51
|
Xin H, Huang R, Zhou M, Chen J, Zhang J, Zhou T, Ji S, Liu X, Tian H, Lam SM, Bao X, Li L, Tong S, Deng F, Shui G, Zhang Z, Wong CCL, Li MD. Daytime-restricted feeding enhances running endurance without prior exercise in mice. Nat Metab 2023; 5:1236-1251. [PMID: 37365376 DOI: 10.1038/s42255-023-00826-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 05/17/2023] [Indexed: 06/28/2023]
Abstract
Physical endurance and energy conservation are essential for survival in the wild. However, it remains unknown whether and how meal timing regulates physical endurance and muscle diurnal rhythms. Here, we show that day/sleep time-restricted feeding (DRF) enhances running endurance by 100% throughout the circadian cycle in both male and female mice, compared to either ad libitum feeding or night/wake time-restricted feeding. Ablation of the circadian clock in the whole body or the muscle abolished the exercise regulatory effect of DRF. Multi-omics analysis revealed that DRF robustly entrains diurnal rhythms of a mitochondrial oxidative metabolism-centric network, compared to night/wake time-restricted feeding. Remarkably, muscle-specific knockdown of the myocyte lipid droplet protein perilipin-5 completely mimics DRF in enhancing endurance, enhancing oxidative bioenergetics and outputting rhythmicity to circulating energy substrates, including acylcarnitine. Together, our work identifies a potent dietary regimen to enhance running endurance without prior exercise, as well as providing a multi-omics atlas of muscle circadian biology regulated by meal timing.
Collapse
Affiliation(s)
- Haoran Xin
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Rongfeng Huang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Meiyu Zhou
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jianghui Chen
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
- Department of Cardiology, Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianxin Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Tingting Zhou
- Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Shushen Ji
- Department of Bioinformatics, GFK Biotech, Shanghai, China
| | - Xiao Liu
- Department of Bioinformatics, GFK Biotech, Shanghai, China
| | - He Tian
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- LipidALL Technologies, Changzhou, China
| | - Xinyu Bao
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Lihua Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China
| | - Shifei Tong
- Department of Cardiology, Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Deng
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Zhihui Zhang
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China.
| | - Catherine C L Wong
- Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.
- Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China.
| | - Min-Dian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, Southwest Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
52
|
Liu J, Song Y, Wang Y, Hong H. Vitamin D/vitamin D receptor pathway in non-alcoholic fatty liver disease. Expert Opin Ther Targets 2023; 27:1145-1157. [PMID: 37861098 DOI: 10.1080/14728222.2023.2274099] [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: 03/16/2023] [Accepted: 10/18/2023] [Indexed: 10/21/2023]
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease worldwide, but underlying mechanisms are not fully understood. In recent years, a growing body of evidence has emphasized the therapeutic role of vitamin D in NAFLD, but the specific mechanism remains to be investigated. AREAS COVERED This review summarized the roles of vitamin D/VDR (vitamin D receptor) pathway in different types of liver cells (such as hepatocytes, hepatic stellate cells, liver macrophages, T lymphocytes, and other hepatic immune cells) in case of NAFLD. Meanwhile, the effects of pathways in the gut-liver axis, adipose tissue-liver axis, and skeletal muscle-liver axis on the development of NAFLD were further reviewed. Relevant literature was searched on PubMed for the writing of this review. EXPERT OPINION The precise regulation of regional vitamin D/VDR signaling pathway based on cell-specific or tissue-specific function will help clarify the potential mechanism of vitamin D in NAFLD, which may provide new therapeutic targets to improve the safety and efficacy of vitamin D based drugs.
Collapse
Affiliation(s)
- Jingqi Liu
- Fujian Key Laboratory of Vascular Aging, Department of Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Xiamen Institute of Geriatric Rehabilitation, Department of Geriatrics, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Yang Song
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Ye Wang
- Xiamen Institute of Geriatric Rehabilitation, Department of Geriatrics, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Huashan Hong
- Fujian Key Laboratory of Vascular Aging, Department of Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
53
|
Correa-de-Araujo R. The Public Health Need and Strategic Opportunities for the Accelerated Development of Function-Promoting Therapies for Older Adults. J Gerontol A Biol Sci Med Sci 2023; 78:1-3. [PMID: 37325964 PMCID: PMC10272975 DOI: 10.1093/gerona/glad076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Indexed: 06/17/2023] Open
Affiliation(s)
- Rosaly Correa-de-Araujo
- Division of Geriatrics and Clinical Gerontology, National Institute on Aging, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, USA
| |
Collapse
|
54
|
Golec M, Zembala-John J, Fronczek M, Konka A, Bochenek A, Wystyrk K, Botor H, Zalewska M, Chrapiec M, Kasperczyk S, Brzoza Z, Bułdak RJ. Relationship between anthropometric and body composition parameters and anti-SARS-CoV-2 specific IgG titers in females vaccinated against COVID-19 according to the heterologous vaccination course: A cohort study. PLoS One 2023; 18:e0287128. [PMID: 37310975 DOI: 10.1371/journal.pone.0287128] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
INTRODUCTION The aim of this cohort study was to evaluate the relationship between anthropometric and body composition parameters and anti-SARS-CoV-2 IgG titers in a group of females who were vaccinated against COVID-19 with two doses of ChAdOx1 vaccine and then boosted with the BNT162b2 vaccine. MATERIALS AND METHODS The study group consisted of 63 women. Basic demographic and clinical data were collected. To assess the anti-SARS-CoV-2 immunoglobulin G titers following the vaccination, five blood draws were performed: 1) before the first dose, 2) before the second dose, 3) 14-21 days after the primary vaccination, 4) before the booster, and 5) 21 days after the booster. Blood samples were analyzed using a two-step enzymatic chemiluminescent assay. Body mass index and body composition were evaluated using bioelectrical impedance analysis. To select the most distinguishing parameters and correlations between anthropometric and body composition parameters and anti-SARS-CoV-2 IgG titers, factor analysis using the Principal Component Analysis was conducted. RESULTS Sixty-three females (mean age: 46.52 years) who met the inclusion criteria were enrolled. 40 of them (63.50%) participated in the post-booster follow-up. After receiving two doses of the ChAdOx1 vaccine, the study group's anti-SARS-CoV-2 IgG titers were 67.19 ± 77.44 AU/mL (mean ± SD), whereas after receiving a heterologous mRNA booster, the level of anti-SARS-CoV-2 IgG titers was about three-times higher and amounted to 212.64 ± 146.40 AU/mL (mean ± SD). Our data shows that seropositivity, obesity, non-fat-related, and fat-related body composition parameters all had a significant effect on the level of IgG titer after a two-dose vaccination of ChAdOx1. However, only non-fat-related and fat-related body composition parameters had a significant effect on the IgG titer after booster vaccination. CONCLUSION COVID-19 infection before the first dose of vaccination is not related to IgG titer after booster administration. Body composition has a significant effect on the production of anti-SARS-CoV-2 IgG after booster vaccination in females.
Collapse
Affiliation(s)
- Marlena Golec
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
| | - Joanna Zembala-John
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
- Department of Medicine and Environmental Epidemiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
- Silesian Center for Heart Diseases, Zabrze, Poland
| | - Martyna Fronczek
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
- Department of Pharmacology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Adam Konka
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
| | - Aneta Bochenek
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
| | - Karolina Wystyrk
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
| | | | - Marzena Zalewska
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
- Department of Basic Medical Sciences, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Bytom, Poland
| | - Martyna Chrapiec
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
| | - Sławomir Kasperczyk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Zenon Brzoza
- Department of Internal Diseases, Allergology, Endocrinology and Gastroenterology, Institute of Medical Sciences, University of Opole, Opole, Poland
| | - Rafał J Bułdak
- Silesian Park of Medical Technology Kardio-Med Silesia, Zabrze, Poland
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Opole, Poland
| |
Collapse
|
55
|
Wang Y, Luo D, Liu J, Song Y, Jiang B, Jiang H. Low skeletal muscle mass index and all-cause mortality risk in adults: A systematic review and meta-analysis of prospective cohort studies. PLoS One 2023; 18:e0286745. [PMID: 37285331 DOI: 10.1371/journal.pone.0286745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/22/2023] [Indexed: 06/09/2023] Open
Abstract
OBJECTIVE The relationship between low skeletal muscle mass index (SMI) and all-cause mortality risk in the general adults remains unclear. Our study was conducted to examine and quantify the associations between low SMI and all-cause mortality risks. METHODS PubMed, Web of Science, and Cochrane Library for primary data sources and references to relevant publications retrieved until 1 April 2023. A random-effect model, subgroup analyses, meta-regression, sensitivity analysis, and publication bias were conducted using STATA 16.0. RESULTS Sixteen prospective studies were included in the meta-analysis of low SMI and the risk of all-cause mortality. A total of 11696 deaths were ascertained among 81358 participants during the 3 to 14.4 years follow-up. The pooled RR of all-cause mortality risk was 1.57 (95% CI, 1.25 to 1.96, P < 0.001) across the lowest to the normal muscle mass category. The results of meta-regression showed that BMI (P = 0.086) might be sources of heterogeneity between studies. Subgroup analysis showed that low SMI was significantly associated with an increased risk of all-cause mortality in studies with a body mass index (BMI) between 18.5 to 25 (1.34, 95% CI, 1.24-1.45, P<0.001), 25 to 30 (1.91, 95% CI, 1.16-3.15, P = 0.011), and over 30 (2.58, 95% CI, 1.20-5.54 P = 0.015). CONCLUSIONS Low SMI was significantly associated with the increased risk of all-cause mortality, and the risk of all-cause mortality associated with low SMI was higher in adults with a higher BMI. Low SMI Prevention and treatment might be significant for reducing mortality risk and promoting healthy longevity.
Collapse
Affiliation(s)
- Yahai Wang
- College of Arts and Physical Education, Nanchang Normal College of Applied Technology, Nanchang, Jiangxi, China
| | - Donglin Luo
- Faculty of Health Service, Naval Medical University, Shanghai, China
| | - Jiahao Liu
- Faculty of Health Service, Naval Medical University, Shanghai, China
| | - Yu Song
- College of Arts and Physical Education, Nanchang Normal College of Applied Technology, Nanchang, Jiangxi, China
| | - Binggang Jiang
- Faculty of Health Service, Naval Medical University, Shanghai, China
| | - Haichao Jiang
- Faculty of Health Service, Naval Medical University, Shanghai, China
| |
Collapse
|
56
|
Musci RV, Andrie KM, Walsh MA, Valenti ZJ, Linden MA, Afzali MF, Bork S, Campbell M, Johnson T, Kail TE, Martinez R, Nguyen T, Sanford J, Wist S, Murrell MD, McCord JM, Hybertson BM, Zhang Q, Javors MA, Santangelo KS, Hamilton KL. Phytochemical compound PB125 attenuates skeletal muscle mitochondrial dysfunction and impaired proteostasis in a model of musculoskeletal decline. J Physiol 2023; 601:2189-2216. [PMID: 35924591 PMCID: PMC9898472 DOI: 10.1113/jp282273] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 07/28/2022] [Indexed: 02/06/2023] Open
Abstract
Impaired mitochondrial function and disrupted proteostasis contribute to musculoskeletal dysfunction. However, few interventions simultaneously target these two drivers to prevent musculoskeletal decline. Nuclear factor erythroid 2-related factor 2 (Nrf2) activates a transcriptional programme promoting cytoprotection, metabolism, and proteostasis. We hypothesized daily treatment with a purported Nrf2 activator, PB125, in Hartley guinea pigs, a model of musculoskeletal decline, would attenuate the progression of skeletal muscle mitochondrial dysfunction and impaired proteostasis and preserve musculoskeletal function. We treated 2- and 5-month-old male and female Hartley guinea pigs for 3 and 10 months, respectively, with the phytochemical compound PB125. Longitudinal assessments of voluntary mobility were measured using Any-MazeTM open-field enclosure monitoring. Cumulative skeletal muscle protein synthesis rates were measured using deuterium oxide over the final 30 days of treatment. Mitochondrial oxygen consumption in soleus muscles was measured using high resolution respirometry. In both sexes, PB125 (1) increased electron transfer system capacity; (2) attenuated the disease/age-related decline in coupled and uncoupled mitochondrial respiration; and (3) attenuated declines in protein synthesis in the myofibrillar, mitochondrial and cytosolic subfractions of the soleus. These effects were not associated with statistically significant prolonged maintenance of voluntary mobility in guinea pigs. Collectively, treatment with PB125 contributed to maintenance of skeletal muscle mitochondrial respiration and proteostasis in a pre-clinical model of musculoskeletal decline. Further investigation is necessary to determine if these documented effects of PB125 are also accompanied by slowed progression of other aspects of musculoskeletal dysfunction. KEY POINTS: Aside from exercise, there are no effective interventions for musculoskeletal decline, which begins in the fifth decade of life and contributes to disability and cardiometabolic diseases. Targeting both mitochondrial dysfunction and impaired protein homeostasis (proteostasis), which contribute to the age and disease process, may mitigate the progressive decline in overall musculoskeletal function (e.g. gait, strength). A potential intervention to target disease drivers is to stimulate nuclear factor erythroid 2-related factor 2 (Nrf2) activation, which leads to the transcription of genes responsible for redox homeostasis, proteome maintenance and mitochondrial energetics. Here, we tested a purported phytochemical Nrf2 activator, PB125, to improve mitochondrial function and proteostasis in male and female Hartley guinea pigs, which are a model for musculoskeletal ageing. PB125 improved mitochondrial respiration and attenuated disease- and age-related declines in skeletal muscle protein synthesis, a component of proteostasis, in both male and female Hartley guinea pigs.
