mediates the restoration of homeostasis and proliferation, reduction of the rate of aging and delay of the aging process. On the other hand, under more intense stress conditions, an inadequate suppression of autophagy can lead to cellular aging. Conversely, autophagy activity decreases during aging. Due to the interrelationship between aging and autophagy, limited literature exists on this topic. Therefore, the objective of this review is to summarize the current concepts on aging and autophagy in the musculoskeletal system. The aim is to better understand the mechanisms of age-related changes in bone, joint and muscle, as well as the interaction relationship between autophagy and aging. Its goal is to provide a comprehensive perspective for the improvement of diseases of the musculoskeletal system.
Collapse
Affiliation(s)
- Haifeng Zhang
- Department of Orthopedics Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenhui Gu
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, Nantong, Jiangsu, China
| | - Genbin Wu
- Department of Orthopedics Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinxian Yu
- Department of Orthopedics Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Di Cola S, Khan S, Lapenna L, Merli M. Emerging drugs for the treatment of sarcopenia in cirrhosis of the liver. Expert Opin Emerg Drugs 2024; 29:81-91. [PMID: 38549232 DOI: 10.1080/14728214.2024.2332428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/14/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION Malnutrition and sarcopenia are common and impact the prognosis in patients with liver cirrhosis. The etiology is multifactorial and includes periods of reduced caloric intake, increased catabolism and direct molecular mechanisms that inhibit muscle synthesis. Although these conditions are widely acknowledged, and there is a growing interest in their diagnosis, robust evidence regarding the treatment and reversibility of these conditions is still lacking. AREAS COVERED We have explored the current evidence on the pharmacological treatment of sarcopenia in patients with cirrhosis. Additionally, we have searched for drugs already in use and ongoing trials for other chronic diseases. EXPERT OPINION The current guidelines recommend the use of a protein-adequate diet and moderate physical activity for treating sarcopenia in patients with cirrhosis. Currently, robust evidence is derived only from the supplementation of Branched-Chain Amino Acids, capable of increasing muscle mass and function. There are many drugs targeting various pathways that contribute to sarcopenia. However, evidence is sporadic and insufficient to suggest their use in clinical practice.Novel drugs specifically designed to enhance muscle mass and function should be developed. Finally, gender significantly influences the type of muscle alteration and therapeutic mechanisms; therefore, future studies should be designed taking gender differences into consideration.
Collapse
Affiliation(s)
- Simone Di Cola
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Saniya Khan
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia Lapenna
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Manuela Merli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
15
|
Kuai D, Tang Q, Wang X, Yan Q, Tian W, Zhang H. Relationship between serum apelin, visfatin levels, and body composition in Polycystic Ovary Syndrome patients. Eur J Obstet Gynecol Reprod Biol 2024; 297:24-29. [PMID: 38555852 DOI: 10.1016/j.ejogrb.2024.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/08/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
OBJECTIVE To investigate the relationship between body composition and serum visfatin and apelin levels in patients with polycystic ovary syndrome (PCOS). METHODS In this prospective observational study, the differences in body composition, levels of gonadal hormone concentrations, glucose metabolism, apelin, and visfatin were compared between PCOS patients and the control group. PCOS patients were further divided into different subgroups according to different obesity criteria and the differences between serum visfatin and apelin levels in different subgroups were compared. Finally, the correlation of serum visfatin levels and apelin levels with body composition, and metabolism-related indicators in PCOS patients was explored. RESULTS A total collected 178 cases of PCOS patients and 172 cases of healthy women (control group) between 2020 July and 2021 November. In PCOS patients, their weight, Body Mass Index (BMI), Waist Hip Rate (WHR), Fat-Free Mass Index (FFMI), Percent Body Fat (PBF), Fat mass index (FMI), PBF of Arm, PBF of Leg, PBF of the Trunk, Visceral Fat Level (VFL), fasting insulin (FINS), Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) and Luteinizing hormone (LH) were significantly higher than in the control group (all P < 0.001), Percent Skeletal Muscle (PSM), PSM of Leg, and PSM of the Trunk were significantly decreased than in the control group (all P < 0.001). The PCOS patients had significantly higher serum visfatin levels and apelin levels compared with the control group (all P < 0.001). In PBF > 35 % PCOS patients, the apelin and visfatin levels were significantly higher than the PBF ≤ 35 % PCOS patients. In WHR ≥ 0.85 and BMI ≥ 24 kg/m2 PCOS patients, the visfatin levels were significantly higher than the WHR < 0.85 and BMI < 24 kg/m2 PCOS patients. Serum apelin and visfatin positively correlated with BMI level, WHR, FFMI, PBF, FMI, PBF of arms, PBF of legs, PBF of the trunk, VFL, FBG, HOMA-IR index and negatively correlated with PSM, PSM of legs, and PSM of the trunk (all P < 0.001). CONCLUSIONS Compared with healthy women, Patients with PCOS have an increased fat content in various parts of the body, reduced skeletal muscle content, and are often complicated by metabolic abnormalities. Serum visfatin and apelin correlated not only with obesity, fat mass, and fat distribution but also with muscle mass and distribution. It may be possible to reduce the long-term risk of metabolic disease in PCOS through the monitoring and management of the body composition in PCOS patients or to reflect the therapeutic effect of PCOS.
Collapse
Affiliation(s)
- Dan Kuai
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Qingtao Tang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiang Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Qi Yan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wenyan Tian
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Huiying Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
16
|
Li J, Shen Z, Lin Y, Wang Z, Li M, Sun H, Wang Q, Zhao C, Xu J, Lu X, Gao W. DNA methylation of skeletal muscle function-related secretary factors identifies FGF2 as a potential biomarker for sarcopenia. J Cachexia Sarcopenia Muscle 2024; 15:1209-1217. [PMID: 38641928 PMCID: PMC11154778 DOI: 10.1002/jcsm.13472] [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: 06/09/2023] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Sarcopenia is characterized by progressive loss of muscle mass and function due to aging. DNA methylation has been identified to play important roles in the dysfunction of skeletal muscle. The aim of our present study was to explore the whole blood sample-based methylation changes of skeletal muscle function-related factors in patients with sarcopenia. METHODS The overall DNA methylation levels were analysed by using MethlTarget™ DNA Methylation Analysis platform in a discovery set consistent of 50 sarcopenic older adults (aged ≥65 years) and 50 age- and sex-matched non-sarcopenic individuals. The candidate differentially methylated regions (DMRs) were further validated by Methylation-specific PCR (MSP) in another two independent larger sets and confirmed by pyrosequencing. Receiver operating characteristic (ROC) curve analysis was used to determine the optimum cut-off levels of fibroblast growth factor 2 (FGF2)_30 methylation best predicting sarcopenia and area under the ROC curve (AUC) was measured. The correlation between candidate DMRs and the risk of sarcopenia was investigated by univariate analysis and multivariate logistic regression analysis. RESULTS Among 1149 cytosine-phosphate-guanine (CpG) sites of 27 skeletal muscle function-related secretary factors, 17 differentially methylated CpG sites and 7 differentially methylated regions (DMRs) were detected between patients with sarcopenia and control subjects in the discovery set. Further methylation-specific PCR identified that methylation of fibroblast growth factor 2 (FGF2)_30 was lower in patients with sarcopenia and the level was decreased as the severity of sarcopenia increased, which was confirmed by pyrosequencing. Correlation analysis demonstrated that the methylation level of FGF2_30 was positively correlated to ASMI (r = 0.372, P < 0.001), grip strength (r = 0.334, P < 0.001), and gait speed (r = 0.411, P < 0.001). ROC curve analysis indicated that the optimal cut-off value of FGF2_30 methylation level that predicted sarcopenia was 0.15 with a sensitivity of 84.6% and a specificity of 70.1% (AUC = 0.807, 95% CI = 0.756-0.858, P < 0.001). Multivariate logistic regression analyses showed that lower FGF2_30 methylation level (<0.15) was significantly associated with increased risk of sarcopenia even after adjustment for potential confounders including age, sex, and BMI (adjusted OR = 9.223, 95% CI: 6.614-12.861, P < 0.001). CONCLUSIONS Our results suggest that lower FGF2_30 methylation is correlated with the risk and severity of sarcopenia in the older adults, indicating that FGF2 methylation serve as a surrogate biomarker for the screening and evaluation of sarcopenia.
Collapse
Affiliation(s)
- Jia‐Wen Li
- Department of Geriatrics, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| | - Zheng‐Kai Shen
- Jiangsu Province Center for Disease Control and PreventionNanjingChina
| | - Yu‐Shuang Lin
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Zhi‐Yue Wang
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Mei‐Lin Li
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Hui‐Xian Sun
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Quan Wang
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Can Zhao
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Jin‐Shui Xu
- Jiangsu Province Center for Disease Control and PreventionNanjingChina
| | - Xiang Lu
- Department of Geriatrics, Sir Run Run HospitalNanjing Medical UniversityNanjingChina
| | - Wei Gao
- Department of Geriatrics, Zhongda Hospital, School of MedicineSoutheast UniversityNanjingChina
| |
Collapse
|
17
|
Walzik D, Wences Chirino TY, Zimmer P, Joisten N. Molecular insights of exercise therapy in disease prevention and treatment. Signal Transduct Target Ther 2024; 9:138. [PMID: 38806473 PMCID: PMC11133400 DOI: 10.1038/s41392-024-01841-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/30/2024] Open
Abstract
Despite substantial evidence emphasizing the pleiotropic benefits of exercise for the prevention and treatment of various diseases, the underlying biological mechanisms have not been fully elucidated. Several exercise benefits have been attributed to signaling molecules that are released in response to exercise by different tissues such as skeletal muscle, cardiac muscle, adipose, and liver tissue. These signaling molecules, which are collectively termed exerkines, form a heterogenous group of bioactive substances, mediating inter-organ crosstalk as well as structural and functional tissue adaption. Numerous scientific endeavors have focused on identifying and characterizing new biological mediators with such properties. Additionally, some investigations have focused on the molecular targets of exerkines and the cellular signaling cascades that trigger adaption processes. A detailed understanding of the tissue-specific downstream effects of exerkines is crucial to harness the health-related benefits mediated by exercise and improve targeted exercise programs in health and disease. Herein, we review the current in vivo evidence on exerkine-induced signal transduction across multiple target tissues and highlight the preventive and therapeutic value of exerkine signaling in various diseases. By emphasizing different aspects of exerkine research, we provide a comprehensive overview of (i) the molecular underpinnings of exerkine secretion, (ii) the receptor-dependent and receptor-independent signaling cascades mediating tissue adaption, and (iii) the clinical implications of these mechanisms in disease prevention and treatment.