Collapse
Affiliation(s)
- Robert V. Musci
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Kendra M. Andrie
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Maureen A. Walsh
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Zackary J. Valenti
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Melissa A. Linden
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Maryam F. Afzali
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Sydney Bork
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Margaret Campbell
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Taylor Johnson
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Thomas E. Kail
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Richard Martinez
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Tessa Nguyen
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Joseph Sanford
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Sara Wist
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | | | - Joe M. McCord
- Pathways Bioscience, Aurora, CO
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Brooks M. Hybertson
- Pathways Bioscience, Aurora, CO
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Qian Zhang
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | | | - Kelly S. Santangelo
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Karyn L. Hamilton
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
57
|
Cui CY, Ferrucci L, Gorospe M. Macrophage Involvement in Aging-Associated Skeletal Muscle Regeneration. Cells 2023; 12:1214. [PMID: 37174614 PMCID: PMC10177543 DOI: 10.3390/cells12091214] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
The skeletal muscle is a dynamic organ composed of contractile muscle fibers, connective tissues, blood vessels and nerve endings. Its main function is to provide motility to the body, but it is also deeply involved in systemic metabolism and thermoregulation. The skeletal muscle frequently encounters microinjury or trauma, which is primarily repaired by the coordinated actions of muscle stem cells (satellite cells, SCs), fibro-adipogenic progenitors (FAPs), and multiple immune cells, particularly macrophages. During aging, however, the capacity of skeletal muscle to repair and regenerate declines, likely contributing to sarcopenia, an age-related condition defined as loss of muscle mass and function. Recent studies have shown that resident macrophages in skeletal muscle are highly heterogeneous, and their phenotypes shift during aging, which may exacerbate skeletal muscle deterioration and inefficient regeneration. In this review, we highlight recent insight into the heterogeneity and functional roles of macrophages in skeletal muscle regeneration, particularly as it declines with aging.
Collapse
Affiliation(s)
- Chang-Yi Cui
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
58
|
Di Credico A, Gaggi G, Izzicupo P, Vitucci D, Buono P, Di Baldassarre A, Ghinassi B. Betaine Treatment Prevents TNF-α-Mediated Muscle Atrophy by Restoring Total Protein Synthesis Rate and Morphology in Cultured Myotubes. J Histochem Cytochem 2023; 71:199-209. [PMID: 37013268 PMCID: PMC10149894 DOI: 10.1369/00221554231165326] [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] [Accepted: 01/12/2023] [Indexed: 04/05/2023] Open
Abstract
Skeletal muscle atrophy is represented by a dramatic decrease in muscle mass, and it is related to a lower life expectancy. Among the different causes, chronic inflammation and cancer promote protein loss through the effect of inflammatory cytokines, leading to muscle shrinkage. Thus, the availability of safe methods to counteract inflammation-derived atrophy is of high interest. Betaine is a methyl derivate of glycine and it is an important methyl group donor in transmethylation. Recently, some studies found that betaine could promote muscle growth, and it is also involved in anti-inflammatory mechanisms. Our hypothesis was that betaine would be able to prevent tumor necrosis factor-α (TNF-α)-mediated muscle atrophy in vitro. We treated differentiated C2C12 myotubes for 72 hr with either TNF-α, betaine, or a combination of them. After the treatment, we analyzed total protein synthesis, gene expression, and myotube morphology. Betaine treatment blunted the decrease in muscle protein synthesis rate exerted by TNF-α, and upregulated Mhy1 gene expression in both control and myotube treated with TNF-α. In addition, morphological analysis revealed that myotubes treated with both betaine and TNF-α did not show morphological features of TNF-α-mediated atrophy. We demonstrated that in vitro betaine supplementation counteracts the muscle atrophy led by inflammatory cytokines.
Collapse
Affiliation(s)
- Andrea Di Credico
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti, Italy
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology, Chieti, Italy
| | - Giulia Gaggi
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti, Italy
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology, Chieti, Italy
| | - Pascal Izzicupo
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Daniela Vitucci
- Department of Movement Sciences and Wellness, University Parthenope, Napoli, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Napoli, Italy
| | - Pasqualina Buono
- Department of Movement Sciences and Wellness, University Parthenope, Napoli, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Napoli, Italy
| | - Angela Di Baldassarre
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti, Italy
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology, Chieti, Italy
| | - Barbara Ghinassi
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, Chieti, Italy
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology, Chieti, Italy
| |
Collapse
|
59
|
Methenitis S, Nomikos T, Kontou E, Kiourelli KM, Papadimas G, Papadopoulos C, Terzis G. Skeletal muscle fiber composition may modify the effect of nutrition on body composition in young females. Nutr Metab Cardiovasc Dis 2023; 33:817-825. [PMID: 36725423 DOI: 10.1016/j.numecd.2022.12.027] [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: 09/29/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIM The aim of this study was to investigate the hypothesis that healthy, normal-weight females with greater proportions and sizes of the oxidative muscle fibers would also be characterized by a healthier body composition compared with individuals with increased glycolytic fibers, even if both follow similar nutritional plans. METHODS AND RESULTS Vastus lateralis muscle fiber-type composition, body composition through dual-energy X-ray absorptiometry, and dietary intakes through questionnaire were evaluated in twenty-two young, healthy, non-obese females (age: 21.3±1.8yrs, body mass: 67.5±6.2 kg, body height: 1.66±0.05m, body mass index (BMI): 24.2±2.6 kg m-2). The participants were allocated into two groups according to their type I muscle fibers percentage [high (HI) and low (LI)]. The participants of the LI group were characterized by significantly higher body mass, fat mass, BMI, and cross-sectional and percentage cross-sectional area (%CSA) of type IIx muscle fibers compared with participants of the HI group (p < 0.021). In contrast, the HI group was characterized by higher cross-sectional and %CSA of type I muscle fibers compared with the LI group (p < 0.038). Significant correlations were observed between body fat mass, lean body mass, total energy intake, fat energy intake, and %CSAs of type I and IIx muscle fibers (r: -0.505 to 0.685; p < 0.05). CONCLUSION In conclusion, this study suggests that muscle fiber composition is an important factor that at least partly could explain the observed differential inter-individual responses of the body composition to nutrition in female individuals. Increased %CSAs of type I muscle fibers seem to act as a protective mechanism against obesity and favor a healthier body composition, neutralizing the negative effect of increased caloric fats intake on body composition, probably because of their greater oxidative metabolic properties and fat utilization capacities. In contrast, female individuals with low type I and high type IIx %CSAs of type I seem to be more metabolically inflexible and dietinduced obesity prone, even if they consume fewer total daily calories and fats.
Collapse
Affiliation(s)
- Spyridon Methenitis
- Sports Performance Laboratory, School of Physical Education & Sports Science, National and Kapodistrian University of Athens, Greece.
| | - Tzortzis Nomikos
- Department of Nutrition and Dietetics, School of Health Sciences & Education, Harokopio University, Greece.
| | - Eleni Kontou
- Sports Performance Laboratory, School of Physical Education & Sports Science, National and Kapodistrian University of Athens, Greece; Theseus, Physical Medicine and Rehabilitation Center, Athens, Greece.
| | - Kleio-Maria Kiourelli
- Department of Nutrition and Dietetics, School of Health Sciences & Education, Harokopio University, Greece.
| | - George Papadimas
- A' Neurology Clinic, Aiginition Hospital, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Constantinos Papadopoulos
- A' Neurology Clinic, Aiginition Hospital, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Gerasimos Terzis
- Sports Performance Laboratory, School of Physical Education & Sports Science, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
60
|
Kajabadi N, Low M, Jacques E, Lad H, Tung LW, Babaeijandaghi F, Gamu D, Zelada D, Wong CK, Chang C, Yi L, Wosczyna MN, Rando TA, Henríquez JP, Gibson WT, Gilbert PM, Rossi FMV. Activation of β-catenin in mesenchymal progenitors leads to muscle mass loss. Dev Cell 2023; 58:489-505.e7. [PMID: 36898377 DOI: 10.1016/j.devcel.2023.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/12/2022] [Accepted: 02/10/2023] [Indexed: 03/11/2023]
Abstract
Loss of muscle mass is a common manifestation of chronic disease. We find the canonical Wnt pathway to be activated in mesenchymal progenitors (MPs) from cancer-induced cachectic mouse muscle. Next, we induce β-catenin transcriptional activity in murine MPs. As a result, we observe expansion of MPs in the absence of tissue damage, as well as rapid loss of muscle mass. Because MPs are present throughout the organism, we use spatially restricted CRE activation and show that the induction of tissue-resident MP activation is sufficient to induce muscle atrophy. We further identify increased expression of stromal NOGGIN and ACTIVIN-A as key drivers of atrophic processes in myofibers, and we verify their expression by MPs in cachectic muscle. Finally, we show that blocking ACTIVIN-A rescues the mass loss phenotype triggered by β-catenin activation in MPs, confirming its key functional role and strengthening the rationale for targeting this pathway in chronic disease.
Collapse
Affiliation(s)
- Nasim Kajabadi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Marcela Low
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Carrera de Química y Farmacia, Facultad de Medicina y Ciencia, Universidad San Sebastián, General Lagos 1163, 5090000 Valdivia, Chile
| | - Erik Jacques
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Heta Lad
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Lin Wei Tung
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Farshad Babaeijandaghi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Daniel Gamu
- BC Children's Hospital Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, C201, 4500 Oak Street, Vancouver, BC V6H 3N1, Canada
| | - Diego Zelada
- Neuromuscular Studies Laboratory (NeSt Lab), GDeP, Department of Cell Biology, Universidad de Concepción, Concepción, Chile
| | - Chi Kin Wong
- BC Children's Hospital Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, C201, 4500 Oak Street, Vancouver, BC V6H 3N1, Canada
| | - Chihkai Chang
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Lin Yi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Michael N Wosczyna
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Musculoskeletal Research Center, Bioengineering Institute, Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY 10010, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Juan Pablo Henríquez
- Neuromuscular Studies Laboratory (NeSt Lab), GDeP, Department of Cell Biology, Universidad de Concepción, Concepción, Chile
| | - William T Gibson
- BC Children's Hospital Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, C201, 4500 Oak Street, Vancouver, BC V6H 3N1, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Fabio M V Rossi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
61
|
Mancin L, Wu GD, Paoli A. Gut microbiota-bile acid-skeletal muscle axis. Trends Microbiol 2023; 31:254-269. [PMID: 36319506 DOI: 10.1016/j.tim.2022.10.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 01/13/2023]
Abstract
The gut microbiota represents a 'metabolic organ' that can regulate human metabolism. Intact gut microbiota contributes to host homeostasis, whereas compositional perturbations, termed dysbiosis, are associated with a wide range of diseases. Recent evidence demonstrates that dysbiosis, and the accompanying loss of microbiota-derived metabolites, results in a substantial alteration of skeletal muscle metabolism. As an example, bile acids, produced in the liver and further metabolized by intestinal microbiota, are of considerable interest since they regulate several host metabolic pathways by activating nuclear receptors, including the farnesoid X receptor (FXR). Indeed, alteration of gut microbiota may lead to skeletal muscle atrophy via a bile acid-FXR pathway. This Review aims to suggest a new pathway that connects different mechanisms, involving the gut-muscle axis, that are often seen as unrelated, and, starting from preclinical studies, we hypothesize new strategies aimed at optimizing skeletal muscle functionality.
Collapse
Affiliation(s)
- Laura Mancin
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Human Inspired Technology Research Center HIT, University of Padua, Padua, Italy.
| | - Gary D Wu
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Antonio Paoli
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Human Inspired Technology Research Center HIT, University of Padua, Padua, Italy; Research Center for High Performance Sport, UCAM, Catholic University of Murcia, Murcia, Spain
| |
Collapse
|
62
|
Dong J, Zhao J, Liu X, Lee WN. Nondestructive ultrasound evaluation of microstructure-related material parameters of skeletal muscle: An in silico and in vitro study. J Mech Behav Biomed Mater 2023; 142:105807. [PMID: 37030170 DOI: 10.1016/j.jmbbm.2023.105807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
Direct and nondestructive assessment of material properties of skeletal muscle in vivo shall advance our understanding of intact muscle mechanics and facilitate personalized interventions. However, this is challenged by intricate hierarchical microstructure of the skeletal muscle. We have previously regarded the skeletal muscle as a composite of myofibers and extracellular matrix (ECM), formulated shear wave propagation in the undeformed muscle using the acoustoelastic theory, and preliminarily demonstrated that ultrasound-based shear wave elastography (SWE) could estimate microstructure-related material parameters (MRMPs): myofiber stiffness μf, ECM stiffness μm, and myofiber volume ratio Vf. The proposed method warrants further validation but is hampered by the lack of ground truth values of MRMPs. In this study, we presented analytical and experimental validations of the proposed method using finite-element (FE) simulations and 3D-printed hydrogel phantoms, respectively. Three combinations of different physiologically relevant MRMPs were used in the FE simulations where shear wave propagations in the corresponding composite media were simulated. Two 3D-printed hydrogel phantoms with the MRMPs close to those of a real skeletal muscle (i.e., μf=2.02kPa, μm=52.42kPa, and Vf=0.675,0.832) for ultrasound imaging were fabricated by an alginate-based hydrogel printing protocol that we modified and optimized from the freeform reversible embedding of suspended hydrogels (FRESH) method in literature. Average percent errors of (μf,μm,Vf) estimates were found to be (2.7%,7.3%,2.4%)in silico and (3.0%,8.0%,9.9%)in vitro. This quantitative study corroborated the potential of our proposed theoretical model along with ultrasound SWE for uncovering microstructural characteristics of the skeletal muscle in an entirely nondestructive way.
Collapse
|
63
|
ELbialy ZI, Atef E, Al-Hawary II, Salah AS, Aboshosha AA, Abualreesh MH, Assar DH. Myostatin-mediated regulation of skeletal muscle damage post-acute Aeromonas hydrophila infection in Nile tilapia (Oreochromis niloticus L.). FISH PHYSIOLOGY AND BIOCHEMISTRY 2023; 49:1-17. [PMID: 36622623 DOI: 10.1007/s10695-022-01165-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
This study focuses on the relationship between myostatin (MyoS), myogenin (MyoG), and the growth hormone/insulin-like growth factor-1 (GH/IGF-1) axis for muscle growth and histopathological changes in muscle after an Aeromonas hydrophila infection. A total number of 90 Nile tilapia (55.85 g) were randomly allocated into two equal groups of three replicates each. The first group was an uninfected control group that was injected intraperitoneally (ip) with 0.2 ml phosphate buffer saline (PBS), while the second group was injected ip with 0.2 ml (1.3 × 108 CFU/ml) Aeromonas hydrophila culture suspension. Sections of white muscle and liver tissues were taken from each group 24 h, 48 h, 72 h, and 1 week after infection for molecular analysis and histopathological examination. The results revealed that with time progression, the severity of muscle lesions increased from edema between bundles and mononuclear inflammatory cell infiltration 24 h post-challenge to severe atrophy of muscle bundles with irregular and curved fibers with hyalinosis of the fibers 1 week postinfection. The molecular analysis showed that bacterial infection was able to induce the muscle expression levels of GH with reduced ILGF-1, MyoS, and MyoG at 24 h postinfection. However, time progression postinfection reversed these findings through elevated muscle expression levels of MyoS with regressed expression levels of muscle GH, ILGF-1, and MyoG. There have been no previous reports on the molecular expression analysis of the aforementioned genes and muscle histopathological changes in Nile tilapia following acute Aeromonas hydrophila infection. Our findings, collectively, revealed that the up-and down-regulation of the myostatin signaling is likely to be involved in the postinfection-induced muscle wasting through the negative regulation of genes involved in muscle growth, such as GH, ILGF-1, and myogenin, in response to acute Aeromonas hydrophila infection in Nile tilapia, Oreochromis niloticus.
Collapse
Affiliation(s)
- Zizy I ELbialy
- Fish Processing and Biotechnology Department, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt.
| | - Eman Atef
- Fish Processing and Biotechnology Department, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Ibrahim I Al-Hawary
- Fish Processing and Biotechnology Department, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Abdallah S Salah
- Department of Aquaculture, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
- Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA, UK
| | - Ali A Aboshosha
- Department of Genetics, Faculty of Agriculture, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Muyassar H Abualreesh
- Department of Marine Biology, Faculty of Marine Sciences, King Abdul-Aziz University (KAU), Jeddah, 21589, Saudi Arabia
| | - Doaa H Assar
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt.
| |
Collapse
|
64
|
Liu Z, Feng Y, Zhao H, Hu J, Chen Y, Liu D, Wang H, Zhu X, Yang H, Shen Z, Xia X, Ye J, Liu Y. Pharmacokinetics and tissue distribution of Ramulus Mori (Sangzhi) alkaloids in rats and its effects on liver enzyme activity. Front Pharmacol 2023; 14:1136772. [PMID: 36873997 PMCID: PMC9981942 DOI: 10.3389/fphar.2023.1136772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/07/2023] [Indexed: 02/19/2023] Open
Abstract
Ramulus Mori (Sangzhi) alkaloids (SZ-A) derived from twigs of mulberry (Morus alba L., genus Morus in the Moraceae family) was approved by the National Medical Products Administration in 2020 for the treatment of type 2 diabetes mellitus. In addition to excellent hypoglycemic effect, increasing evidence has confirmed that SZ-A exerts multiple pharmacological effects, such as protecting pancreatic ß-cell function, stimulating adiponectin expression, and alleviating hepatic steatosis. Importantly, a specific distribution of SZ-A in target tissues following oral absorption into the blood is essential for the induction of multiple pharmacological effects. However, there is a lack of studies thoroughly exploring the pharmacokinetic profiles and tissue distribution of SZ-A following oral absorption into the blood, particularly dose-linear pharmacokinetics and target tissue distribution associated with glycolipid metabolic diseases. In the present study, we systematically investigated the pharmacokinetics and tissue distribution of SZ-A and its metabolites in human and rat liver microsomes, and rat plasma, as well as its effects on the activity of hepatic cytochrome P450 enzymes (CYP450s). The results revealed that SZ-A was rapidly absorbed into the blood, exhibited linear pharmacokinetic characteristics in the dose range of 25-200 mg/kg, and was broadly distributed in glycolipid metabolism-related tissues. The highest SZ-A concentrations were observed in the kidney, liver, and aortic vessels, followed by the brown and subcutaneous adipose tissues, and the heart, spleen, lung, muscle, pancreas, and brain. Except for the trace oxidation products produced by fagomine, other phase I or phase II metabolites were not detected. SZ-A had no inhibitory or activating effects on major CYP450s. Conclusively, SZ-A is rapidly and widely distributed in target tissues, with good metabolic stability and a low risk of triggering drug-drug interactions. This study provides a framework for deciphering the material basis of the multiple pharmacological functions of SZ-A, its rational clinical use, and the expansion of its indications.
Collapse
Affiliation(s)
- Zhihua Liu
- Beijing Wehand-Bio Pharmaceutical Co, Ltd., Beijing, China
| | - Yu Feng
- Beijing Wehand-Bio Pharmaceutical Co, Ltd., Beijing, China
| | - Hang Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jinping Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yanmin Chen
- Beijing Wehand-Bio Pharmaceutical Co, Ltd., Beijing, China.,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Dongdong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hongliang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiangyang Zhu
- Beijing Wehand-Bio Pharmaceutical Co, Ltd., Beijing, China
| | - Hongzhen Yang
- Beijing Wehand-Bio Pharmaceutical Co, Ltd., Beijing, China
| | - Zhufang Shen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xuejun Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
65
|
Xiang S, Li Y, Li Y, Zhang J, Pan W, Lu Y, Liu S. Increased Dietary Niacin Intake Improves Muscle Strength, Quality, and Glucose Homeostasis in Adults over 40 Years of Age. J Nutr Health Aging 2023; 27:709-718. [PMID: 37754210 DOI: 10.1007/s12603-023-1967-0] [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: 05/12/2023] [Accepted: 07/17/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND AND AIMS Age-related loss of skeletal muscle mass and strength begins at 40 years of age, and limited evidence suggests that niacin supplementation increases levels of nicotinamide adenine dinucleotide in mouse muscle tissue. In addition, skeletal muscle has a key role in the body's processing of glucose. Therefore, this study aimed to investigate the relationship between dietary niacin and skeletal muscle mass, strength, and glucose homeostasis in people aged 40 years and older. METHODS This study was an American population-based cross-sectional analysis using data from the National Health and Nutrition Examination Survey (NHANES). Considering that some outcomes are only measured in specific survey cycles and subsamples, we established three data sets: a grip strength dataset (2011-2014, n=3772), a body mass components dataset (2011-2018, n=3279), and a glucose homeostasis dataset (1999-2018, n=9189). Dietary niacin and covariates were measured in all survey cycles. Linear regression or logistic regression models that adjusted for several main covariates, such as physical activity and diet, was used to evaluate the relationship between dietary niacin and grip strength, total lean mass, appendicular lean mass, total fat, trunk fat, total bone mineral content, homeostasis model assessment of insulin resistance (HOMA-IR), fasting blood glycose, fasting insulin and sarcopenia risk. Subgroup analyses, a trend test, an interaction test, and a restricted cubic spline were used for further exploration. RESULTS Higher dietary niacin intake was significantly correlated with higher grip strength (β 0.275, 95% confidence intervals [CI] 0.192-0.357), higher total lean mass (β 0.060, 95% CI 0.045-0.074), higher appendicular lean mass (β 0.025, 95% CI 0.018-0.033), and higher total bone mineral content (β 0.005, 95% CI 0.004-0.007). By contrast, higher dietary niacin intake was significantly associated with lower total fat (β -0.061, 95% CI -0.076 to -0.046), lower trunk fat (β -0.041, 95% CI -0.050 to -0.032) and lower sarcopenia risk (OR 0.460, 95% CI 0.233 to 0.907). In addition, dietary niacin significantly reduced HOMA-IR, fasting blood glucose (in participants without diabetes), and fasting insulin (p <0.05). CONCLUSION Niacin is associated with improved body composition (characterized by increased muscle mass and decreased fat content) and improved glucose homeostasis in dietary doses. Dietary niacin supplementation is a feasible way to alleviate age-related muscular loss.
Collapse
Affiliation(s)
- S Xiang
- Yun Lu, MD, PhD, Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China, , 0000-0003-2253-2983; Shang-Long Liu, MD, PhD, Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China, , 0000-0002-5828-4718
| | | | | | | | | | | | | |
Collapse
|
66
|
Methenitis S, Papadopoulou SK, Panayiotou G, Kaprara A, Hatzitolios A, Skepastianos P, Karali K, Feidantsis K. Nutrition, body composition and physical activity have differential impact on the determination of lipidemic blood profiles between young females with different blood cholesterol concentrations. Obes Res Clin Pract 2023; 17:25-33. [PMID: 36641266 DOI: 10.1016/j.orcp.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/01/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023]
Abstract
INTRODUCTION This cross-sectional study explored whether nutrition, body composition, and physical activity energy expenditure (PAΕΝ) have a differential impact on lipidemic blood profiles among young females with different blood cholesterol concentrations. METHODS One hundred thirty-five young female students (N = 135) were allocated into three groups according to their blood cholesterol concentrations (Chol): (A) Normal [NL; Chol: < 200 mg·dL-1; n = 56 Age: 21.4 ± 2.6 yrs, Body Mass Index (BMI): 22.1 ± 2.0 kg·m-2], (B) Borderline (BL; Chol: ≥200 mg·dL-1 and <240 mg·dL-1; n = 44 Age: 21.6 ± 2.5 yrs, BMI: 24.2 ± 3.1 kg·m-2) and (C) High level (HL; Chol: ≥240 mg·dL-1; n = 35 Age: 22.5 ± 2.4 yrs, BMI: 28.9 ± 2.1 kg·m-2). Body composition [bioelectrical impedance analysis including lean body mass (LBM) and body fat mass], nutritional intake (recall questionnaire), daily physical activity energy expenditure through activity trackers and resting blood lipids concentrations were evaluated. RESULTS Multiple linear regression analyses revealed that in the NL group, lean mass, daily PAΕΝ and daily energy balance were the determinant parameters of blood lipidemic profiles (B: -0.815 to 0.700). In the BL group, nutrition, body composition and daily physical activity energy expenditure exhibited similar impacts (B: -0.440 to 0.478). In the HL group, nutritional intake and body fat mass determined blood lipidemic profile (B: -0.740 to 0.725). CONCLUSION Nutrition, body composition and daily PAΕΝ impact on blood lipids concentration is not universal among young females. In NL females, PAEN, energy expenditure and LBM are the strongest determinants of blood lipids, while in HL females, nutritional intake and body fat mass are. As PAΕΝ increases, the importance of nutrition and body fat decreases, and vice versa.