Collapse
Affiliation(s)
- David Walzik
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Tiffany Y Wences Chirino
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Philipp Zimmer
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
| | - Niklas Joisten
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
- Division of Exercise and Movement Science, Institute for Sport Science, University of Göttingen, 37075, Göttingen, Lower Saxony, Germany.
| |
Collapse
|
18
|
Heitman K, Alexander MS, Faul C. Skeletal Muscle Injury in Chronic Kidney Disease-From Histologic Changes to Molecular Mechanisms and to Novel Therapies. Int J Mol Sci 2024; 25:5117. [PMID: 38791164 PMCID: PMC11121428 DOI: 10.3390/ijms25105117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic kidney disease (CKD) is associated with significant reductions in lean body mass and in the mass of various tissues, including skeletal muscle, which causes fatigue and contributes to high mortality rates. In CKD, the cellular protein turnover is imbalanced, with protein degradation outweighing protein synthesis, leading to a loss of protein and cell mass, which impairs tissue function. As CKD itself, skeletal muscle wasting, or sarcopenia, can have various origins and causes, and both CKD and sarcopenia share common risk factors, such as diabetes, obesity, and age. While these pathologies together with reduced physical performance and malnutrition contribute to muscle loss, they cannot explain all features of CKD-associated sarcopenia. Metabolic acidosis, systemic inflammation, insulin resistance and the accumulation of uremic toxins have been identified as additional factors that occur in CKD and that can contribute to sarcopenia. Here, we discuss the elevation of systemic phosphate levels, also called hyperphosphatemia, and the imbalance in the endocrine regulators of phosphate metabolism as another CKD-associated pathology that can directly and indirectly harm skeletal muscle tissue. To identify causes, affected cell types, and the mechanisms of sarcopenia and thereby novel targets for therapeutic interventions, it is important to first characterize the precise pathologic changes on molecular, cellular, and histologic levels, and to do so in CKD patients as well as in animal models of CKD, which we describe here in detail. We also discuss the currently known pathomechanisms and therapeutic approaches of CKD-associated sarcopenia, as well as the effects of hyperphosphatemia and the novel drug targets it could provide to protect skeletal muscle in CKD.
Collapse
Affiliation(s)
- Kylie Heitman
- Division of Nephrology and Section of Mineral Metabolism, Department of Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Matthew S. Alexander
- Division of Neurology, Department of Pediatrics, The University of Alabama at Birmingham and Children’s of Alabama, Birmingham, AL 35294, USA
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christian Faul
- Division of Nephrology and Section of Mineral Metabolism, Department of Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| |
Collapse
|
19
|
Kilpiö T, Skarp S, Perjés Á, Swan J, Kaikkonen L, Saarimäki S, Szokodi I, Penninger JM, Szabó Z, Magga J, Kerkelä R. Apelin regulates skeletal muscle adaptation to exercise in a high-intensity interval training model. Am J Physiol Cell Physiol 2024; 326:C1437-C1450. [PMID: 38525542 DOI: 10.1152/ajpcell.00427.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024]
Abstract
Plasma apelin levels are reduced in aging and muscle wasting conditions. We aimed to investigate the significance of apelin signaling in cardiac and skeletal muscle responses to physiological stress. Apelin knockout (KO) and wild-type (WT) mice were subjected to high-intensity interval training (HIIT) by treadmill running. The effects of apelin on energy metabolism were studied in primary mouse skeletal muscle myotubes and cardiomyocytes. Apelin increased mitochondrial ATP production and mitochondrial coupling efficiency in myotubes and promoted the expression of mitochondrial genes both in primary myotubes and cardiomyocytes. HIIT induced mild concentric cardiac hypertrophy in WT mice, whereas eccentric growth was observed in the left ventricles of apelin KO mice. HIIT did not affect myofiber size in skeletal muscles of WT mice but decreased the myofiber size in apelin KO mice. The decrease in myofiber size resulted from a fiber type switch toward smaller slow-twitch type I fibers. The increased proportion of slow-twitch type I fibers in apelin KO mice was associated with upregulation of myosin heavy chain slow isoform expression, accompanied with upregulated expression of genes related to fatty acid transport and downregulated expression of genes related to glucose metabolism. Mechanistically, skeletal muscles of apelin KO mice showed defective induction of insulin-like growth factor-1 signaling in response to HIIT. In conclusion, apelin is required for proper skeletal and cardiac muscle adaptation to high-intensity exercise. Promoting apelinergic signaling may have benefits in aging- or disease-related muscle wasting conditions.NEW & NOTEWORTHY Apelin levels decline with age. This study demonstrates that in trained mice, apelin deficiency results in a switch from fast type II myofibers to slow oxidative type I myofibers. This is associated with a concomitant change in gene expression profile toward fatty acid utilization, indicating an aged-muscle phenotype in exercised apelin-deficient mice. These data are of importance in the design of exercise programs for aging individuals and could offer therapeutic target to maintain muscle mass.
Collapse
Affiliation(s)
- Teemu Kilpiö
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Sini Skarp
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Ábel Perjés
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Julia Swan
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Leena Kaikkonen
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Samu Saarimäki
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - István Szokodi
- Heart Institute, Medical School, and Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zoltán Szabó
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Johanna Magga
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Risto Kerkelä
- Research Unit of Biomedicine and Internal Medicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
20
|
Deprez A, Poletto Bonetto JH, Ravizzoni Dartora D, Dodin P, Nuyt AM, Luu TM, Dumont NA. Impact of preterm birth on muscle mass and function: a systematic review and meta-analysis. Eur J Pediatr 2024; 183:1989-2002. [PMID: 38416257 DOI: 10.1007/s00431-023-05410-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 02/29/2024]
Abstract
Individuals born preterm present lower exercise capacity. Along with the cardiopulmonary responses and activity level, muscle strength is a key determinant of exercise capacity. This systematic review aimed to summarize the current knowledge on the impact of preterm birth on skeletal muscle mass and function across the lifespan. The databases PubMed, MEDLINE, EBM, Embase, CINAHL Plus, Global Index Medicus, and Google Scholar were searched using keywords and MeSH terms related to skeletal muscle, preterm birth, and low birth weight. Two independent reviewers undertook study selection, data extraction, and quality appraisal using Covidence review management. Data were pooled to estimate the prematurity effect on muscle mass and function using the R software. From 4378 studies retrieved, 132 were full-text reviewed and 25 met the inclusion/exclusion criteria. Five studies presented a low risk of bias, and 5 had a higher risk of bias due to a lack of adjustment for confounding factors and presenting incomplete outcomes. Meta-analyses of pooled data from homogenous studies indicated a significant reduction in muscle thickness and jump test (muscle power) in individuals born preterm versus full-term with standardized mean difference and confidence interval of - 0.58 (0.27, 0.89) and - 0.45 (0.21, 0.69), respectively. Conclusion: Overall, this systematic review summarizing the existing literature on the impact of preterm birth on skeletal muscle indicates emerging evidence that individuals born preterm, display alteration in the development of their skeletal muscle mass and function. This work also highlights a clear knowledge gap in understanding the effect of preterm birth on skeletal muscle development. What is Known: • Preterm birth, which occurs at a critical time of skeletal muscle development and maturation, impairs the development of different organs and tissues leading to a higher risk of comorbidities such as cardiovascular diseases. • Preterm birth is associated with reduced exercise capacity. What is New: • Individuals born preterm display alterations in muscle mass and function compared to individuals born at term from infancy to adulthood. • There is a need to develop preventive or curative interventions to improve skeletal muscle health in preterm-born individuals.
Collapse
Affiliation(s)
- Alyson Deprez
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Jéssica H Poletto Bonetto
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Daniela Ravizzoni Dartora
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Philippe Dodin
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Anne Monique Nuyt
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Thuy Mai Luu
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Nicolas A Dumont
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada.
- School of Rehabilitation, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
21
|
Zhou N, Gong L, Zhang E, Wang X. Exploring exercise-driven exerkines: unraveling the regulation of metabolism and inflammation. PeerJ 2024; 12:e17267. [PMID: 38699186 PMCID: PMC11064867 DOI: 10.7717/peerj.17267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/28/2024] [Indexed: 05/05/2024] Open
Abstract
Exercise has many beneficial effects that provide health and metabolic benefits. Signaling molecules are released from organs and tissues in response to exercise stimuli and are widely termed exerkines, which exert influence on a multitude of intricate multi-tissue processes, such as muscle, adipose tissue, pancreas, liver, cardiovascular tissue, kidney, and bone. For the metabolic effect, exerkines regulate the metabolic homeostasis of organisms by increasing glucose uptake and improving fat synthesis. For the anti-inflammatory effect, exerkines positively influence various chronic inflammation-related diseases, such as type 2 diabetes and atherosclerosis. This review highlights the prospective contribution of exerkines in regulating metabolism, augmenting the anti-inflammatory effects, and providing additional advantages associated with exercise. Moreover, a comprehensive overview and analysis of recent advancements are provided in this review, in addition to predicting future applications used as a potential biomarker or therapeutic target to benefit patients with chronic diseases.