Collapse
Affiliation(s)
- Spyridon Methenitis
- Sports Performance Laboratory, School of Physical Education & Sports Science, National and Kapodistrian University of Athens, Athens GR-17237, Greece; Department of Nutrition Sciences and Dietetics, Faculty of Health Sciences, International Hellenic University, P.O. Box 141, Sindos GR-57400, Thessaloniki, Greece; Theseus, Physical Medicine and Rehabilitation Center, Athens, Greece
| | - Sousana K Papadopoulou
- Department of Nutrition Sciences and Dietetics, Faculty of Health Sciences, International Hellenic University, P.O. Box 141, Sindos GR-57400, Thessaloniki, Greece
| | - George Panayiotou
- Laboratory of Exercise, Health and Human Performance, Applied Sport Science Postgraduate Program, Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Athina Kaprara
- Laboratory of Sports Medicine, School of Physical Education and Sports Science, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - Apostolos Hatzitolios
- 1st Department of Cardiology, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - Petros Skepastianos
- Department of Biomedical Sciences, Faculty of Health Sciences, International Hellenic University, P.O. Box 141, Sindos GR-57400, Thessaloniki, Greece
| | - Konstantina Karali
- 1st Department of Cardiology, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - Konstantinos Feidantsis
- Department of Nutrition Sciences and Dietetics, Faculty of Health Sciences, International Hellenic University, P.O. Box 141, Sindos GR-57400, Thessaloniki, Greece.
| |
Collapse
|
67
|
Fujimaki S, Ono Y. Murine Models of Tenotomy-Induced Mechanical Overloading and Tail-Suspension-Induced Mechanical Unloading. Methods Mol Biol 2023; 2640:207-215. [PMID: 36995597 DOI: 10.1007/978-1-0716-3036-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Skeletal muscle is a highly plastic tissue that can alter its mass and strength in response to mechanical stimulation, such as overloading and unloading, which lead to muscle hypertrophy and atrophy, respectively. Mechanical loading in the muscle influences muscle stem cell dynamics, including activation, proliferation, and differentiation. Although experimental models of mechanical overloading and unloading have been widely used for the investigation of the molecular mechanisms regulating muscle plasticity and stem cell function, few studies have described the methods in detail. Here, we describe the appropriate procedures for tenotomy-induced mechanical overloading and tail-suspension-induced mechanical unloading, which are the most common and simple methods to induce muscle hypertrophy and atrophy in mouse models.
Collapse
Affiliation(s)
- Shin Fujimaki
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
68
|
Li C, Li N, Zhang Z, Song Y, Li J, Wang Z, Bo H, Zhang Y. The specific mitochondrial unfolded protein response in fast- and slow-twitch muscles of high-fat diet-induced insulin-resistant rats. Front Endocrinol (Lausanne) 2023; 14:1127524. [PMID: 37008907 PMCID: PMC10061072 DOI: 10.3389/fendo.2023.1127524] [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: 12/19/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
INTRODUCTION Skeletal muscle insulin resistance (IR) plays an important role in the pathogenesis of type 2 diabetes mellitus. Skeletal muscle is a heterogeneous tissue composed of different muscle fiber types that contribute distinctly to IR development. Glucose transport shows more protection in slow-twitch muscles than in fast-twitch muscles during IR development, while the mechanisms involved remain unclear. Therefore, we investigated the role of the mitochondrial unfolded protein response (UPRmt) in the distinct resistance of two types of muscle in IR. METHODS Male Wistar rats were divided into high-fat diet (HFD) feeding and control groups. We measured glucose transport, mitochondrial respiration, UPRmt and histone methylation modification of UPRmt-related proteins to examine the UPRmt in the slow fiber-enriched soleus (Sol) and fast fiber-enriched tibialis anterior (TA) under HFD conditions. RESULTS Our results indicate that 18 weeks of HFD can cause systemic IR, while the disturbance of Glut4-dependent glucose transport only occurred in fast-twitch muscle. The expression levels of UPRmt markers, including ATF5, HSP60 and ClpP, and the UPRmt-related mitokine MOTS-c were significantly higher in slow-twitch muscle than in fast-twitch muscle under HFD conditions. Mitochondrial respiratory function is maintained only in slow-twitch muscle. Additionally, in the Sol, histone methylation at the ATF5 promoter region was significantly higher than that in the TA after HFD feeding. CONCLUSION The expression of proteins involved in glucose transport in slow-twitch muscle remains almost unaltered after HFD intervention, whereas a significant decline of these proteins was observed in fast-twitch muscle. Specific activation of the UPRmt in slow-twitch muscle, accompanied by higher mitochondrial respiratory function and MOTS-c expression, may contribute to the higher resistance to HFD in slow-twitch muscle. Notably, the different histone modifications of UPRmt regulators may underlie the specific activation of the UPRmt in different muscle types. However, future work applying genetic or pharmacological approaches should further uncover the relationship between the UPRmt and insulin resistance.
Collapse
Affiliation(s)
- Can Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Nan Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Ziyi Zhang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Yu Song
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Jialin Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Zhe Wang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Hai Bo
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin, China
- Department of Military Training Medicines, Logistics University of Chinese People’s Armed Police Force, Tianjin, China
- *Correspondence: Hai Bo, ; Yong Zhang,
| | - Yong Zhang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin, China
- *Correspondence: Hai Bo, ; Yong Zhang,
| |
Collapse
|
69
|
Oikawa S, Akimoto T. Functional Analysis of MicroRNAs in Skeletal Muscle. Methods Mol Biol 2023; 2640:339-349. [PMID: 36995606 DOI: 10.1007/978-1-0716-3036-5_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that are highly conserved in vertebrates and play important roles in diverse biological processes. miRNAs function to fine-tune gene expression by accelerating the degradation of mRNA and/or by inhibiting protein translation. Identification of muscle-specific miRNAs has extended our knowledge of the molecular network in skeletal muscle. Here we describe methods that are commonly used to analyze the function of miRNAs in skeletal muscle.
Collapse
Affiliation(s)
- Satoshi Oikawa
- Laboratory of Muscle Biology, Faculty of Sport Sciences, Waseda University, Saitama, Japan
| | - Takayuki Akimoto
- Laboratory of Muscle Biology, Faculty of Sport Sciences, Waseda University, Saitama, Japan.
| |
Collapse
|
70
|
Han X, Goh KY, Lee WX, Choy SM, Tang HW. The Importance of mTORC1-Autophagy Axis for Skeletal Muscle Diseases. Int J Mol Sci 2022; 24:297. [PMID: 36613741 PMCID: PMC9820406 DOI: 10.3390/ijms24010297] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) complex 1, mTORC1, integrates nutrient and growth factor signals with cellular responses and plays critical roles in regulating cell growth, proliferation, and lifespan. mTORC1 signaling has been reported as a central regulator of autophagy by modulating almost all aspects of the autophagic process, including initiation, expansion, and termination. An increasing number of studies suggest that mTORC1 and autophagy are critical for the physiological function of skeletal muscle and are involved in diverse muscle diseases. Here, we review recent insights into the essential roles of mTORC1 and autophagy in skeletal muscles and their implications in human muscle diseases. Multiple inhibitors targeting mTORC1 or autophagy have already been clinically approved, while others are under development. These chemical modulators that target the mTORC1/autophagy pathways represent promising potentials to cure muscle diseases.
Collapse
Affiliation(s)
- Xujun Han
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Kah Yong Goh
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Wen Xing Lee
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Sze Mun Choy
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Hong-Wen Tang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Division of Cellular & Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore
| |
Collapse
|
71
|
Structural functionality of skeletal muscle mitochondria and its correlation with metabolic diseases. Clin Sci (Lond) 2022; 136:1851-1871. [PMID: 36545931 DOI: 10.1042/cs20220636] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022]
Abstract
The skeletal muscle is one of the largest organs in the mammalian body. Its remarkable ability to swiftly shift its substrate selection allows other organs like the brain to choose their preferred substrate first. Healthy skeletal muscle has a high level of metabolic flexibility, which is reduced in several metabolic diseases, including obesity and Type 2 diabetes (T2D). Skeletal muscle health is highly dependent on optimally functioning mitochondria that exist in a highly integrated network with the sarcoplasmic reticulum and sarcolemma. The three major mitochondrial processes: biogenesis, dynamics, and mitophagy, taken together, determine the quality of the mitochondrial network in the muscle. Since muscle health is primarily dependent on mitochondrial status, the mitochondrial processes are very tightly regulated in the skeletal muscle via transcription factors like peroxisome proliferator-activated receptor-γ coactivator-1α, peroxisome proliferator-activated receptors, estrogen-related receptors, nuclear respiratory factor, and Transcription factor A, mitochondrial. Physiological stimuli that enhance muscle energy expenditure, like cold and exercise, also promote a healthy mitochondrial phenotype and muscle health. In contrast, conditions like metabolic disorders, muscle dystrophies, and aging impair the mitochondrial phenotype, which is associated with poor muscle health. Further, exercise training is known to improve muscle health in aged individuals or during the early stages of metabolic disorders. This might suggest that conditions enhancing mitochondrial health can promote muscle health. Therefore, in this review, we take a critical overview of current knowledge about skeletal muscle mitochondria and the regulation of their quality. Also, we have discussed the molecular derailments that happen during various pathophysiological conditions and whether it is an effect or a cause.
Collapse
|
72
|
Yan Y, Li M, Lin J, Ji Y, Wang K, Yan D, Shen Y, Wang W, Huang Z, Jiang H, Sun H, Qi L. Adenosine monophosphate activated protein kinase contributes to skeletal muscle health through the control of mitochondrial function. Front Pharmacol 2022; 13:947387. [PMID: 36339617 PMCID: PMC9632297 DOI: 10.3389/fphar.2022.947387] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/06/2022] [Indexed: 11/26/2022] Open
Abstract
Skeletal muscle is one of the largest organs in the body and the largest protein repository. Mitochondria are the main energy-producing organelles in cells and play an important role in skeletal muscle health and function. They participate in several biological processes related to skeletal muscle metabolism, growth, and regeneration. Adenosine monophosphate-activated protein kinase (AMPK) is a metabolic sensor and regulator of systemic energy balance. AMPK is involved in the control of energy metabolism by regulating many downstream targets. In this review, we propose that AMPK directly controls several facets of mitochondrial function, which in turn controls skeletal muscle metabolism and health. This review is divided into four parts. First, we summarize the properties of AMPK signal transduction and its upstream activators. Second, we discuss the role of mitochondria in myogenesis, muscle atrophy, regeneration post-injury of skeletal muscle cells. Third, we elaborate the effects of AMPK on mitochondrial biogenesis, fusion, fission and mitochondrial autophagy, and discuss how AMPK regulates the metabolism of skeletal muscle by regulating mitochondrial function. Finally, we discuss the effects of AMPK activators on muscle disease status. This review thus represents a foundation for understanding this biological process of mitochondrial dynamics regulated by AMPK in the metabolism of skeletal muscle. A better understanding of the role of AMPK on mitochondrial dynamic is essential to improve mitochondrial function, and hence promote skeletal muscle health and function.
Collapse
Affiliation(s)
- Yan Yan
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Ming Li
- Department of Laboratory Medicine, Binhai County People’s Hospital Affiliated to Kangda College of Nanjing Medical University, Yancheng, China
| | - Jie Lin
- Department of Infectious Disease, Affiliated Hospital of Nantong University, Nantong, China
| | - Yanan Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Kexin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Dajun Yan
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Wei Wang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Department of Pathology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Zhongwei Huang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Haiyan Jiang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Haiyan Jiang, ; Hualin Sun, ; Lei Qi,
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
- *Correspondence: Haiyan Jiang, ; Hualin Sun, ; Lei Qi,
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Haiyan Jiang, ; Hualin Sun, ; Lei Qi,
| |
Collapse
|
73
|
Nomikos T, Methenitis S, Panagiotakos DB. The emerging role of skeletal muscle as a modulator of lipid profile the role of exercise and nutrition. Lipids Health Dis 2022; 21:81. [PMID: 36042487 PMCID: PMC9425975 DOI: 10.1186/s12944-022-01692-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 08/16/2022] [Indexed: 11/10/2022] Open
Abstract
The present article aims to discuss the hypothesis that skeletal muscle per se but mostly its muscle fiber composition could be significant determinants of lipid metabolism and that certain exercise modalities may improve metabolic dyslipidemia by favorably affecting skeletal muscle mass, fiber composition and functionality. It discusses the mediating role of nutrition, highlights the lack of knowledge on mechanistic aspects of this relationship and proposes possible experimental directions in this field.
Collapse
Affiliation(s)
- Tzortzis Nomikos
- Department of Nutrition and Dietetics, School of Health Sciences & Education, Harokopio University, Athens, Greece.
| | - Spyridon Methenitis
- Sports Performance Laboratory, School of Physical Education and Sports. Science, National and Kapodistrian University of Athens, Athens, Greece.,Theseus, Physical Medicine and Rehabilitation Center, Athens, Greece
| | - Demosthenes B Panagiotakos
- Department of Nutrition and Dietetics, School of Health Sciences & Education, Harokopio University, Athens, Greece
| |
Collapse
|
74
|
Qi Z, Xia J, Xue X, Liu W, Huang Z, Zhang X, Zou Y, Liu J, Liu J, Li X, Cao L, Li L, Cui Z, Ji B, Zhang Q, Ding S, Liu W. Codon-optimized FAM132b gene therapy prevents dietary obesity by blockading adrenergic response and insulin action. Int J Obes (Lond) 2022; 46:1970-1982. [PMID: 35922561 DOI: 10.1038/s41366-022-01189-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND FAM132b (myonectin) has been identified as a muscle-derived myokine with exercise and has hormone activity in circulation to regulate iron homeostasis and lipid metabolism via unknown receptors. Here, we aim to explore the potential of adeno-associated virus to deliver FAM132b in vivo to develop a gene therapy against obesity. METHODS Adeno-associated virus AAV9 were engineered to induce overexpression of FAM132b with two mutations, A136T and P159A. Then, AAV9 was delivered into high-fat diet mice through tail vein, and glucose homeostasis and obesity development of mice were observed. Methods of structural biology were used to predict the action site or receptor of the FAM132b mutant. RESULTS Treatment of high-fat diet-fed mice with AAV9 improved glucose intolerance and insulin resistance, and resulted in reductions in body weight, fat depot, and adipocyte size. Codon-optimized FAM132b (coFAM132b) reduced the glycemic response to epinephrine (EPI) in the whole body and increased the lipolytic response to EPI in adipose tissues. However, FAM132b knockdown by shRNA significantly increased the glycemic response to EPI in vivo and reduced adipocyte response to EPI and adipose tissue browning. Structural analysis predicted that the FAM132b mutant with A136T and P159A may form a weak bond with β2 adrenergic receptor (ADRB2) and may have more affinity for insulin and insulin-receptor complexes. CONCLUSIONS Our study underscores the potential of FAM132b gene therapy with codon optimization to treat obesity by modulating the adrenergic response and insulin action. Both structural biological analysis and in vivo experiments suggest that the adrenergic response and insulin action are most likely blockaded by FAM132b mutants.
Collapse
Affiliation(s)
- Zhengtang Qi
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Jie Xia
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Xiangli Xue
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Wenbin Liu
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Zhuochun Huang
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Xue Zhang
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Yong Zou
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Jianchao Liu
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Jiatong Liu
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Xingtian Li
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Lu Cao
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Lingxia Li
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Zhiming Cui
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Benlong Ji
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Qiang Zhang
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China.,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| | - Shuzhe Ding
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China. .,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China.
| | - Weina Liu
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, 200241, China. .,School of Physical Education and Health, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
75
|
Lin H, Ma X, Sun Y, Peng H, Wang Y, Thomas SS, Hu Z. Decoding the transcriptome of denervated muscle at single-nucleus resolution. J Cachexia Sarcopenia Muscle 2022; 13:2102-2117. [PMID: 35726356 PMCID: PMC9398230 DOI: 10.1002/jcsm.13023] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 02/18/2022] [Accepted: 05/09/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Skeletal muscle exhibits remarkable plasticity under both physiological and pathological conditions. One major manifestation of this plasticity is muscle atrophy that is an adaptive response to catabolic stimuli. Because the heterogeneous transcriptome responses to catabolism in different types of muscle cells are not fully characterized, we applied single-nucleus RNA sequencing (snRNA-seq) to unveil muscle atrophy related transcriptional changes at single nucleus resolution. METHODS Using a sciatic denervation mouse model of muscle atrophy, snRNA-seq was performed to generate single-nucleus transcriptional profiles of the gastrocnemius muscle from normal and denervated mice. Various bioinformatics analyses, including unsupervised clustering, functional enrichment analysis, trajectory analysis, regulon inference, metabolic signature characterization and cell-cell communication prediction, were applied to illustrate the transcriptome changes of the individual cell types. RESULTS A total of 29 539 muscle nuclei (normal vs. denervation: 15 739 vs. 13 800) were classified into 13 nuclear types according to the known cell markers. Among these, the type IIb myonuclei were further divided into two subgroups, which we designated as type IIb1 and type IIb2 myonuclei. In response to denervation, the proportion of type IIb2 myonuclei increased sharply (78.12% vs. 38.45%, P < 0.05). Concomitantly, trajectory analysis revealed that denervated type IIb2 myonuclei clearly deviated away from the normal type IIb2 myonuclei, indicating that this subgroup underwent robust transcriptional reprogramming upon denervation. Signature genes in denervated type IIb2 myonuclei included Runx1, Gadd45a, Igfn1, Robo2, Dlg2, and Sh3d19 (P < 0.001). The gene regulatory network analysis captured a group of atrophy-related regulons (Foxo3, Runx1, Elk4, and Bhlhe40) whose activities were enhanced (P < 0.01), especially in the type IIb2 myonuclei. The metabolic landscape in the myonuclei showed that most of the metabolic pathways were down-regulated by denervation (P < 0.001), while some of the metabolic signalling, such as glutathione metabolism, was specifically activated in the denervated type IIb2 myonulei. We also investigated the transcriptomic alterations in the type I myofibres, muscle stem cells, fibro-adipogenic progenitors, macrophages, endothelial cells and pericytes and characterized their signature responses to denervation. By predicting the cell-cell interactions, we observed that the communications between myofibres and muscle resident cells were diminished by denervation. CONCLUSIONS Our results define the myonuclear transition, metabolic remodelling, and gene regulation networks reprogramming associated with denervation-induced muscle atrophy and illustrate the molecular basis of the heterogeneity and plasticity of muscle cells in response to catabolism. These results provide a useful resource for exploring the molecular mechanism of muscle atrophy.
Collapse
Affiliation(s)
- Hongchun Lin
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Nephrology Division, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xinxin Ma
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Yuxiang Sun
- Nephrology Division, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hui Peng
- Nephrology Division, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanlin Wang
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Sandhya Sara Thomas
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Zhaoyong Hu
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
76
|
Cardoso TF, Bruscadin JJ, Afonso J, Petrini J, Andrade BGN, de Oliveira PSN, Malheiros JM, Rocha MIP, Zerlotini A, Ferraz JBS, Mourão GB, Coutinho LL, Regitano LCA. EEF1A1 transcription cofactor gene polymorphism is associated with muscle gene expression and residual feed intake in Nelore cattle. Mamm Genome 2022; 33:619-628. [PMID: 35816191 DOI: 10.1007/s00335-022-09959-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 06/22/2022] [Indexed: 12/01/2022]
Abstract
Cis-acting effects of noncoding variants on gene expression and regulatory molecules constitute a significant factor for phenotypic variation in complex traits. To provide new insights into the impacts of single-nucleotide polymorphisms (SNPs) on transcription factors (TFs) and transcription cofactors (TcoF) coding genes, we carried out a multi-omic analysis to identify cis-regulatory effects of SNPs on these genes' expression in muscle and describe their association with feed efficiency-related traits in Nelore cattle. As a result, we identified one SNP, the rs137256008C > T, predicted to impact the EEF1A1 gene expression (β = 3.02; P-value = 3.51E-03) and the residual feed intake trait (β = - 3.47; P-value = 0.02). This SNP was predicted to modify transcription factor sites and overlaps with several QTL for feed efficiency traits. In addition, co-expression network analyses showed that animals containing the T allele of the rs137256008 SNP may be triggering changes in the gene network. Therefore, our analyses reinforce and contribute to a better understanding of the biological mechanisms underlying gene expression control of feed efficiency traits in bovines. The cis-regulatory SNP can be used as biomarker for feed efficiency in Nelore cattle.
Collapse
Affiliation(s)
- T F Cardoso
- Embrapa Southeast Livestock, São Carlos, SP, Brazil
| | - J J Bruscadin
- Program on Evolutionary Genetics and Molecular Biology, Federal University of São Carlos, São Carlos, SP, Brazil
| | - J Afonso
- Embrapa Southeast Livestock, São Carlos, SP, Brazil
| | - J Petrini
- Department of Animal Science, "Luiz de Queiroz" College of Agriculture, University of São Paulo/ESALQ, Piracicaba, SP, Brazil
| | - B G N Andrade
- Computer Science Department, Munster Technological University, MTU/ADAPT, Cork, Ireland
| | - P S N de Oliveira
- Program on Evolutionary Genetics and Molecular Biology, Federal University of São Carlos, São Carlos, SP, Brazil
| | - J M Malheiros
- Federal University of Latin American Integration, Foz do Iguaçu, Paraná, Brazil
| | - M I P Rocha
- Program on Evolutionary Genetics and Molecular Biology, Federal University of São Carlos, São Carlos, SP, Brazil
| | - A Zerlotini
- Embrapa Agricultural Informatics, Campinas, SP, Brazil
| | - J B S Ferraz
- Department of Veterinary Medicine, University of São Paulo/FZEA, Pirassununga, Brazil
| | - G B Mourão
- Department of Animal Science, "Luiz de Queiroz" College of Agriculture, University of São Paulo/ESALQ, Piracicaba, SP, Brazil
| | - L L Coutinho
- Department of Animal Science, "Luiz de Queiroz" College of Agriculture, University of São Paulo/ESALQ, Piracicaba, SP, Brazil
| | | |
Collapse
|
77
|
Chen J, Li Z, Zhang Y, Zhang X, Zhang S, Liu Z, Yuan H, Pang X, Liu Y, Tao W, Chen X, Zhang P, Chen GQ. Mechanism of reduced muscle atrophy via ketone body (D)-3-hydroxybutyrate. Cell Biosci 2022; 12:94. [PMID: 35725651 PMCID: PMC9208164 DOI: 10.1186/s13578-022-00826-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/03/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Muscle atrophy is an increasingly global health problem affecting millions, there is a lack of clinical drugs or effective therapy. Excessive loss of muscle mass is the typical characteristic of muscle atrophy, manifesting as muscle weakness accompanied by impaired metabolism of protein and nucleotide. (D)-3-hydroxybutyrate (3HB), one of the main components of the ketone body, has been reported to be effective for the obvious hemodynamic effects in atrophic cardiomyocytes and exerts beneficial metabolic reprogramming effects in healthy muscle. This study aims to exploit how the 3HB exerts therapeutic effects for treating muscle atrophy induced by hindlimb unloaded mice. RESULTS Anabolism/catabolism balance of muscle protein was maintained with 3HB via the Akt/FoxO3a and the mTOR/4E-BP1 pathways; protein homeostasis of 3HB regulation includes pathways of ubiquitin-proteasomal, autophagic-lysosomal, responses of unfolded-proteins, heat shock and anti-oxidation. Metabolomic analysis revealed the effect of 3HB decreased purine degradation and reduced the uric acid in atrophied muscles; enhanced utilization from glutamine to glutamate also provides evidence for the promotion of 3HB during the synthesis of proteins and nucleotides. CONCLUSIONS 3HB significantly inhibits the loss of muscle weights, myofiber sizes and myofiber diameters in hindlimb unloaded mouse model; it facilitates positive balance of proteins and nucleotides with enhanced accumulation of glutamate and decreased uric acid in wasting muscles, revealing effectiveness for treating muscle atrophy.
Collapse
Affiliation(s)
- Jin Chen
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zihua Li
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yudian Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xu Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Shujie Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zonghan Liu
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Huimei Yuan
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Xiangsheng Pang
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yaxuan Liu
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Wuchen Tao
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Xiaoping Chen
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing, 100094, China.
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Peng Zhang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Guo-Qiang Chen
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China.
- MOE Key Lab of Industrial Biocatalysis, Dept of Chemical Engineering, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
78
|
Dlamini SN, Norris SA, Mendham AE, Mtintsilana A, Ward KA, Olsson T, Goedecke JH, Micklesfield LK. Targeted proteomics of appendicular skeletal muscle mass and handgrip strength in black South Africans: a cross-sectional study. Sci Rep 2022; 12:9512. [PMID: 35680977 PMCID: PMC9178538 DOI: 10.1038/s41598-022-13548-9] [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: 02/13/2022] [Accepted: 05/25/2022] [Indexed: 11/20/2022] Open
Abstract
Although appendicular skeletal muscle mass (ASM) and handgrip strength (HGS) are key components of sarcopenia, their underlying biological mechanisms remain poorly understood. We aimed to investigate associations of circulating biomarkers with ASM and HGS in middle-aged black South Africans. This study consisted of 934 black South Africans (469 men and 465 women, aged 41-72 years) from the Middle-aged Soweto cohort. Linear regression models were used to examine relationships between 182 biomarkers (measured with proximity extension assay) and dual-energy X-ray absorptiometry-measured ASM and dynamometer-measured HGS. Age, height, sex, smoking, alcohol, food insecurity, physical activity, visceral adipose tissue, HIV and menopausal status were included as confounders. Regression models showing sex-interactions were stratified by sex. The Benjamini-Hochberg false discovery rate (FDR) was used to control for multiple testing, and FDR-adjusted P values were reported. In the total sample, 10 biomarkers were associated with higher ASM and 29 with lower ASM (P < 0.05). Out of these 39 biomarkers, 8 were also associated with lower HGS (P < 0.05). MMP-7 was associated with lower HGS only (P = 0.011) in the total sample. Sex-interactions (P < 0.05) were identified for 52 biomarkers for ASM, and 6 for HGS. For men, LEP, MEPE and SCF were associated with higher ASM (P < 0.001, = 0.004, = 0.006, respectively), and MEPE and SCF were also associated with higher HGS (P = 0.001, 0.012, respectively). Also in men, 37 biomarkers were associated with lower ASM (P < 0.05), with none of these being associated with lower HGS. Furthermore, DLK-1 and MYOGLOBIN were associated with higher HGS only (P = 0.004, 0.006, respectively), while GAL-9 was associated with lower HGS only (P = 0.005), among men. For women, LEP, CD163, IL6, TNF-R1 and TNF-R2 were associated with higher ASM (P < 0.001, = 0.014, = 0.027, = 0.014, = 0.048, respectively), while IGFBP-2, CTRC and RAGE were associated with lower ASM (P = 0.043, 0.001, 0.014, respectively). No biomarker was associated with HGS in women. In conclusion, most biomarkers were associated with ASM and not HGS, and the associations of biomarkers with ASM and HGS displayed sex-specificity in middle-aged black South Africans. Proteomic studies should examine ASM and HGS individually. Future research should also consider sexual dimorphism in the pathophysiology of sarcopenia for development of sex-specific treatment and diagnostic methods.