Collapse
Affiliation(s)
- Nihong Zhou
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
- School of Sport Science, Beijing Sport University, Beijing, China
| | - Lijing Gong
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
- Key Laboratory for Performance Training & Recovery of General Administration of Sport, Beijing Sport University, Beijing, China
| | - Enming Zhang
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmö, Sweden
- NanoLund Center for NanoScience, Lund University, Lund, Sweden
| | - Xintang Wang
- Key Laboratory for Performance Training & Recovery of General Administration of Sport, Beijing Sport University, Beijing, China
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| |
Collapse
|
22
|
Hur H, Kim HJ, Lee D, Jo C. Beef peptides mitigate skeletal muscle atrophy in C2C12 myotubes through protein degradation, protein synthesis, and the oxidative stress pathway. Food Funct 2024; 15:4564-4574. [PMID: 38584588 DOI: 10.1039/d3fo03911k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
This study aimed to investigate the potential of beef peptides (BPs) in mitigating muscle atrophy induced by dexamethasone (DEX) with underlying three mechanisms in vitro (protein degradation, protein synthesis, and the oxidative stress pathway). Finally, the anti-atrophic effect of BPs was enhanced through purification and isolation. BPs were generated using beef loin hydrolyzed with alcalase/ProteAX/trypsin, each at a concentration of 0.67%, followed by ultrafiltration through a 3 kDa cut-off. BPs (10-100 μg mL-1) dose-dependently counteracted the DEX-induced reductions in myotube diameters, differentiation, fusion, and maturation indices (p < 0.05). Additionally, BPs significantly reduced FoxO1 protein dephosphorylation, thereby suppressing muscle-specific E3 ubiquitin ligases such as muscle RING-finger containing protein-1 and muscle atrophy F-box protein in C2C12 myotubes at concentrations exceeding 25 μg mL-1 (p < 0.05). BPs also enhanced the phosphorylation of protein synthesis markers, including mTOR, 4E-BP1, and p70S6K1, in a dose-dependent manner (p < 0.05) and increased the mRNA expression of antioxidant enzymes. Fractionated peptides derived from BPs, through size exclusion and polarity-based fractionation, also demonstrated enhanced anti-atrophic effects compared to BPs. These peptides downregulated the mRNA expression of primary muscle atrophy markers while upregulated that of antioxidant enzymes. Specifically, peptides GAGAAGAPAGGA (MW 924.5) and AFRSSTKK (MW 826.4) were identified from fractionated peptides of BPs. These findings suggest that BPs, specifically the peptide fractions GAGAAGAPAGGA and AFRSSTKK, could be a potential strategy to mitigate glucocorticoid-induced skeletal muscle atrophy by reducing the E3 ubiquitin ligase activity.
Collapse
Affiliation(s)
- Hyeonjin Hur
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea.
| | - Hye-Jin Kim
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea.
| | - Dongheon Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea.
| | - Cheorun Jo
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea.
- Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang 25354, Republic of Korea
- Department of Animal Product Technology, Faculty of Animal Husbandry, Universitas Padjadjaran, West Java 45363, Indonesia
| |
Collapse
|
23
|
Roberts BM, Geddis AV, Ciuciu A, Reynoso M, Mehta N, Varanoske AN, Kelley AM, Walker RJ, Munoz R, Kolb AL, Staab JS, Naimo MA, Tomlinson RE. Acetaminophen influences musculoskeletal signaling but not adaptations to endurance exercise training. FASEB J 2024; 38:e23586. [PMID: 38568858 DOI: 10.1096/fj.202302642r] [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: 12/22/2023] [Revised: 03/01/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024]
Abstract
Acetaminophen (ACE) is a widely used analgesic and antipyretic drug with various applications, from pain relief to fever reduction. Recent studies have reported equivocal effects of habitual ACE intake on exercise performance, muscle growth, and risks to bone health. Thus, this study aimed to assess the impact of a 6-week, low-dose ACE regimen on muscle and bone adaptations in exercising and non-exercising rats. Nine-week-old Wistar rats (n = 40) were randomized to an exercise or control (no exercise) condition with ACE or without (placebo). For the exercise condition, rats ran 5 days per week for 6 weeks at a 5% incline for 2 min at 15 cm/s, 2 min at 20 cm/s, and 26 min at 25 cm/s. A human equivalent dose of ACE was administered (379 mg/kg body weight) in drinking water and adjusted each week based on body weight. Food, water intake, and body weight were measured daily. At the beginning of week 6, animals in the exercise group completed a maximal treadmill test. At the end of week 6, rats were euthanized, and muscle cross-sectional area (CSA), fiber type, and signaling pathways were measured. Additionally, three-point bending and microcomputer tomography were measured in the femur. Follow-up experiments in human primary muscle cells were used to explore supra-physiological effects of ACE. Data were analyzed using a two-way ANOVA for treatment (ACE or placebo) and condition (exercise or non-exercise) for all animal outcomes. Data for cell culture experiments were analyzed via ANOVA. If omnibus significance was found in either ANOVA, a post hoc analysis was completed, and a Tukey's adjustment was used. ACE did not alter body weight, water intake, food intake, or treadmill performance (p > .05). There was a treatment-by-condition effect for Young's Modulus where placebo exercise was significantly lower than placebo control (p < .05). There was no treatment by condition effects for microCT measures, muscle CSA, fiber type, or mRNA expression. Phosphorylated-AMPK was significantly increased with exercise (p < .05) and this was attenuated with ACE treatment. Furthermore, phospho-4EBP1 was depressed in the exercise group compared to the control (p < .05) and increased in the ACE control and ACE exercise group compared to placebo exercise (p < .05). A low dose of ACE did not influence chronic musculoskeletal adaptations in exercising rodents but acutely attenuated AMPK phosphorylation and 4EBP1 dephosphorylation post-exercise.
Collapse
Affiliation(s)
- Brandon M Roberts
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| | - Alyssa V Geddis
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| | - Alexandra Ciuciu
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Marinaliz Reynoso
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| | - Nikhil Mehta
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Alyssa N Varanoske
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
- Oak Ridge Institute for Science and Education, Belcamp, Maryland, USA
| | - Alyssa M Kelley
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| | - Raymond J Walker
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| | - Rigoberto Munoz
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| | - Alexander L Kolb
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| | - Jeffery S Staab
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| | - Marshall A Naimo
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts, USA
| | - Ryan E Tomlinson
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
24
|
De Spiegeleer A, Descamps A, Wynendaele E, Naumovski P, Crombez L, Planas M, Feliu L, Knappe D, Mouly V, Bigot A, Bielza R, Hoffmann R, Van Den Noortgate N, Elewaut D, De Spiegeleer B. Streptococcal quorum sensing peptide CSP-7 contributes to muscle inflammation and wasting. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167094. [PMID: 38428683 DOI: 10.1016/j.bbadis.2024.167094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/04/2024] [Accepted: 02/20/2024] [Indexed: 03/03/2024]
Abstract
Muscle wasting diseases, such as cancer cachexia and age-associated sarcopenia, have a profound and detrimental impact on functional independence, quality of life, and survival. Our understanding of the underlying mechanisms is currently limited, which has significantly hindered the development of targeted therapies. In this study, we explored the possibility that the streptococcal quorum sensing peptide Competence Stimulating Peptide 7 (CSP-7) might be a previously unidentified contributor to clinical muscle wasting. We found that CSP-7 selectively triggers muscle cell inflammation in vitro, specifically the release of IL-6. Furthermore, we demonstrated that CSP-7 can traverse the gastrointestinal barrier in vitro and is present in the systemic circulation in humans in vivo. Importantly, CSP-7 was associated with a muscle wasting phenotype in mice in vivo. Overall, our findings provide new mechanistic insights into the pathophysiology of muscle inflammation and wasting.
Collapse
Affiliation(s)
- Anton De Spiegeleer
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University Hospital, Ghent, Belgium; Department of Geriatrics, Faculty of Medicine and Health Sciences, Ghent University Hospital, Ghent, Belgium
| | - Amélie Descamps
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University Hospital, Ghent, Belgium; Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Evelien Wynendaele
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University Hospital, Ghent, Belgium; Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Petar Naumovski
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University Hospital, Ghent, Belgium; Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Liesbeth Crombez
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University Hospital, Ghent, Belgium; Department of Geriatrics, Faculty of Medicine and Health Sciences, Ghent University Hospital, Ghent, Belgium
| | - Marta Planas
- LIPPSO, Department of Chemistry, Universitat de Girona, Maria Aurèlia Capmany 69, Girona, Spain
| | - Lidia Feliu
- LIPPSO, Department of Chemistry, Universitat de Girona, Maria Aurèlia Capmany 69, Girona, Spain
| | - Daniel Knappe
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany; Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
| | - Vincent Mouly
- Centre de Recherche en Myologie, Sorbonne Université, Paris, France
| | - Anne Bigot
- Centre de Recherche en Myologie, Sorbonne Université, Paris, France
| | - Rafael Bielza
- Department of Geriatric Medicine, Hospital Universitario Infanta Sofía, Madrid, Spain
| | - Ralf Hoffmann
- Center for Biotechnology and Biomedicine, University of Leipzig, Leipzig, Germany; Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Leipzig, Germany
| | - Nele Van Den Noortgate
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University Hospital, Ghent, Belgium; Department of Geriatrics, Faculty of Medicine and Health Sciences, Ghent University Hospital, Ghent, Belgium
| | - Dirk Elewaut
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University Hospital, Ghent, Belgium; VIB Inflammation Research Center, Unit for Molecular Immunology and Inflammation, Ghent University, Ghent, Belgium; Department of Rheumatology, Faculty of Medicine and Health Sciences, Ghent University Hospital, Ghent, Belgium
| | - Bart De Spiegeleer
- Translational Research in Immunosenescence, Gerontology and Geriatrics (TRIGG) Group, Ghent University Hospital, Ghent, Belgium; Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium.
| |
Collapse
|
25
|
Qi H, Tian D, Luan F, Yang R, Zeng N. Pathophysiological changes of muscle after ischemic stroke: a secondary consequence of stroke injury. Neural Regen Res 2024; 19:737-746. [PMID: 37843207 PMCID: PMC10664100 DOI: 10.4103/1673-5374.382221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/30/2023] [Accepted: 06/01/2023] [Indexed: 10/17/2023] Open
Abstract
Sufficient clinical evidence suggests that the damage caused by ischemic stroke to the body occurs not only in the acute phase but also during the recovery period, and that the latter has a greater impact on the long-term prognosis of the patient. However, current stroke studies have typically focused only on lesions in the central nervous system, ignoring secondary damage caused by this disease. Such a phenomenon arises from the slow progress of pathophysiological studies examining the central nervous system. Further, the appropriate therapeutic time window and benefits of thrombolytic therapy are still controversial, leading scholars to explore more pragmatic intervention strategies. As treatment measures targeting limb symptoms can greatly improve a patient's quality of life, they have become a critical intervention strategy. As the most vital component of the limbs, skeletal muscles have become potential points of concern. Despite this, to the best of our knowledge, there are no comprehensive reviews of pathophysiological changes and potential treatments for post-stroke skeletal muscle. The current review seeks to fill a gap in the current understanding of the pathological processes and mechanisms of muscle wasting atrophy, inflammation, neuroregeneration, mitochondrial changes, and nutritional dysregulation in stroke survivors. In addition, the challenges, as well as the optional solutions for individualized rehabilitation programs for stroke patients based on motor function are discussed.
Collapse
Affiliation(s)
- Hu Qi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Dan Tian
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Fei Luan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Ruocong Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| |
Collapse
|
26
|
Palzkill VR, Tan J, Tice AL, Ferriera LF, Ryan TE. A 6-minute Limb Function Assessment for Therapeutic Testing in Experimental Peripheral Artery Disease Models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586197. [PMID: 38585832 PMCID: PMC10996543 DOI: 10.1101/2024.03.21.586197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Background The translation of promising therapies from pre-clinical models of hindlimb ischemia (HLI) to patients with peripheral artery disease (PAD) has been inadequate. While this failure is multifactorial, primary outcome measures in preclinical HLI models and clinical trials involving patients with PAD are not aligned well. For example, laser Doppler perfusion recovery measured under resting conditions is the most used outcome in HLI studies, whereas clinical trials involving patients with PAD primarily assess walking performance. Here, we sought to develop a 6-min limb function test for preclinical HLI models that assess muscular performance and hemodynamics congruently. Methods We developed an in situ 6-min limb function test that involves repeated isotonic (shortening) contractions performed against a submaximal load. Continuous measurement of muscle blood flow was performed using laser Doppler flowmetry. Quantification of muscle power, work, and perfusion are obtained across the test. To assess the efficacy of this test, we performed HLI via femoral artery ligation on several mouse strains: C57BL6J, BALBc/J, and MCK-PGC1α (muscle-specific overexpression of PGC1α). Additional experiments were performed using an exercise intervention (voluntary wheel running) following HLI. Results The 6-min limb function test was successful at detecting differences in limb function of C57BL6/J and BALBc/J mice subjected to HLI with effect sizes superior to laser Doppler perfusion recovery. C57BL6/J mice randomized to exercise therapy following HLI had smaller decline in muscle power, greater hyperemia, and performed more work across the 6-min limb function test compared to non-exercise controls with HLI. Mice with muscle-specific overexpression of PGC1α had no differences in perfusion recovery in resting conditions, but exhibited greater capillary density, increased muscle mass and absolute force levels, and performed more work across the 6-min limb function test compared to their wildtype littermates without the transgene. Conclusion These results demonstrate the efficacy of the 6-min limb function test to detect differences in the response to HLI across several interventions including where traditional perfusion recovery, capillary density, and muscle strength measures were unable to detect therapeutic differences.
Collapse
Affiliation(s)
- Victoria R. Palzkill
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | - Jianna Tan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
| | | | - Leonardo F. Ferriera
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- The Myology Institute, The University of Florida, Gainesville, FL, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, The University of Florida, Gainesville, FL, USA
- Center for Exercise Science, The University of Florida, Gainesville, FL, USA
- The Myology Institute, The University of Florida, Gainesville, FL, USA
| |
Collapse
|
27
|
Le Moal E, Liu Y, Collerette-Tremblay J, Dumontier S, Fabre P, Molina T, Dort J, Orfi Z, Denault N, Boutin J, Michaud J, Giguère H, Desroches A, Trân K, Ellezam B, Vézina F, Bedard S, Raynaud C, Balg F, Sarret P, Boudreault PL, Scott MS, Denault JB, Marsault E, Feige JN, Auger-Messier M, Dumont NA, Bentzinger CF. Apelin stimulation of the vascular skeletal muscle stem cell niche enhances endogenous repair in dystrophic mice. Sci Transl Med 2024; 16:eabn8529. [PMID: 38507466 DOI: 10.1126/scitranslmed.abn8529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/28/2024] [Indexed: 03/22/2024]
Abstract
Impaired skeletal muscle stem cell (MuSC) function has long been suspected to contribute to the pathogenesis of muscular dystrophy (MD). Here, we showed that defects in the endothelial cell (EC) compartment of the vascular stem cell niche in mouse models of Duchenne MD, laminin α2-related MD, and collagen VI-related myopathy were associated with inefficient mobilization of MuSCs after tissue damage. Using chemoinformatic analysis, we identified the 13-amino acid form of the peptide hormone apelin (AP-13) as a candidate for systemic stimulation of skeletal muscle ECs. Systemic administration of AP-13 using osmotic pumps generated a pro-proliferative EC-rich niche that supported MuSC function through angiocrine factors and markedly improved tissue regeneration and muscle strength in all three dystrophic mouse models. Moreover, EC-specific knockout of the apelin receptor led to regenerative defects that phenocopied key pathological features of MD, including vascular defects, fibrosis, muscle fiber necrosis, impaired MuSC function, and reduced force generation. Together, these studies provide in vivo proof of concept that enhancing endogenous skeletal muscle repair by targeting the vascular niche is a viable therapeutic avenue for MD and characterized AP-13 as a candidate for further study for the systemic treatment of MuSC dysfunction.
Collapse
Affiliation(s)
- Emmeran Le Moal
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Yuguo Liu
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jasmin Collerette-Tremblay
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Simon Dumontier
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Paul Fabre
- CHU Sainte-Justine Research Center, Department of Pharmacology and Physiology, School of Rehabilitation, Faculty of Medicine Université de Montréal, Montreal, QC H3T 1C5, Canada
| | - Thomas Molina
- CHU Sainte-Justine Research Center, Department of Pharmacology and Physiology, School of Rehabilitation, Faculty of Medicine Université de Montréal, Montreal, QC H3T 1C5, Canada
| | - Junio Dort
- CHU Sainte-Justine Research Center, Department of Pharmacology and Physiology, School of Rehabilitation, Faculty of Medicine Université de Montréal, Montreal, QC H3T 1C5, Canada
| | - Zakaria Orfi
- CHU Sainte-Justine Research Center, Department of Pharmacology and Physiology, School of Rehabilitation, Faculty of Medicine Université de Montréal, Montreal, QC H3T 1C5, Canada
| | - Nicolas Denault
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Joël Boutin
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Joris Michaud
- Nestlé Institute of Health Sciences, Nestlé Research, 1015 Lausanne, Switzerland
| | - Hugo Giguère
- Département de Médecine-Service de Cardiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Alexandre Desroches
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Kien Trân
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Benjamin Ellezam
- CHU Sainte-Justine Department of Pathology and Cell Biology, Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - François Vézina
- Department of Surgery, Division of Orthopedics, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Sonia Bedard
- Department of Surgery, Division of Orthopedics, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Catherine Raynaud
- Department of Surgery, Division of Orthopedics, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Frederic Balg
- Department of Surgery, Division of Orthopedics, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Philippe Sarret
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Pierre-Luc Boudreault
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Michelle S Scott
- Département de Biochimie et Génomique Fonctionnelle, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jean-Bernard Denault
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Eric Marsault
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jerome N Feige
- Nestlé Institute of Health Sciences, Nestlé Research, 1015 Lausanne, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Mannix Auger-Messier
- Département de Médecine-Service de Cardiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Nicolas A Dumont
- CHU Sainte-Justine Research Center, Department of Pharmacology and Physiology, School of Rehabilitation, Faculty of Medicine Université de Montréal, Montreal, QC H3T 1C5, Canada
| | - C Florian Bentzinger
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CHUS), Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
28
|
Suganuma M, Furutani M, Hosoyama T, Mitsumori R, Otsuka R, Takemura M, Matsui Y, Nakano Y, Niida S, Ozaki K, Satake S, Shigemizu D. Identification of Potential Blood-Based Biomarkers for Frailty by Using an Integrative Approach. Gerontology 2024; 70:630-638. [PMID: 38484720 DOI: 10.1159/000538313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 03/05/2024] [Indexed: 06/15/2024] Open
Abstract
INTRODUCTION Although frailty is a geriatric syndrome that is associated with disability, hospitalization, and mortality, it can be reversible and preventable with the appropriate interventions. Additionally, as the current diagnostic criteria for frailty include only physical, psychological, cognitive, and social measurements, there is a need for promising blood-based molecular biomarkers to aid in the diagnosis of frailty. METHODS To identify candidate blood-based biomarkers that can enhance current diagnosis of frailty, we conducted a comprehensive analysis of clinical data, messenger RNA-sequencing (RNA-seq), and aging-related factors using a total of 104 older adults aged 65-90 years (61 frail subjects and 43 robust subjects) in a cross-sectional case-control study. RESULTS We identified two candidate biomarkers of frailty from the clinical data analysis, nine from the RNA-seq analysis, and six from the aging-related factors analysis. By using combinations of the candidate biomarkers and clinical information, we constructed risk prediction models. The best models used combinations that included skeletal muscle mass index measured by dual-energy X-ray absorptiometry (adjusted p = 0.026), GDF15 (adjusted p = 1.46E-03), adiponectin (adjusted p = 0.012), CXCL9 (adjusted p = 0.011), or apelin (adjusted p = 0.020) as the biomarker. These models achieved a high area under the curve of 0.95 in an independent validation cohort (95% confidence interval: 0.79-0.97). Our risk prediction models showed significantly higher areas under the curve than did models constructed using only basic clinical information (Welch's t test p < 0.001). CONCLUSION All five biomarkers showed statistically significant correlations with components of the frailty diagnostic criteria. We discovered several potential biomarkers for the diagnosis of frailty. Further refinement may lead to their future clinical use.
Collapse
Affiliation(s)
- Mutsumi Suganuma
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Motoki Furutani
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Tohru Hosoyama
- Geroscience Research Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Risa Mitsumori
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Rei Otsuka
- Center for Gerontology and Social Science, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Marie Takemura
- Center for Frailty and Locomotive Syndrome, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yasumoto Matsui
- Center for Frailty and Locomotive Syndrome, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Shumpei Niida
- Core Facility Administration, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kouichi Ozaki
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shosuke Satake
- Center for Gerontology and Social Science, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Daichi Shigemizu
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
29
|
He Y, Liu S, Lin H, Ding F, Shao Z, Xiong L. Roles of organokines in intervertebral disc homeostasis and degeneration. Front Endocrinol (Lausanne) 2024; 15:1340625. [PMID: 38532900 PMCID: PMC10963452 DOI: 10.3389/fendo.2024.1340625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/19/2024] [Indexed: 03/28/2024] Open
Abstract
The intervertebral disc is not isolated from other tissues. Recently, abundant research has linked intervertebral disc homeostasis and degeneration to various systemic diseases, including obesity, metabolic syndrome, and diabetes. Organokines are a group of diverse factors named for the tissue of origin, including adipokines, osteokines, myokines, cardiokines, gastrointestinal hormones, and hepatokines. Through endocrine, paracrine, and autocrine mechanisms, organokines modulate energy homeostasis, oxidative stress, and metabolic balance in various tissues to mediate cross-organ communication. These molecules are involved in the regulation of cellular behavior, inflammation, and matrix metabolism under physiological and pathological conditions. In this review, we aimed to summarize the impact of organokines on disc homeostasis and degeneration and the underlying signaling mechanism. We focused on the regulatory mechanisms of organokines to provide a basis for the development of early diagnostic and therapeutic strategies for disc degeneration.