Collapse
Affiliation(s)
- Siphiwe N Dlamini
- SAMRC/Wits Developmental Pathways for Health Research Unit, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Shane A Norris
- SAMRC/Wits Developmental Pathways for Health Research Unit, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Global Health Research Institute, School of Health and Human Development, University of Southampton, Southampton, UK
| | - Amy E Mendham
- SAMRC/Wits Developmental Pathways for Health Research Unit, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Health Through Physical Activity, Lifestyle and Sport Research Centre, FIMS International Collaborating Centre of Sports Medicine, Division of Physiological Sciences, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Asanda Mtintsilana
- SAMRC/Wits Developmental Pathways for Health Research Unit, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Kate A Ward
- SAMRC/Wits Developmental Pathways for Health Research Unit, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Medical Research Council Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Tommy Olsson
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| | - Julia H Goedecke
- SAMRC/Wits Developmental Pathways for Health Research Unit, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Lisa K Micklesfield
- SAMRC/Wits Developmental Pathways for Health Research Unit, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
79
|
Riparini G, Simone JM, Sartorelli V. FACS-isolation and Culture of Fibro-Adipogenic Progenitors and Muscle Stem Cells from Unperturbed and Injured Mouse Skeletal Muscle. J Vis Exp 2022:10.3791/63983. [PMID: 35758697 PMCID: PMC11077435 DOI: 10.3791/63983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Fibro-adipogenic progenitor cells (FAPs) are a population of skeletal muscle-resident mesenchymal stromal cells (MSCs) capable of differentiating along fibrogenic, adipogenic, osteogenic, or chondrogenic lineage. Together with muscle stem cells (MuSCs), FAPs play a critical role in muscle homeostasis, repair, and regeneration, while actively maintaining and remodeling the extracellular matrix (ECM). In pathological conditions, such as chronic damage and muscular dystrophies, FAPs undergo aberrant activation and differentiate into collagen-producing fibroblasts and adipocytes, leading to fibrosis and intramuscular fatty infiltration. Thus, FAPs play a dual role in muscle regeneration, either by sustaining MuSC turnover and promoting tissue repair or contributing to fibrotic scar formation and ectopic fat infiltrates, which compromise the integrity and function of the skeletal muscle tissue. A proper purification of FAPs and MuSCs is a prerequisite for understanding the biological role of these cells in physiological as well as in pathological conditions. Here, we describe a standardized method for the simultaneous isolation of FAPs and MuSCs from limb muscles of adult mice using fluorescence-activated cell sorting (FACS). The protocol describes in detail the mechanical and enzymatic dissociation of mononucleated cells from whole limb muscles and injured tibialis anterior (TA) muscles. FAPs and MuSCs are subsequently isolated using a semi-automated cell sorter to obtain pure cell populations. We additionally describe an optimized method for culturing quiescent and activated FAPs and MuSCs, either alone or in coculture conditions.
Collapse
Affiliation(s)
- Giulia Riparini
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis, Musculoskeletal, and Skin Diseases (NIAMS), National Institutes of Health (NIH);
| | - James M Simone
- Flow Cytometry Section, National Institute of Arthritis, Musculoskeletal, and Skin Diseases (NIAMS), National Institutes of Health (NIH)
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis, Musculoskeletal, and Skin Diseases (NIAMS), National Institutes of Health (NIH);
| |
Collapse
|
80
|
Cirillo F, Zimmers TA, Mangiavini L. Editorial: Metabolic Adaptation of Muscle Tissue in Diseases Associated With Cachexia. Front Cell Dev Biol 2022; 10:947902. [PMID: 35721487 PMCID: PMC9204193 DOI: 10.3389/fcell.2022.947902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Federica Cirillo
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, Italy
- *Correspondence: Federica Cirillo,
| | - Teresa A. Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, United States
- Indiana Center of Musculoskeletal Health, Indianapolis, IN, United States
- Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States
| | - Laura Mangiavini
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|
81
|
Forouhan M, Lim WF, Zanetti-Domingues LC, Tynan CJ, Roberts TC, Malik B, Manzano R, Speciale AA, Ellerington R, Garcia-Guerra A, Fratta P, Sorarú G, Greensmith L, Pennuto M, Wood MJA, Rinaldi C. AR cooperates with SMAD4 to maintain skeletal muscle homeostasis. Acta Neuropathol 2022; 143:713-731. [PMID: 35522298 PMCID: PMC9107400 DOI: 10.1007/s00401-022-02428-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/10/2022] [Accepted: 04/27/2022] [Indexed: 12/27/2022]
Abstract
Androgens and androgen-related molecules exert a plethora of functions across different tissues, mainly through binding to the transcription factor androgen receptor (AR). Despite widespread therapeutic use and misuse of androgens as potent anabolic agents, the molecular mechanisms of this effect on skeletal muscle are currently unknown. Muscle mass in adulthood is mainly regulated by the bone morphogenetic protein (BMP) axis of the transforming growth factor (TGF)-β pathway via recruitment of mothers against decapentaplegic homolog 4 (SMAD4) protein. Here we show that, upon activation, AR forms a transcriptional complex with SMAD4 to orchestrate a muscle hypertrophy programme by modulating SMAD4 chromatin binding dynamics and enhancing its transactivation activity. We challenged this mechanism of action using spinal and bulbar muscular atrophy (SBMA) as a model of study. This adult-onset neuromuscular disease is caused by a polyglutamine expansion (polyQ) in AR and is characterized by progressive muscle weakness and atrophy secondary to a combination of lower motor neuron degeneration and primary muscle atrophy. Here we found that the presence of an elongated polyQ tract impairs AR cooperativity with SMAD4, leading to an inability to mount an effective anti-atrophy gene expression programme in skeletal muscle in response to denervation. Furthermore, adeno-associated virus, serotype 9 (AAV9)-mediated muscle-restricted delivery of BMP7 is able to rescue the muscle atrophy in SBMA mice, supporting the development of treatments able to fine-tune AR-SMAD4 transcriptional cooperativity as a promising target for SBMA and other conditions associated with muscle loss.
Collapse
Affiliation(s)
- Mitra Forouhan
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Wooi Fang Lim
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Laura C Zanetti-Domingues
- Central Laser Facility, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Didcot, Oxfordshire, UK
| | - Christopher J Tynan
- Central Laser Facility, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Didcot, Oxfordshire, UK
| | - Thomas C Roberts
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Bilal Malik
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Raquel Manzano
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Alfina A Speciale
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Ruth Ellerington
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Antonio Garcia-Guerra
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Pietro Fratta
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Gianni Sorarú
- Department of Neurosciences, Neurology Unit, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Linda Greensmith
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Maria Pennuto
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Matthew J A Wood
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
- MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | - Carlo Rinaldi
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK.
- MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK.
| |
Collapse
|
82
|
Bollen SE, Bass JJ, Fujita S, Wilkinson D, Hewison M, Atherton PJ. The Vitamin D/Vitamin D receptor (VDR) axis in muscle atrophy and sarcopenia. Cell Signal 2022; 96:110355. [PMID: 35595176 DOI: 10.1016/j.cellsig.2022.110355] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 12/22/2022]
Abstract
Muscle atrophy and sarcopenia (the term given to the age-related decline in muscle mass and function), influence an individuals risk of falls, frailty, functional decline, and, ultimately, impaired quality of life. Vitamin D deficiency (low serum levels of 25-hydroxyvitamin D (25(OH)D3)) has been reported to impair muscle strength and increase risk of sarcopenia. The mechanisms that underpin the link between low 25(OH)D3 and sarcopenia are yet to be fully understood but several lines of evidence have highlighted the importance of both genomic and non-genomic effects of active vitamin D (1,25-dihydroxyvitamin D (1,25(OH)2D3)) and its nuclear vitamin D receptor (VDR), in skeletal muscle functioning. Studies in vitro have demonstrated a key role for the vitamin D/VDR axis in regulating biological processes central to sarcopenic muscle atrophy, such as proteolysis, mitochondrial function, cellular senescence, and adiposity. The aim of this review is to provide a mechanistic overview of the proposed mechanisms for the vitamin D/VDR axis in sarcopenic muscle atrophy.
Collapse
Affiliation(s)
- Shelby E Bollen
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, DE22 3DT, UK.
| | - Joseph J Bass
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, DE22 3DT, UK
| | - Satoshi Fujita
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Daniel Wilkinson
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, DE22 3DT, UK
| | - Martin Hewison
- Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Philip J Atherton
- MRC/ARUK Centre for Musculoskeletal Ageing Research and National Institute for Health Research (NIHR), Nottingham Biomedical Research Centre (BRC), School of Medicine, University of Nottingham, DE22 3DT, UK.
| |
Collapse
|
83
|
Lombardo M, Feraco A, Bellia C, Prisco L, D’Ippolito I, Padua E, Storz MA, Lauro D, Caprio M, Bellia A. Influence of Nutritional Status and Physical Exercise on Immune Response in Metabolic Syndrome. Nutrients 2022; 14:nu14102054. [PMID: 35631195 PMCID: PMC9145042 DOI: 10.3390/nu14102054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/25/2022] Open
Abstract
Metabolic Syndrome (MetS) is a cluster of metabolic alterations mostly related to visceral adiposity, which in turn promotes glucose intolerance and a chronic systemic inflammatory state, characterized by immune cell infiltration. Such immune system activation increases the risk of severe disease subsequent to viral infections. Strong correlations between elevated body mass index (BMI), type-2-diabetes and increased risk of hospitalization after pandemic influenza H1N1 infection have been described. Similarly, a correlation between elevated blood glucose level and SARS-CoV-2 infection severity and mortality has been described, indicating MetS as an important predictor of clinical outcomes in patients with COVID-19. Adipose secretome, including two of the most abundant and well-studied adipokines, leptin and interleukin-6, is involved in the regulation of energy metabolism and obesity-related low-grade inflammation. Similarly, skeletal muscle hormones—called myokines—released in response to physical exercise affect both metabolic homeostasis and immune system function. Of note, several circulating hormones originate from both adipose tissue and skeletal muscle and display different functions, depending on the metabolic context. This review aims to summarize recent data in the field of exercise immunology, investigating the acute and chronic effects of exercise on myokines release and immune system function.
Collapse
Affiliation(s)
- Mauro Lombardo
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.F.); (L.P.); (E.P.); (M.C.); (A.B.)
- Correspondence:
| | - Alessandra Feraco
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.F.); (L.P.); (E.P.); (M.C.); (A.B.)
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy
| | - Chiara Bellia
- Department of Biomedicine, Neurosciences, and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy;
| | - Luigi Prisco
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.F.); (L.P.); (E.P.); (M.C.); (A.B.)
| | - Ilenia D’Ippolito
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (I.D.); (D.L.)
| | - Elvira Padua
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.F.); (L.P.); (E.P.); (M.C.); (A.B.)
- School of Human Movement Science, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Maximilian Andreas Storz
- Department of Internal Medicine II, Center for Complementary Medicine, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| | - Davide Lauro
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (I.D.); (D.L.)
| | - Massimiliano Caprio
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.F.); (L.P.); (E.P.); (M.C.); (A.B.)
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy
| | - Alfonso Bellia
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy; (A.F.); (L.P.); (E.P.); (M.C.); (A.B.)
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (I.D.); (D.L.)
| |
Collapse
|
84
|
"HIIT the Inflammation": Comparative Effects of Low-Volume Interval Training and Resistance Exercises on Inflammatory Indices in Obese Metabolic Syndrome Patients Undergoing Caloric Restriction. Nutrients 2022; 14:nu14101996. [PMID: 35631137 PMCID: PMC9145085 DOI: 10.3390/nu14101996] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 02/01/2023] Open
Abstract
Exercise is a cornerstone in metabolic syndrome (MetS) treatment. However, the effects of low-volume exercise modalities on MetS-associated low-grade inflammation are unclear. A total of 106 MetS patients (53.7 ± 11.4 years) were randomized to low-volume high-intensity interval training (LOW-HIIT, 14 min/session), single-set resistance training (1-RT, ~15 min/session), whole-body electromyostimulation (WB-EMS, 20 min/session), three-set resistance training (3-RT, ~50 min/session), each performed 2 ×/week for 12 weeks, or a control group (CON). All groups received nutritional counseling for weight loss. Inflammatory and cardiometabolic indices were analyzed pre- and post-intervention. All groups significantly reduced body weight by an average of 3.6%. Only LOW-HIIT reduced C-reactive protein (CRP) (−1.6 mg/L, p = 0.001) and interleukin-6 (−1.1 pg/mL, p = 0.020). High-sensitivity CRP and lipopolysaccharide-binding protein decreased following LOW-HIIT (−1.4 mg/L, p = 0.001 and −2.1 ng/mL, p = 0.004) and 3-RT (−0.6 mg/L, p = 0.044 and −2.0 ng/mL, p < 0.001). MetS severity score improved with LOW-HIIT (−1.8 units, p < 0.001), 1-RT (−1.6 units, p = 0.005), and 3-RT (−2.3 units, p < 0.001). Despite similar effects on body weight, low-volume exercise modalities have different impact on inflammatory and cardiometabolic outcomes in MetS patients. LOW-HIIT has superior efficacy for improving inflammation compared to 1-RT and WB-EMS. Resistance-based exercise appears to require a higher volume to promote beneficial impact on inflammation.