Collapse
Affiliation(s)
- Yuxin He
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Ding
- Department of Orthopaedics, JingMen Central Hospital, Jingmen, China
- Hubei Minzu University, Enshi, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liming Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Chen J, Chen H, Dong X, Hui T, Yan M, Ren D, Zou S, Wang S, Fei E, Zhang W, Lai X. Deficiency of skeletal muscle Agrin contributes to the pathogenesis of age-related sarcopenia in mice. Cell Death Dis 2024; 15:201. [PMID: 38461287 PMCID: PMC10925061 DOI: 10.1038/s41419-024-06581-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/11/2024]
Abstract
Sarcopenia, a progressive and prevalent neuromuscular disorder, is characterized by age-related muscle wasting and weakening. Despite its widespread occurrence, the molecular underpinnings of this disease remain poorly understood. Herein, we report that levels of Agrin, an extracellular matrix (ECM) protein critical for neuromuscular formation, were decreased with age in the skeletal muscles of mice. The conditional loss of Agrin in myogenic progenitors and satellite cells (SCs) (Pax7 Cre:: Agrin flox/flox) causes premature muscle aging, manifesting a distinct sarcopenic phenotype in mice. Conversely, the elevation of a miniaturized form of Agrin in skeletal muscle through adenovirus-mediated gene transfer induces enhanced muscle capacity in aged mice. Mechanistic investigations suggest that Agrin-mediated improvement in muscle function occurs through the stimulation of Yap signaling and the concurrent upregulation of dystroglycan expression. Collectively, our findings underscore the pivotal role of Agrin in the aging process of skeletal muscles and propose Agrin as a potential therapeutic target for addressing sarcopenia.
Collapse
Affiliation(s)
- Jie Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Hong Chen
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Xia Dong
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Tiankun Hui
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Min Yan
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Dongyan Ren
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Suqi Zou
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Shunqi Wang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Erkang Fei
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Wenhua Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Xinsheng Lai
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China.
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, Jiangxi, China.
| |
Collapse
|
31
|
Zheng ZF, Lin GT, Zhong Q, Wu D, Lu J, Wang JB, Chen QY, Lin JX, Cao LL, Lin M, Zheng SP, Xie JW, Zheng CH, Huang CM, Li P. Effect of sarcopenia on short-term and long-term outcomes of older patients with locally advanced gastric cancer: a multicenter study. Surg Endosc 2024; 38:1151-1162. [PMID: 38082017 DOI: 10.1007/s00464-023-10638-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 11/29/2023] [Indexed: 02/23/2024]
Abstract
OBJECTIVE To assess the effect of preoperative sarcopenia on the short-term and long-term outcomes in older patients with locally advanced gastric cancer (LAGC). METHODS Clinicopathological data of older patients with LAGC who underwent radical surgery were retrospectively analyzed. Sarcopenia was defined as a skeletal muscle index of less than 36.4 cm2/m2 for men and less than 28.4 cm2/m2 for women. Comparing the postoperative complications and survival between sarcopenia and non-sarcopenia groups using multicenter data. RESULTS A total of 406 older patients with LAGC were included in the analysis, including 145 (35.7%) with sarcopenia and 261 (64.3%) with non-sarcopenia. Multivariate logistic regression analysis showed that sarcopenia was an independent risk factor for postoperative complications with CD grade ≥ II (OR 1.616; P < 0.05). Kaplan-Meier survival curve analysis showed that the 5-year overall survival (OS) and 5-year recurrence-free survival (RFS) in the sarcopenia group were lower than those in the non-sarcopenia group (P both < 0.05). Multivariate Cox regression analyses showed that sarcopenia was an independent prognostic factor for 5-year OS and RFS (P both < 0.05). The 5-year recurrence rate in the sarcopenia group was 57.2%, which was significantly higher than that in the non-sarcopenia group (46.4%; P = 0.036). Recurrence pattern analysis showed that the incidence of distant metastases in patients with sarcopenia (42.8%) was significantly higher than non-sarcopenia (31.4%; P = 0.022). CONCLUSION Sarcopenia serves as a valuable predictor of both short-term and long-term outcomes in older patients with LAGC. Therefore, the significance of assessing preoperative nutritional status and implementing thorough postoperative follow-up for older LAGC patients with sarcopenia should be emphasized.
Collapse
Affiliation(s)
- Zi-Fang Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Department of General Surgery, Affiliated Hospital of Putian University, Putian, China
| | - Guang-Tan Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Dong Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Shu-Ping Zheng
- Public Technology Service Center, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
32
|
Jin L, Diaz-Canestro C, Wang Y, Tse MA, Xu A. Exerkines and cardiometabolic benefits of exercise: from bench to clinic. EMBO Mol Med 2024; 16:432-444. [PMID: 38321233 PMCID: PMC10940599 DOI: 10.1038/s44321-024-00027-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Regular exercise has both immediate and long-lasting benefits on cardiometabolic health, and has been recommended as a cornerstone of treatment in the management of diabetes and cardiovascular conditions. Exerkines, which are defined as humoral factors responsive to acute or chronic exercise, have emerged as important players conferring some of the multiple cardiometabolic benefits of exercise. Over the past decades, hundreds of exerkines released from skeletal muscle, heart, liver, adipose tissue, brain, and gut have been identified, and several exerkines (such as FGF21, IL-6, and adiponectin) have been exploited therapeutically as exercise mimetics for the treatment of various metabolic and cardiovascular diseases. Recent advances in metagenomics have led to the identification of gut microbiota, a so-called "hidden" metabolic organ, as an additional class of exerkines determining the efficacy of exercise in diabetes prevention, cardiac protection, and exercise performance. Furthermore, multiomics-based studies have shown the feasibility of using baseline exerkine signatures to predict individual responses to exercise with respect to metabolic and cardiorespiratory health. This review aims to explore the molecular pathways whereby exerkine networks mediate the cardiometabolic adaptations to exercise by fine-tuning inter-organ crosstalk, and discuss the roadmaps for translating exerkine-based discovery into the therapeutic application and personalized medicine in the management of the cardiometabolic disease.
Collapse
Affiliation(s)
- Leigang Jin
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Candela Diaz-Canestro
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Michael Andrew Tse
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Centre for Sports and Exercise, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.
- Department of Medicine, The University of Hong Kong, Hong Kong, China.
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
33
|
Zhu YH, Hu P, Luo YX, Yao XQ. Knowledge mapping of trends and hotspots in the field of exercise and cognition research over the past decade. Aging Clin Exp Res 2024; 36:19. [PMID: 38308660 PMCID: PMC10838253 DOI: 10.1007/s40520-023-02661-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/09/2023] [Indexed: 02/05/2024]
Abstract
Exercise elicits a wide range of physiological responses in mammalian tissues that enhance a broad range of functions, particularly in improving cognitive performance. However, the field lacks a comprehensive bibliometric analysis that clarifies its knowledge structure and research hotspots. This study aims to address this gap and map the research landscape regarding the role of exercise in cognitive function enhancement. Firstly, the frequencies and co-occurrence of keywords were analysed to identify six main clusters: aging, cognitive impairment, rehabilitation, obesity, fatigue, and hippocampus. Secondly, reference timeline co-citation analysis revealed that hippocampus and aging were the major bursts with high intensity and long attention span while children had recently emerged as a topical subject. Finally, the evolution of themes from 2012 to 2022 was analysed, and found that older adults had been the leading research theme for exercise affecting cognition. Childhood obesity was an emerging theme that attracted increasing research attention in recent years while the hippocampus research theme expanded rapidly during the decade but remained a niche topic with less relevance to others. This research identified and summarised research priorities and evolutionary trends in exercise to improve cognition by constructing knowledge networks through visual analysis. It provides researchers with a comprehensive insight into the current state of the field to facilitate further research.
Collapse
Affiliation(s)
- Ying-Hai Zhu
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Hu
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ya-Xi Luo
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Xiu-Qing Yao
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Chongqing Municipality Clinical Research Center for Geriatric Medicine, Chongqing, China.
| |
Collapse
|
34
|
Espino-Gonzalez E, Tickle PG, Altara R, Gallagher H, Cheng CW, Engman V, Wood N, Justo da Silva GJ, Scalabrin M, Yu X, Zhong Z, Colman MA, Yuldasheva NY, Booz GW, Adams V, Pereira MG, Cataliotti A, Roberts LD, Egginton S, Bowen TS. Caloric Restriction Rejuvenates Skeletal Muscle Growth in Heart Failure With Preserved Ejection Fraction. JACC Basic Transl Sci 2024; 9:223-240. [PMID: 38510717 PMCID: PMC10950401 DOI: 10.1016/j.jacbts.2023.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 03/22/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major clinical problem, with limited treatments. HFpEF is characterized by a distinct, but poorly understood, skeletal muscle pathology, which could offer an alternative therapeutic target. In a rat model, we identified impaired myonuclear accretion as a mechanism for low myofiber growth in HFpEF following resistance exercise. Acute caloric restriction rescued skeletal muscle pathology in HFpEF, whereas cardiac therapies had no effect. Mechanisms regulating myonuclear accretion were dysregulated in patients with HFpEF. Overall, these findings may have widespread implications in HFpEF, indicating combined dietary with exercise interventions as a beneficial approach to overcome skeletal muscle pathology.
Collapse
Affiliation(s)
- Ever Espino-Gonzalez
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Peter G. Tickle
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Raffaele Altara
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Harrison Gallagher
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Chew W. Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine, University of Leeds, Leeds, United Kingdom
| | - Viktor Engman
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Nathanael Wood
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | | | - Mattia Scalabrin
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Xinyue Yu
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Ziyi Zhong
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Michael A. Colman
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Nadira Y. Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine, University of Leeds, Leeds, United Kingdom
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Volker Adams
- Heart Center Dresden, TU-Dresden, Dresden, Germany
| | - Marcelo G. Pereira
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Alessandro Cataliotti
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Lee D. Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine, University of Leeds, Leeds, United Kingdom
| | - Stuart Egginton
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - T. Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
35
|
Zhang K, Xie N, Ye H, Miao J, Xia B, Yang Y, Peng H, Xu S, Wu T, Tao C, Ruan J, Wang Y, Yang S. Glucose restriction enhances oxidative fiber formation: A multi-omic signal network involving AMPK and CaMK2. iScience 2024; 27:108590. [PMID: 38161415 PMCID: PMC10755363 DOI: 10.1016/j.isci.2023.108590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/23/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Skeletal muscle is a highly plastic organ that adapts to different metabolic states or functional demands. This study explored the impact of permanent glucose restriction (GR) on skeletal muscle composition and metabolism. Using Glut4m mice with defective glucose transporter 4, we conducted multi-omics analyses at different ages and after low-intensity treadmill training. The oxidative fibers were significantly increased in Glut4m muscles. Mechanistically, GR activated AMPK pathway, promoting mitochondrial function and beneficial myokine expression, and facilitated slow fiber formation via CaMK2 pathway. Phosphorylation-activated Perm1 may synergize AMPK and CaMK2 signaling. Besides, MAPK and CDK kinases were also implicated in skeletal muscle protein phosphorylation during GR response. This study provides a comprehensive signaling network demonstrating how GR influences muscle fiber types and metabolic patterns. These insights offer valuable data for understanding oxidative fiber formation mechanisms and identifying clinical targets for metabolic diseases.
Collapse
Affiliation(s)
- Kaiyi Zhang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA Teaching and Research Centre, Liège University, 5030 Gembloux, Belgium
| | - Ning Xie
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Huaqiong Ye
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Jiakun Miao
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Boce Xia
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Yu Yang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Huanqi Peng
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Shuang Xu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Tianwen Wu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Cong Tao
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Jinxue Ruan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Yanfang Wang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| | - Shulin Yang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China
| |
Collapse
|
36
|
Bian X, Wang Q, Wang Y, Lou S. The function of previously unappreciated exerkines secreted by muscle in regulation of neurodegenerative diseases. Front Mol Neurosci 2024; 16:1305208. [PMID: 38249295 PMCID: PMC10796786 DOI: 10.3389/fnmol.2023.1305208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/05/2023] [Indexed: 01/23/2024] Open
Abstract
The initiation and progression of neurodegenerative diseases (NDs), distinguished by compromised nervous system integrity, profoundly disrupt the quality of life of patients, concurrently exerting a considerable strain on both the economy and the social healthcare infrastructure. Exercise has demonstrated its potential as both an effective preventive intervention and a rehabilitation approach among the emerging therapeutics targeting NDs. As the largest secretory organ, skeletal muscle possesses the capacity to secrete myokines, and these myokines can partially improve the prognosis of NDs by mediating the muscle-brain axis. Besides the well-studied exerkines, which are secreted by skeletal muscle during exercise that pivotally exert their beneficial function, the physiological function of novel exerkines, e.g., apelin, kynurenic acid (KYNA), and lactate have been underappreciated previously. Herein, this review discusses the roles of these novel exerkines and their mechanisms in regulating the progression and improvement of NDs, especially the significance of their functions in improving NDs' prognoses through exercise. Furthermore, several myokines with potential implications in ameliorating ND progression are proposed as the future direction for investigation. Elucidation of the function of exerkines secreted by skeletal muscle in the regulation of NDs advances the understanding of its pathogenesis and facilitates the development of therapeutics that intervene in these processes to cure NDs.
Collapse
Affiliation(s)
- Xuepeng Bian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Qian Wang
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Yibing Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Shujie Lou
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
37
|
Zhao Z, Yan K, Guan Q, Guo Q, Zhao C. Mechanism and physical activities in bone-skeletal muscle crosstalk. Front Endocrinol (Lausanne) 2024; 14:1287972. [PMID: 38239981 PMCID: PMC10795164 DOI: 10.3389/fendo.2023.1287972] [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: 09/03/2023] [Accepted: 12/05/2023] [Indexed: 01/22/2024] Open
Abstract
Bone and skeletal muscle work in coordination to maintain the function of the musculoskeletal system, in which skeletal muscle contraction drives the movement of the bone lever system while bone provides insert sites for skeletal muscle through the bone-muscle junction. Existing evidence suggests that factors secreted by skeletal muscle and bone mediate the interaction between the two tissues. Herein, we focused on the relationship between skeletal muscle and bone and the underlying mechanism of the interaction. Exercise can promote bone strength and secrete osteocalcin and insulin-like growth factor I into the blood, thus improving muscle quality. In addition, exercise can also promote myostatin, interleukin-6, Irisin, and apelin in muscles to enter the blood so that they can act on bones to maintain the balance between bone absorption and bone formation. There is a special regulatory axis interleukin-6/osteocalcin between myokines and osteokines, which is mainly influenced by exercise. Therefore, we pay attention to the important factors in the bone-muscle intersection that are affected by exercise, which were found or their functions were expanded, which strengthened the connection between organs of the whole body, highlighting the importance of exercise and contributing to the diagnosis, prevention, and treatment of osteoporosis and sarcopenia in the clinic.
Collapse
Affiliation(s)
- Zhonghan Zhao
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Kai Yan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Qiao Guan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Qiang Guo
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Can Zhao
- College of Athletic Performance, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
38
|
Hosseini L, Shahabi P, Fakhari A, Zangbar HS, Seyedaghamiri F, Sadeghzadeh J, Abolhasanpour N. Aging and age-related diseases with a focus on therapeutic potentials of young blood/plasma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1-13. [PMID: 37552316 DOI: 10.1007/s00210-023-02657-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 07/31/2023] [Indexed: 08/09/2023]
Abstract
Aging is accompanied by alterations in the body with time-related to decline of physiological integrity and functionality process, responsible for increasing diseases and vulnerability to death. Several ages associated with biomarkers were observed in red blood cells, and consequently plasma proteins have a critical rejuvenating role in the aging process and age-related disorders. Advanced age is a risk factor for a broad spectrum of diseases and disorders such as cardiovascular diseases, musculoskeletal disorders and liver, chronic kidney disease, neurodegenerative diseases, and cancer because of loss of regenerative capacity, correlated to reduced systemic factors and raise of pro-inflammatory cytokines. Most studies have shown that systemic factors in young blood/plasma can strongly protect against age-related diseases in various tissues by restoring autophagy, increasing neurogenesis, and reducing oxidative stress, inflammation, and apoptosis. Here, we focus on the current advances in using young plasma or blood to combat aging and age-related diseases and summarize the experimental and clinical evidence supporting this approach. Based on reports, young plasma or blood is new a therapeutic approach to aging and age-associated diseases.
Collapse
Affiliation(s)
- Leila Hosseini
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Parviz Shahabi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, IR, Iran
| | - Ali Fakhari
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Soltani Zangbar
- Department of Neurosciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemehsadat Seyedaghamiri
- Department of Neurosciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Sadeghzadeh
- Department of Neurosciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasrin Abolhasanpour
- Research Center for Evidence-Based Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
39
|
Feng Z, Wu Z, Ma Y, Chen Q, Liu Z, Qiu Y, Zhu Z. Higher Baseline Serum Myokine of FSTL1 May Serve as a Potential Predictive Biomarker for Successful Brace Treatment in Girls With Adolescent Idiopathic Scoliosis. Spine (Phila Pa 1976) 2023; 48:1756-1762. [PMID: 37339276 DOI: 10.1097/brs.0000000000004751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/04/2023] [Indexed: 06/22/2023]
Abstract
STUDY DESIGN A retrospective case-control study. OBJECTIVE This study aimed to investigate whether myokine, which is related to exercise and muscle mass, could serve as a biomarker for predicting bracing outcomes. SUMMARY OF BACKGROUND DATA Several risk factors have been documented to be associated with bracing failure in patients with adolescent idiopathic scoliosis (AIS). However, serum biomarkers have not been extensively explored. PATIENTS AND METHODS Skeletally immature females with AIS, without previous histories of bracing or surgery, were included. Peripheral blood was collected at the time of the bracing prescription. Baseline serum concentrations of 8 myokines [apelin, fractalkine, brain-derived neurotrophic factor, erythropoietin, osteonectin, fatty-acid-binding protein 3, follistatin-like 1 (FSTL1), and musclin] were measured by multiplex assays. Patients were followed up until weaned from bracing and then designated as a "failure" (defined as Cobb angle progression >5°) or "success." A logistic regression analysis was performed that accounted for serum myokines and skeletal maturity. RESULTS We included 117 patients, with 27 in the failure group. Patients in the failure group had lower initial Risser sign and lower baseline serum levels of myokines, including FSTL1 (2217.3 ± 617.0 vs . 1369.3 ± 704.9, P = 0.002), apelin [116.5 (12.0, 335.9) vs . 83.5 (10.5, 221.1), P = 0.016], fractalkine (979.6 ± 457.8 vs . 743.8 ± 456.1, P = 0.020), and musclin [211.3 (16.3, 370.3) vs . 67.8 (15.5, 325.6), P = 0.049]. Following adjusted analysis, serum FSTL1 [odds ratio = 10.460; (2.213-49.453)] was determined to be predictive of bracing effectiveness. CONCLUSION Patients who failed AIS bracing had significantly lower mean baseline levels of FSTL1 than those who achieved success. FSTL1 may serve as a biomarker that can inform outcomes after bracing.