Collapse
|
85
|
Kim YJ, Kim HJ, Lee SG, Kim DH, In Jang S, Go HS, Lee WJ, Seong JK. Aerobic exercise for eight weeks provides protective effects towards liver and cardiometabolic health and adipose tissue remodeling under metabolic stress for one week: A study in mice. Metabolism 2022; 130:155178. [PMID: 35227728 DOI: 10.1016/j.metabol.2022.155178] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/07/2022] [Accepted: 02/20/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND The relationship between exercise training and health benefits is under thorough investigation. However, the effects of exercise training on the maintenance of metabolic health are unclear. METHODS Our experimental design involved initial exercise training followed by a high-fat diet (HFD) challenge. Eight-week-old male was trained under voluntary wheel running aerobic exercise for eight weeks to determine the systemic metabolic changes induced by exercise training and whether such changes persisted even after discontinuing exercise. The mice were given either a normal chow diet (NCD) or HFD ad libitum for one week after discontinuation of exercise (CON-NCD, n = 29; EX-NCD, n = 29; CON-HFD, n = 30; EX-HFD, n = 31). RESULTS Our study revealed that metabolic stress following the transition to an HFD in mice that discontinued training failed to reverse the aerobic exercise training-induced improvement in metabolism. We report that the mice subjected to exercise training could better counteract weight gain, adipose tissue hypertrophy, insulin resistance, fatty liver, and mitochondrial dysfunction in response to an HFD compared with untrained mice. This observation could be attributed to the fact that exercise enhances the browning of white fat, whole-body oxygen uptake, and heat generation. Furthermore, we suggest that the effects of exercise persist due to PPARα-FGF21-FGFR1 mechanisms, although additional pathways cannot be excluded and require further research. Although our study suggests the preventive potential of exercise, appropriate human trials are needed to demonstrate the efficacy in subjects who cannot perform sustained exercise; this may provide an important basis regarding human health.
Collapse
Affiliation(s)
- Youn Ju Kim
- Laboratory of Developmental Biology and Genomics, BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea; The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Korea Mouse Phenotyping Center (KMPC), Seoul National University, 08826 Seoul, Republic of Korea
| | - Hye Jin Kim
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Korea Mouse Phenotyping Center (KMPC), Seoul National University, 08826 Seoul, Republic of Korea
| | - Sang Gyu Lee
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, 08826 Seoul, Republic of Korea
| | - Do Hyun Kim
- Laboratory of Developmental Biology and Genomics, BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea; The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Korea Mouse Phenotyping Center (KMPC), Seoul National University, 08826 Seoul, Republic of Korea
| | - Su In Jang
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, 08826 Seoul, Republic of Korea
| | - Hye Sun Go
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, 08826 Seoul, Republic of Korea
| | - Won Jun Lee
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, 08826 Seoul, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea; The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Korea Mouse Phenotyping Center (KMPC), Seoul National University, 08826 Seoul, Republic of Korea; Interdisciplinary Program for Bioinformatics, Program for Cancer Biology, BIO-MAX/N-Bio Institute, Seoul National University, 08826 Seoul, Republic of Korea.
| |
Collapse
|
86
|
JO AL, HAN JW, AN JI, CHO KH, JEOUNG NH. Cuban Policosanol Prevents the Apoptosis and the Mitochondrial Dysfunction Induced by Lipopolysaccharide in C2C12 Myoblast via Activation of Akt and Erk Pathways. J Nutr Sci Vitaminol (Tokyo) 2022; 68:79-86. [DOI: 10.3177/jnsv.68.79] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Ae Lim JO
- Department of Pharmaceutical Engineering, Deagu Catholic University
| | - Ji Won HAN
- Department of Pharmaceutical Engineering, Deagu Catholic University
| | - Ji In AN
- Department of Pharmaceutical Engineering, Deagu Catholic University
| | | | - Nam Ho JEOUNG
- Department of Pharmaceutical Engineering, Deagu Catholic University
| |
Collapse
|
87
|
Monotropein Improves Dexamethasone-Induced Muscle Atrophy via the AKT/mTOR/FOXO3a Signaling Pathways. Nutrients 2022; 14:nu14091859. [PMID: 35565825 PMCID: PMC9103778 DOI: 10.3390/nu14091859] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
The present study aimed to investigate the effects of monotropein (MON) on improving dexamethasone (DEX)-induced muscle atrophy in mice and C2C12 mouse skeletal muscle cells. The body weights, grip strengths, and muscle weights of mice were assessed. The histological change in the gastrocnemius tissues was also observed through H&E staining. The expression of myosin heavy chain (MyHC), muscle ring finger 1 (MuRF1), and muscle atrophy F-box (Atrogin1) and the phosphorylation of AKT, mTOR, and FOXO3a in the muscle tissues of mice and C2C12 myotubes were analyzed using Western blotting. MON improved muscle atrophy in mice and C2C12 myotubes by regulating catabolic states via the AKT/mTOR/FOXO3a signaling pathways, and enhanced muscle function by the increases of muscle mass and strength in mice. This suggests that MON could be used for the prevention and treatment of muscle atrophy in patients.
Collapse
|
88
|
Vaca-Dempere M, Kumar A, Sica V, Muñoz-Cánoves P. Running skeletal muscle clocks on time- the determining factors. Exp Cell Res 2022; 413:112989. [PMID: 35081395 DOI: 10.1016/j.yexcr.2021.112989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 12/08/2021] [Accepted: 12/19/2021] [Indexed: 11/23/2022]
Abstract
Circadian rhythms generate 24 h-long oscillations, which are key regulators of many aspects of behavior and physiology. Recent circadian transcriptome studies have discovered rhythmicity at the transcriptional level of hundreds of skeletal muscle genes, with roles in skeletal muscle growth, maintenance, and metabolic functions. These rhythms allow this tissue to perform molecular functions at the appropriate time of the day in order to anticipate environmental changes. However, while the last decade of research has characterized several aspects of the skeletal muscle molecular clock, many still are unexplored, including its functions, regulatory mechanisms, and interactions with other tissues. The central clock is believed to be located in the suprachiasmatic nucleus (SCN) of the brain hypothalamus, providing entrainment to peripheral organs through humoral and neuronal signals. However, these mechanisms of action are still unknown. Conversely, muscle tissue can be entrained through extrinsic, SCN-independent factors, such as feeding and physical activity. In this review, we provide an overview of the recent research about the extrinsic and intrinsic factors required for skeletal muscle clock regulation. Furthermore, we discuss the need for future studies to elucidate the mechanisms behind this regulation, which will in turn help dissect the role of circadian disruption at the onset of aging and diseases.
Collapse
Affiliation(s)
- Mireia Vaca-Dempere
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain
| | - Arun Kumar
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain
| | - Valentina Sica
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain; ICREA, 08010, Barcelona, Spain; Spanish National Center on Cardiovascular Research (CNIC), 28029, Madrid, Spain.
| |
Collapse
|
89
|
McNally BD, Ashley DF, Hänschke L, Daou HN, Watt NT, Murfitt SA, MacCannell ADV, Whitehead A, Bowen TS, Sanders FWB, Vacca M, Witte KK, Davies GR, Bauer R, Griffin JL, Roberts LD. Long-chain ceramides are cell non-autonomous signals linking lipotoxicity to endoplasmic reticulum stress in skeletal muscle. Nat Commun 2022; 13:1748. [PMID: 35365625 PMCID: PMC8975934 DOI: 10.1038/s41467-022-29363-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/09/2022] [Indexed: 12/14/2022] Open
Abstract
The endoplasmic reticulum (ER) regulates cellular protein and lipid biosynthesis. ER dysfunction leads to protein misfolding and the unfolded protein response (UPR), which limits protein synthesis to prevent cytotoxicity. Chronic ER stress in skeletal muscle is a unifying mechanism linking lipotoxicity to metabolic disease. Unidentified signals from cells undergoing ER stress propagate paracrine and systemic UPR activation. Here, we induce ER stress and lipotoxicity in myotubes. We observe ER stress-inducing lipid cell non-autonomous signal(s). Lipidomics identifies that palmitate-induced cell stress induces long-chain ceramide 40:1 and 42:1 secretion. Ceramide synthesis through the ceramide synthase 2 de novo pathway is regulated by UPR kinase Perk. Inactivation of CerS2 in mice reduces systemic and muscle ceramide signals and muscle UPR activation. The ceramides are packaged into extracellular vesicles, secreted and induce UPR activation in naïve myotubes through dihydroceramide accumulation. This study furthers our understanding of ER stress by identifying UPR-inducing cell non-autonomous signals.
Collapse
Affiliation(s)
- Ben D McNally
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK
| | - Dean F Ashley
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK
| | - Lea Hänschke
- Life & Medical Sciences Institute (LIMES) Development, Genetics & Molecular Physiology Unit, University of Bonn, Carl-Troll-Straße, 31, 53115, Bonn, Germany
| | - Hélène N Daou
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Nicole T Watt
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Steven A Murfitt
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK
| | | | - Anna Whitehead
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - T Scott Bowen
- Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | | | - Michele Vacca
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK.,Clinica Medica "Frugoni", Interdisciplinar Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Klaus K Witte
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Graeme R Davies
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Reinhard Bauer
- Life & Medical Sciences Institute (LIMES) Development, Genetics & Molecular Physiology Unit, University of Bonn, Carl-Troll-Straße, 31, 53115, Bonn, Germany
| | - Julian L Griffin
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK.,Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Lee D Roberts
- School of Medicine, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
90
|
Kang BB, Chiang BH. A novel phenolic formulation for treating hepatic and peripheral insulin resistance by regulating GLUT4-mediated glucose uptake. J Tradit Complement Med 2022; 12:195-205. [PMID: 35528476 PMCID: PMC9072824 DOI: 10.1016/j.jtcme.2021.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/07/2021] [Accepted: 08/07/2021] [Indexed: 11/26/2022] Open
|
91
|
Imdad S, Lim W, Kim JH, Kang C. Intertwined Relationship of Mitochondrial Metabolism, Gut Microbiome and Exercise Potential. Int J Mol Sci 2022; 23:ijms23052679. [PMID: 35269818 PMCID: PMC8910986 DOI: 10.3390/ijms23052679] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
The microbiome has emerged as a key player contributing significantly to the human physiology over the past decades. The potential microbial niche is largely unexplored in the context of exercise enhancing capacity and the related mitochondrial functions. Physical exercise can influence the gut microbiota composition and diversity, whereas a sedentary lifestyle in association with dysbiosis can lead to reduced well-being and diseases. Here, we have elucidated the importance of diverse microbiota, which is associated with an individual's fitness, and moreover, its connection with the organelle, the mitochondria, which is the hub of energy production, signaling, and cellular homeostasis. Microbial by-products, such as short-chain fatty acids, are produced during regular exercise that can enhance the mitochondrial capacity. Therefore, exercise can be employed as a therapeutic intervention to circumvent or subside various metabolic and mitochondria-related diseases. Alternatively, the microbiome-mitochondria axis can be targeted to enhance exercise performance. This review furthers our understanding about the influence of microbiome on the functional capacity of the mitochondria and exercise performance, and the interplay between them.
Collapse
Affiliation(s)
- Saba Imdad
- Molecular Metabolism in Health & Disease, Exercise Physiology Laboratory, Sport Science Research Institute, Inha University, Incheon 22212, Korea;
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju 28503, Korea
| | - Wonchung Lim
- Department of Sports Medicine, College of Health Science, Cheongju University, Cheongju 28503, Korea;
| | - Jin-Hee Kim
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju 28503, Korea
- Correspondence: (J.-H.K.); (C.K.)
| | - Chounghun Kang
- Molecular Metabolism in Health & Disease, Exercise Physiology Laboratory, Sport Science Research Institute, Inha University, Incheon 22212, Korea;
- Department of Physical Education, College of Education, Inha University, Incheon 22212, Korea
- Correspondence: (J.-H.K.); (C.K.)
| |
Collapse
|
92
|
Nutrients against Glucocorticoid-Induced Muscle Atrophy. Foods 2022; 11:foods11050687. [PMID: 35267320 PMCID: PMC8909279 DOI: 10.3390/foods11050687] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/15/2022] [Accepted: 02/23/2022] [Indexed: 11/29/2022] Open
Abstract
Glucocorticoid excess is a critical factor contributing to muscle atrophy. Both endogenous and exogenous glucocorticoids negatively affect the preservation of muscle mass and function. To date, the most effective intervention to prevent muscle atrophy is to apply a mechanical load in the form of resistance exercise. However, glucocorticoid-induced skeletal muscle atrophy easily causes fatigue in daily physical activities, such as climbing stairs and walking at a brisk pace, and reduces body movements to cause a decreased ability to perform physical activity. Therefore, providing adequate nutrients in these circumstances is a key factor in limiting muscle wasting and improving muscle mass recovery. The present review will provide an up-to-date review of the effects of various nutrients, including amino acids such as branched-chain amino acids (BCAAs) and β–hydroxy β–methylbutyrate (HMB), fatty acids such as omega-3, and vitamins and their derivates on the prevention and improvement of glucocorticoid-induced muscle atrophy.