Collapse
Affiliation(s)
- Zhenhua Feng
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Joint Scoliosis Research Center of The Chinese University of Hong Kong and Nanjing University, Nanjing and Hong Kong, China
| | - Zhichong Wu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Joint Scoliosis Research Center of The Chinese University of Hong Kong and Nanjing University, Nanjing and Hong Kong, China
| | - Yanyu Ma
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Quanchi Chen
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhen Liu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Joint Scoliosis Research Center of The Chinese University of Hong Kong and Nanjing University, Nanjing and Hong Kong, China
| | - Yong Qiu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Joint Scoliosis Research Center of The Chinese University of Hong Kong and Nanjing University, Nanjing and Hong Kong, China
| | - Zezhang Zhu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Joint Scoliosis Research Center of The Chinese University of Hong Kong and Nanjing University, Nanjing and Hong Kong, China
| |
Collapse
|
40
|
Jia S, Yu Z, Bai L. Exerkines and osteoarthritis. Front Physiol 2023; 14:1302769. [PMID: 38107476 PMCID: PMC10722202 DOI: 10.3389/fphys.2023.1302769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/21/2023] [Indexed: 12/19/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent chronic joint disease, with physical exercise being a widely endorsed strategy in its management guidelines. Exerkines, defined as cytokines secreted in response to acute and chronic exercise, function through endocrine, paracrine, and/or autocrine pathways. Various tissue-specific exerkines, encompassing exercise-induced myokines (muscle), cardiokines (heart), and adipokines (adipose tissue), have been linked to exercise therapy in OA. Exerkines are derived from these kines, but unlike them, only kines regulated by exercise can be called exerkines. Some of these exerkines serve a therapeutic role in OA, such as irisin, metrnl, lactate, secreted frizzled-related protein (SFRP), neuregulin, and adiponectin. While others may exacerbate the condition, such as IL-6, IL-7, IL-15, IL-33, myostatin, fractalkine, follistatin-like 1 (FSTL1), visfatin, activin A, migration inhibitory factor (MIF), apelin and growth differentiation factor (GDF)-15. They exerts anti-/pro-apoptosis/pyroptosis/inflammation, chondrogenic differentiation and cell senescence effect in chondrocyte, synoviocyte and mesenchymal stem cell. The modulation of adipokine effects on diverse cell types within the intra-articular joint emerges as a promising avenue for future OA interventions. This paper reviews recent findings that underscore the significant role of tissue-specific exerkines in OA, delving into the underlying cellular and molecular mechanisms involved.
Collapse
Affiliation(s)
- Shuangshuo Jia
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ziyao Yu
- Imaging Department, Dalian Medical University, Dalian, China
| | - Lunhao Bai
- Department of Orthopedic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
41
|
de Souto Barreto P, Rolland Y, Ferrucci L, Arai H, Bischoff-Ferrari H, Duque G, Fielding RA, Beard JR, Muscedere J, Sierra F, Vellas B, LeBrasseur NK. Looking at frailty and intrinsic capacity through a geroscience lens: the ICFSR & Geroscience Task Force. NATURE AGING 2023; 3:1474-1479. [PMID: 37985720 DOI: 10.1038/s43587-023-00531-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Affiliation(s)
- Philipe de Souto Barreto
- IHU HealthAge, Toulouse, France.
- CERPOP, Inserm 1295, Université de Toulouse, UPS, Toulouse, France.
- Institute on Aging of the Toulouse University Hospital (CHU-Toulouse), Toulouse, France.
| | - Yves Rolland
- IHU HealthAge, Toulouse, France
- CERPOP, Inserm 1295, Université de Toulouse, UPS, Toulouse, France
- Institute on Aging of the Toulouse University Hospital (CHU-Toulouse), Toulouse, France
| | - Luigi Ferrucci
- Intramural Research Program of the National Institute on Aging
| | - Hidenori Arai
- National Center for Geriatrics and Gerontology, Obu, Japan
| | - Heike Bischoff-Ferrari
- IHU HealthAge, Toulouse, France
- Department of Geriatrics and Aging Research, University of Zurich, Zurich, Switzerland
| | - Gustavo Duque
- Bone, Muscle & Geroscience Research Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Roger A Fielding
- Nutrition, Exercise Physiology, and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - John R Beard
- International Longevity Center-USA, Columbia University, New York, NY, USA
| | - John Muscedere
- Department of Critical Care Medicine, Queen's University, Kingston, Ontario, Canada
| | | | - Bruno Vellas
- IHU HealthAge, Toulouse, France
- CERPOP, Inserm 1295, Université de Toulouse, UPS, Toulouse, France
- Institute on Aging of the Toulouse University Hospital (CHU-Toulouse), Toulouse, France
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
42
|
Rolland Y, Dray C, Vellas B, Barreto PDS. Current and investigational medications for the treatment of sarcopenia. Metabolism 2023; 149:155597. [PMID: 37348598 DOI: 10.1016/j.metabol.2023.155597] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/20/2023] [Accepted: 05/25/2023] [Indexed: 06/24/2023]
Abstract
Sarcopenia, defined as the loss of muscle mass and function, is a widely prevalent and severe condition in older adults. Since 2016, it is recognized as a disease. Strength exercise training and nutritional support are the frontline treatment of sarcopenia, with no drug currently approved for this indication. However, new therapeutic options are emerging. In this review, we evidenced that only very few trials have focused on sarcopenia/sarcopenic patients. Most drug trials were performed in different clinical older populations (e.g., men with hypogonadism, post-menopausal women at risk for osteoporosis), and their efficacy were tested separately on the components of sarcopenia (muscle mass, muscle strength and physical performances). Results from trials testing the effects of Testosterone, Selective Androgen Receptor Modulators (SARMs), Estrogen, Dehydroepiandrosterone (DHEA), Insulin-like Growth Factor-1 (IGF-1), Growth Hormone (GH), GH Secretagogue (GHS), drug targeting Myostatin and Activin receptor pathway, Vitamin D, Angiotensin Converting Enzyme inhibitors (ACEi) and Angiotensin Receptor Blockers (ARBs), or β-blockers, were compiled. Although some drugs have been effective in improving muscle mass and/or strength, this was not translated into clinically relevant improvements on physical performance. Finally, some promising molecules investigated in on-going clinical trials and in pre-clinical phase were summarized, including apelin and irisin.
Collapse
Affiliation(s)
- Yves Rolland
- Gérontopôle de Toulouse, IHU HealthAge, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France; CERPOP UMR 1295, University of Toulouse III, Inserm, UPS, Toulouse, France.
| | - Cedric Dray
- Université de Toulouse III Université Paul Sabatier, Toulouse, France; Restore, a geroscience and rejuvenation research center, UMR 1301-Inserm, 5070-CNRS EFS, France
| | - Bruno Vellas
- Gérontopôle de Toulouse, IHU HealthAge, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France; CERPOP UMR 1295, University of Toulouse III, Inserm, UPS, Toulouse, France
| | - Philipe De Souto Barreto
- Gérontopôle de Toulouse, IHU HealthAge, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France; CERPOP UMR 1295, University of Toulouse III, Inserm, UPS, Toulouse, France
| |
Collapse
|
43
|
Memczak S, Belmonte JC. Overcoming muscle stem cell aging. Curr Opin Genet Dev 2023; 83:102127. [PMID: 37839315 DOI: 10.1016/j.gde.2023.102127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/06/2023] [Accepted: 09/19/2023] [Indexed: 10/17/2023]
Abstract
Reduced muscle strength and mass is one of the hallmarks of physiological aging in humans and can result in severe impairment of the quality of life. In part this is caused by a functional loss of the highly specialized muscle stem cells (MuSCs), which in healthy conditions provide maintenance, growth, and regeneration. Recent progress in understanding of the stem cell niche and results from single cell technologies reveal exciting insights at unprecedented detail into MuSCs and muscle biology during aging. Here, we review this field and discuss the implications of current findings with a focus on cellular reprogramming approaches as a novel therapeutic avenue for age-related muscle decline.
Collapse
|
44
|
Bondi D, Bevere M, Piccirillo R, Sorci G, Di Felice V, Re Cecconi AD, D'Amico D, Pietrangelo T, Fulle S. Integrated procedures for accelerating, deepening, and leading genetic inquiry: A first application on human muscle secretome. Mol Genet Metab 2023; 140:107705. [PMID: 37837864 DOI: 10.1016/j.ymgme.2023.107705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/15/2023] [Accepted: 10/01/2023] [Indexed: 10/16/2023]
Abstract
PURPOSE Beyond classical procedures, bioinformatic-assisted approaches and computational biology offer unprecedented opportunities for scholars. However, these amazing possibilities still need epistemological criticism, as well as standardized procedures. Especially those topics with a huge body of data may benefit from data science (DS)-assisted methods. Therefore, the current study dealt with the combined expert-assisted and DS-assisted approaches to address the broad field of muscle secretome. We aimed to apply DS tools to fix the literature research, suggest investigation targets with a data-driven approach, predict possible scenarios, and define a workflow. METHODS Recognized scholars with expertise on myokines were invited to provide a list of the most important myokines. GeneRecommender, GeneMANIA, HumanNet, and STRING were selected as DS tools. Networks were built on STRING and GeneMANIA. The outcomes of DS tools included the top 5 recommendations. Each expert-led discussion has been then integrated with an DS-led approach to provide further perspectives. RESULTS Among the results, 11 molecules had already been described as bona-fide myokines in literature, and 11 molecules were putative myokines. Most of the myokines and the putative myokines recommended by the DS tools were described as present in the cargo of extracellular vesicles. CONCLUSIONS Including both supervised and unsupervised learning methods, as well as encompassing algorithms focused on both protein interaction and gene represent a comprehensive approach to tackle complex biomedical topics. DS-assisted methods for reviewing existent evidence, recommending targets of interest, and predicting original scenarios are worth exploring as in silico recommendations to be integrated with experts' ideas for optimizing molecular studies.
Collapse
Affiliation(s)
- Danilo Bondi
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| | - Michele Bevere
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.
| | - Rosanna Piccirillo
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy.
| | - Guglielmo Sorci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| | - Valentina Di Felice
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| | - Andrea David Re Cecconi
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy.
| | - Daniela D'Amico
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| | - Tiziana Pietrangelo
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| | - Stefania Fulle
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| |
Collapse
|
45
|
Piétri-Rouxel F, Falcone S, Traoré M. [GDF5: a therapeutic candidate for combating sarcopenia]. Med Sci (Paris) 2023; 39 Hors série n° 1:47-53. [PMID: 37975770 DOI: 10.1051/medsci/2023143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023] Open
Abstract
Sarcopenia is a complex age-related muscular disease affecting 10 to 16 % of people over 65 years old. It is characterized by excessive loss of muscle mass and strength. Despite a plethora of studies aimed at understanding the physiological mechanisms underlying this pathology, the pathophysiology of sarcopenia remains poorly understood. To date, there is no pharmacological treatment for this disease. In this context, our team develop therapeutic approaches based on the GDF5 protein to counteract the loss of muscle mass and function in various pathological conditions, including sarcopenia. After deciphering one of the molecular mechanisms governing GDF5 expression, we have demonstrated the therapeutic potential of this protein in the preservation of muscle mass and strength in aged mice.
Collapse
Affiliation(s)
- France Piétri-Rouxel
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Sestina Falcone
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| | - Massiré Traoré
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, F-75013 Paris, France
| |
Collapse
|
46
|
Zhang M, Ning J, Lu Y. Apelin alleviates sepsis-induced acute lung injury in part by modulating the SIRT1/NLRP3 pathway to inhibit endothelial cell pyroptosis. Tissue Cell 2023; 85:102251. [PMID: 39491401 DOI: 10.1016/j.tice.2023.102251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 11/05/2024]
Abstract
BACKGROUND Sepsis, an intricate systemic inflammatory syndrome, gives rise to various life-threatening complications, with acute lung injury (ALI) being prominently encountered. ALI, clinically characterized by pulmonary infiltration, hypoxemia, and edema, stands as a prevailing consequence of sepsis. This work sought to elucidate the mechanism of Apelin in mitigating sepsis-induced ALI (siALI). METHODS A mouse sepsis model was constructed by cecal ligation and puncture surgery, followed utilizing histopathological analysis using HE staining. mRNA levels of inflammatory cytokines (IL-1β, IL-6, and TNF-α) were assessed utilizing qRT-PCR, while ELISA was employed to measure the levels of vWF, VEGF, IL-1β, and IL-18. Western blot was conducted to examine protein levels of NLRP3, Caspase-1 p20, GSDMD-N, and SIRT1. To evaluate the extent of endothelial cell (EC) pyroptosis, immunofluorescence co-staining of CD31, NLRP3, and Caspase-1 p20 was fulfilled. Furthermore, TUNEL staining was utilized to ascertain the degree of plasma membrane damage and cell death. RESULTS Apelin demonstrated its potential in ameliorating siALI in mice by diminishing mRNA expression levels of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) as well as expression levels of vWF and VEGF. Apelin inhibited protein expression of NLRP3, Caspase-1 p20, and GSDMD-N, indicating that EC pyroptosis was suppressed. Finally, Apelin could upregulate the protein expression of SIRT1. This upregulation led to the inhibition of protein expression of NLRP3, Caspase-1 p20, and GSDMD-N, consequently suppressing EC pyroptosis. As a result, a reduction in the expression of inflammatory cytokines IL-1β and IL-18 ultimately alleviated siALI. CONCLUSION Apelin was confirmed to alleviate siALI partially by modulating SIRT1/NLRP3 pathway to inhibit EC pyroptosis, which dawned on the molecular mechanism of siALI and had important clinical significance for treating ALI effectively.
Collapse
Affiliation(s)
- Manyan Zhang
- Department of Respiration, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Jiyu Ning
- Department of General Practice, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yu Lu
- Department of General Practice, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
47
|
Shi M, Fang Y, Liang Y, Hu Y, Huang J, Xia W, Bian H, Zhuo Q, Wu L, Zhao C. Identification and characterization of differentially expressed circular RNAs in extraocular muscle of oculomotor nerve palsy. BMC Genomics 2023; 24:617. [PMID: 37848864 PMCID: PMC10583365 DOI: 10.1186/s12864-023-09733-3] [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: 06/09/2023] [Accepted: 10/11/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Oculomotor nerve palsy (ONP) is a neuroparalytic disorder resulting in dysfunction of innervating extraocular muscles (EOMs), of which the pathological characteristics remain underexplored. METHODS In this study, medial rectus muscle tissue samples from four ONP patients and four constant exotropia (CXT) patients were collected for RNA sequencing. Differentially expressed circular RNAs (circRNAs) were identified and included in functional enrichment analysis, followed by interaction analysis with microRNAs and mRNAs as well as RNA binding proteins. Furthermore, RT-qPCR was used to validate the expression level of the differentially expressed circRNAs. RESULTS A total of 84 differentially expressed circRNAs were identified from 10,504 predicted circRNAs. Functional enrichment analysis indicated that the differentially expressed circRNAs significantly correlated with skeletal muscle contraction. In addition, interaction analyses showed that up-regulated circRNA_03628 was significantly interacted with RNA binding protein AGO2 and EIF4A3 as well as microRNA hsa-miR-188-5p and hsa-miR-4529-5p. The up-regulation of circRNA_03628 was validated by RT-qPCR, followed by further elaboration of the expression, location and clinical significance of circRNA_03628 in EOMs of ONP. CONCLUSIONS Our study may shed light on the role of differentially expressed circRNAs, especially circRNA_03628, in the pathological changes of EOMs in ONP.
Collapse
Affiliation(s)
- Mingsu Shi
- Eye Institute, Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Yanxi Fang
- Eye Institute, Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Yu Liang
- Eye Institute, Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Yuxiang Hu
- Eye Institute, Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Jiaqiu Huang
- Eye Institute, Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Weiyi Xia
- Eye Institute, Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Hewei Bian
- Eye Institute, Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Qiao Zhuo
- Eye Institute, Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Lianqun Wu
- Eye Institute, Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China.
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China.
| | - Chen Zhao
- Eye Institute, Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China.
- NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China.
| |
Collapse
|
48
|
Kim SR, Lee EI, Kim Y, Cho SW, Hong N, Rhee Y, Park JW. Preclinical assessment of rodent jumping power with a novel electrical stimulation-assisted device. Sci Rep 2023; 13:17371. [PMID: 37833409 PMCID: PMC10575903 DOI: 10.1038/s41598-023-44748-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 10/11/2023] [Indexed: 10/15/2023] Open
Abstract
Sarcopenia is a progressive loss of muscle mass and strength that is associated with increasing the risk of falls, musculoskeletal diseases, and chronic metabolic diseases. However, the animal models adopted to study sarcopenia face limitations since the functional tests conducted on human cannot be directly adapted to animals because the animals do not follow instructions. Moreover, current preclinical research tools for muscle function assessment, such as the rotarod, grip strength, and treadmill, have limitations, including low-intensity simple movements, evaluator subjectivity, and limited power indicators. Hence, in this study, we present a new jumping-power assessment tool in a preclinical rodent model to demonstrate muscle functions. To overcome the light weight and command issues in the rodent model, we developed an electrical stimulation-assisted jump power assessment device. Precisely, the device utilizes a load cell with a 0.1 g resolution and a 50 points/s data acquisition rate to capture the short period of the mouse jump. Additionally, interdigitated electrodes are used to electrically stimulate the mice and make them jump. While our primary focus in this article is the validation of the newly developed jump power assessment device, it is worth noting that this tool has several potential utilities. These include the phenotypic comparison of sarcopenia models, the exploration of muscle function reduction mechanisms, muscle function-related blood biomarkers, and the evaluation of drug intervention effects.
Collapse
Affiliation(s)
- Seung-Rok Kim
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Ey-In Lee
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Yongnyun Kim
- Yonsei University Health System, Seoul, 03722, Korea
| | - Sang Wouk Cho
- Department of Integrative Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institue for Innovation in Digital Healthcare (IIDH), Severance Hospital, Seoul, Korea
| | - Namki Hong
- Institue for Innovation in Digital Healthcare (IIDH), Severance Hospital, Seoul, Korea.
- Department of Internal Medicine, Severance Hospital, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.
| | - Yumie Rhee
- Department of Internal Medicine, Severance Hospital, Endocrine Research Institute, Yonsei University College of Medicine, Seoul, 03722, Korea.
| | - Jin-Woo Park
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| |
Collapse
|
49
|
Smith JAB, Murach KA, Dyar KA, Zierath JR. Exercise metabolism and adaptation in skeletal muscle. Nat Rev Mol Cell Biol 2023; 24:607-632. [PMID: 37225892 PMCID: PMC10527431 DOI: 10.1038/s41580-023-00606-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 05/26/2023]
Abstract
Viewing metabolism through the lens of exercise biology has proven an accessible and practical strategy to gain new insights into local and systemic metabolic regulation. Recent methodological developments have advanced understanding of the central role of skeletal muscle in many exercise-associated health benefits and have uncovered the molecular underpinnings driving adaptive responses to training regimens. In this Review, we provide a contemporary view of the metabolic flexibility and functional plasticity of skeletal muscle in response to exercise. First, we provide background on the macrostructure and ultrastructure of skeletal muscle fibres, highlighting the current understanding of sarcomeric networks and mitochondrial subpopulations. Next, we discuss acute exercise skeletal muscle metabolism and the signalling, transcriptional and epigenetic regulation of adaptations to exercise training. We address knowledge gaps throughout and propose future directions for the field. This Review contextualizes recent research of skeletal muscle exercise metabolism, framing further advances and translation into practice.
Collapse
Affiliation(s)
- Jonathon A B Smith
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Kevin A Murach
- Molecular Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, USA
| | - Kenneth A Dyar
- Metabolic Physiology, Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
50
|
Xie G, Jin H, Mikhail H, Pavel V, Yang G, Ji B, Lu B, Li Y. Autophagy in sarcopenia: Possible mechanisms and novel therapies. Biomed Pharmacother 2023; 165:115147. [PMID: 37473679 DOI: 10.1016/j.biopha.2023.115147] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/23/2023] [Accepted: 07/07/2023] [Indexed: 07/22/2023] Open
Abstract
With global population aging, age-related diseases, especially sarcopenia, have attracted much attention in recent years. Characterized by low muscle strength, low muscle quantity or quality and low physical performance, sarcopenia is one of the major factors associated with an increased risk of falls and disability. Much effort has been made to understand the cellular biological and physiological mechanisms underlying sarcopenia. Autophagy is an important cellular self-protection mechanism that relies on lysosomes to degrade misfolded proteins and damaged organelles. Research designed to obtain new insight into human diseases from the autophagic aspect has been carried out and has made new progress, which encourages relevant studies on the relationship between autophagy and sarcopenia. Autophagy plays a protective role in sarcopenia by modulating the regenerative capability of satellite cells, relieving oxidative stress and suppressing the inflammatory response. This review aims to reveal the specific interaction between sarcopenia and autophagy and explore possible therapies in hopes of encouraging more specific research in need and unlocking novel promising therapies to ameliorate sarcopenia.
Collapse
Affiliation(s)
- Guangyang Xie
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China
| | - Hongfu Jin
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Herasimenka Mikhail
- Republican Scientific and Practical Center of Traumatology and Orthopedics, Minsk 220024, Belarus
| | - Volotovski Pavel
- Republican Scientific and Practical Center of Traumatology and Orthopedics, Minsk 220024, Belarus
| | - Guang Yang
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Bingzhou Ji
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Bangbao Lu
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|