Collapse
|
93
|
Renzini A, D’Onghia M, Coletti D, Moresi V. Histone Deacetylases as Modulators of the Crosstalk Between Skeletal Muscle and Other Organs. Front Physiol 2022; 13:706003. [PMID: 35250605 PMCID: PMC8895239 DOI: 10.3389/fphys.2022.706003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 01/31/2022] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle plays a major role in controlling body mass and metabolism: it is the most abundant tissue of the body and a major source of humoral factors; in addition, it is primarily responsible for glucose uptake and storage, as well as for protein metabolism. Muscle acts as a metabolic hub, in a crosstalk with other organs and tissues, such as the liver, the brain, and fat tissue. Cytokines, adipokines, and myokines are pivotal mediators of such crosstalk. Many of these circulating factors modulate histone deacetylase (HDAC) expression and/or activity. HDACs form a numerous family of enzymes, divided into four classes based on their homology to their orthologs in yeast. Eleven family members are considered classic HDACs, with a highly conserved deacetylase domain, and fall into Classes I, II, and IV, while class III members are named Sirtuins and are structurally and mechanistically distinct from the members of the other classes. HDACs are key regulators of skeletal muscle metabolism, both in physiological conditions and following metabolic stress, participating in the highly dynamic adaptative responses of the muscle to external stimuli. In turn, HDAC expression and activity are closely regulated by the metabolic demands of the skeletal muscle. For instance, NAD+ levels link Class III (Sirtuin) enzymatic activity to the energy status of the cell, and starvation or exercise affect Class II HDAC stability and intracellular localization. SUMOylation or phosphorylation of Class II HDACs are modulated by circulating factors, thus establishing a bidirectional link between HDAC activity and endocrine, paracrine, and autocrine factors. Indeed, besides being targets of adipo-myokines, HDACs affect the synthesis of myokines by skeletal muscle, altering the composition of the humoral milieu and ultimately contributing to the muscle functioning as an endocrine organ. In this review, we discuss recent findings on the interplay between HDACs and circulating factors, in relation to skeletal muscle metabolism and its adaptative response to energy demand. We believe that enhancing knowledge on the specific functions of HDACs may have clinical implications leading to the use of improved HDAC inhibitors for the treatment of metabolic syndromes or aging.
Collapse
Affiliation(s)
- Alessandra Renzini
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Marco D’Onghia
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Dario Coletti
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
- Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Sorbonne Université, Paris, France
| | - Viviana Moresi
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
- Institute of Nanotechnology (Nanotec), National Research Council, Rome, Italy
| |
Collapse
|
94
|
Bernacchioni C, Squecco R, Gamberi T, Ghini V, Schumacher F, Mannelli M, Garella R, Idrizaj E, Cencetti F, Puliti E, Bruni P, Turano P, Fiaschi T, Donati C. S1P Signalling Axis Is Necessary for Adiponectin-Directed Regulation of Electrophysiological Properties and Oxidative Metabolism in C2C12 Myotubes. Cells 2022; 11:713. [PMID: 35203362 PMCID: PMC8869893 DOI: 10.3390/cells11040713] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Adiponectin (Adn), released by adipocytes and other cell types such as skeletal muscle, has insulin-sensitizing and anti-inflammatory properties. Sphingosine 1-phosphate (S1P) is reported to act as effector of diverse biological actions of Adn in different tissues. S1P is a bioactive sphingolipid synthesized by the phosphorylation of sphingosine catalyzed by sphingosine kinase (SK) 1 and 2. Consolidated findings support the key role of S1P in the biology of skeletal muscle. METHODS AND RESULTS Here we provide experimental evidence that S1P signalling is modulated by globular Adn treatment being able to increase the phosphorylation of SK1/2 as well as the mRNA expression levels of S1P4 in C2C12 myotubes. These findings were confirmed by LC-MS/MS that showed an increase of S1P levels after Adn treatment. Notably, the involvement of S1P axis in Adn action was highlighted since, when SK1 and 2 were inhibited by PF543 and ABC294640 inhibitors, respectively, not only the electrophysiological changes but also the increase of oxygen consumption and of aminoacid levels induced by the hormone, were significantly inhibited. CONCLUSION Altogether, these findings show that S1P biosynthesis is necessary for the electrophysiological properties and oxidative metabolism of Adn in skeletal muscle cells.
Collapse
Affiliation(s)
- Caterina Bernacchioni
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Roberta Squecco
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (R.S.); (R.G.); (E.I.)
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Veronica Ghini
- Magnetic Resonance Center (CERM), University of Florence, 50019 Florence, Italy; (V.G.); (P.T.)
| | - Fabian Schumacher
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany;
| | - Michele Mannelli
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Rachele Garella
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (R.S.); (R.G.); (E.I.)
| | - Eglantina Idrizaj
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (R.S.); (R.G.); (E.I.)
| | - Francesca Cencetti
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Elisa Puliti
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Paola Bruni
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Paola Turano
- Magnetic Resonance Center (CERM), University of Florence, 50019 Florence, Italy; (V.G.); (P.T.)
| | - Tania Fiaschi
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| |
Collapse
|
95
|
Xu Z, Fu T, Guo Q, Zhou D, Sun W, Zhou Z, Chen X, Zhang J, Liu L, Xiao L, Yin Y, Jia Y, Pang E, Chen Y, Pan X, Fang L, Zhu MS, Fei W, Lu B, Gan Z. Disuse-associated loss of the protease LONP1 in muscle impairs mitochondrial function and causes reduced skeletal muscle mass and strength. Nat Commun 2022; 13:894. [PMID: 35173176 PMCID: PMC8850466 DOI: 10.1038/s41467-022-28557-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 02/02/2022] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial proteolysis is an evolutionarily conserved quality-control mechanism to maintain proper mitochondrial integrity and function. However, the physiological relevance of stress-induced impaired mitochondrial protein quality remains unclear. Here, we demonstrate that LONP1, a major mitochondrial protease resides in the matrix, plays a role in controlling mitochondrial function as well as skeletal muscle mass and strength in response to muscle disuse. In humans and mice, disuse-related muscle loss is associated with decreased mitochondrial LONP1 protein. Skeletal muscle-specific ablation of LONP1 in mice resulted in impaired mitochondrial protein turnover, leading to mitochondrial dysfunction. This caused reduced muscle fiber size and strength. Mechanistically, aberrant accumulation of mitochondrial-retained protein in muscle upon loss of LONP1 induces the activation of autophagy-lysosome degradation program of muscle loss. Overexpressing a mitochondrial-retained mutant ornithine transcarbamylase (ΔOTC), a known protein degraded by LONP1, in skeletal muscle induces mitochondrial dysfunction, autophagy activation, and cause muscle loss and weakness. Thus, these findings reveal a role of LONP1-dependent mitochondrial protein quality-control in safeguarding mitochondrial function and preserving skeletal muscle mass and strength, and unravel a link between mitochondrial protein quality and muscle mass maintenance during muscle disuse. Mitochondrial function is important for muscle maintenance and function, and mitochondrial proteolysis maintains mitochondrial integrity and function. Here the authors report that that loss of LONP1-dependent mitochondrial proteolysis in muscle causes reduced muscle mass and strength via activation of autophagy.
Collapse
Affiliation(s)
- Zhisheng Xu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Tingting Fu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Qiqi Guo
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Danxia Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Wanping Sun
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Zheng Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Xinyi Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Jingzi Zhang
- Jiangsu Key Laboratory of Molecular Medicine & Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing, China
| | - Lin Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Liwei Xiao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Yujing Yin
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Yuhuan Jia
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Erkai Pang
- Sports Medicine Department, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Yuncong Chen
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Xin Pan
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Lei Fang
- Jiangsu Key Laboratory of Molecular Medicine & Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing, China
| | - Min-Sheng Zhu
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Wenyong Fei
- Sports Medicine Department, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Bin Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhenji Gan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing, China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing University, Nanjing, China. .,Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China.
| |
Collapse
|
96
|
Li P, Xu R, Shi Y, Shi X, Zhang X, Li J, Kou G. Luteolin increases slow muscle fibers via FLCN-AMPK-PGC-1α signaling pathway. J Funct Foods 2022. [DOI: 10.1016/j.jff.2021.104876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
97
|
Vladimirsky VE, Vladimirsky EV, Lunina AN, Fesyun AD, Rachin AP, Lebedeva OD, Yakovlev MY, Tubekova MA. [Molecular mechanisms of adaptive and therapeutic effects of physical activity in patients with cardiovascular diseases]. VOPROSY KURORTOLOGII, FIZIOTERAPII, I LECHEBNOI FIZICHESKOI KULTURY 2022; 99:69-77. [PMID: 35485663 DOI: 10.17116/kurort20229902169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Physical activity is one of the main components of the rehabilitation of patients with cardiovascular disease (CVD). As shown by practice and the results of evidence-based studies, the beneficial effects of physical activity on disease outcomes in a number of cardiac nosologies are comparable to drug treatment. This gives the doctor another tool to influence the unfavorable epidemiological situation in developed countries with the spread of diseases of the cardiovascular system and CVD mortality. Reliable positive results of cardiorehabilitation (CR) were obtained using various methods. The goal of CR is to restore the optimal physiological, psychological and professional status, reduce the risk of CVD and mortality. In most current CVD guidelines worldwide, cardiac rehabilitation is a Class I recommendation. The molecular mechanisms described in the review, initiated by physical activity, underlie the multifactorial effect of the latter on the function of the cardiovascular system and the course of cardiac diseases. Physical exercise is an important component of the therapeutic management of patients with CVD, which is supported by the results of a meta-analysis of 63 studies associated with various forms of aerobic exercise of varying intensity (from 50 to 95% VO2) for 1 to 47 months, which showed that CR based on physical exercise improves cardiorespiratory endurance. Knowledge of the molecular basis of the influence of physical activity makes it possible to use biochemical markers to assess the effectiveness of rehabilitation programs.
Collapse
Affiliation(s)
| | | | - A N Lunina
- Wagner Perm State Medical University, Perm, Russia
| | - A D Fesyun
- National Medical Research Center for Rehabilitation and Balneology, Moscow, Russia
| | - A P Rachin
- National Medical Research Center for Rehabilitation and Balneology, Moscow, Russia
| | - O D Lebedeva
- National Medical Research Center for Rehabilitation and Balneology, Moscow, Russia
| | - M Yu Yakovlev
- National Medical Research Center for Rehabilitation and Balneology, Moscow, Russia
| | - M A Tubekova
- National Medical Research Center for Rehabilitation and Balneology, Moscow, Russia
| |
Collapse
|
98
|
No Atrophy Let’s Hypertrophy for Better Sporting Events. TURKISH JOURNAL OF KINESIOLOGY 2021. [DOI: 10.31459/turkjkin.1010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
99
|
Cogliati S, Cabrera-Alarcón JL, Enriquez JA. Regulation and functional role of the electron transport chain supercomplexes. Biochem Soc Trans 2021; 49:2655-2668. [PMID: 34747989 PMCID: PMC8786287 DOI: 10.1042/bst20210460] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 12/17/2022]
Abstract
Mitochondria are one of the most exhaustively investigated organelles in the cell and most attention has been paid to the components of the mitochondrial electron transport chain (ETC) in the last 100 years. The ETC collects electrons from NADH or FADH2 and transfers them through a series of electron carriers within multiprotein respiratory complexes (complex I to IV) to oxygen, therefore generating an electrochemical gradient that can be used by the F1-F0-ATP synthase (also named complex V) in the mitochondrial inner membrane to synthesize ATP. The organization and function of the ETC is a continuous source of surprises. One of the latest is the discovery that the respiratory complexes can assemble to form a variety of larger structures called super-complexes (SCs). This opened an unexpected level of complexity in this well-known and fundamental biological process. This review will focus on the current evidence for the formation of different SCs and will explore how they modulate the ETC organization according to the metabolic state. Since the field is rapidly growing, we also comment on the experimental techniques used to describe these SC and hope that this overview may inspire new technologies that will help to advance the field.
Collapse
Affiliation(s)
- Sara Cogliati
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | | | - Jose Antonio Enriquez
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| |
Collapse
|
100
|
Yun CE, So HK, Vuong TA, Na MW, Anh S, Lee HK, Kim KH, Kang JS, Bae GU, Lee SJ. Aronia Upregulates Myogenic Differentiation and Augments Muscle Mass and Function Through Muscle Metabolism. Front Nutr 2021; 8:753643. [PMID: 34888337 PMCID: PMC8650690 DOI: 10.3389/fnut.2021.753643] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/11/2021] [Indexed: 11/16/2022] Open
Abstract
Black chokeberry or aronia (the fruit of Aronia melanocarpa) has been reported to having pharmacological activities against metabolic syndrome, such as hypertension, obesity, diabetes, and pro-inflammatory conditions. However, the effects of aronia on myogenic differentiation and muscle homoeostasis are uncharacterized. In this study, we investigated the effects of aronia (black chokeberry) on myogenic differentiation and muscle metabolic functions in young mice. Aronia extract (AR) promotes myogenic differentiation and elevates the formation of multinucleated myotubes through Akt activation. AR protects dexamethasone (DEX)-induced myotube atrophy through inhibition of muscle-specific ubiquitin ligases mediated by Akt activation. The treatment with AR increases muscle mass and strength in mice without cardiac hypertrophy. AR treatment enhances both oxidative and glycolytic myofibers and muscle metabolism with elevated mitochondrial genes and glucose metabolism-related genes. Furthermore, AR-fed muscle fibers display increased levels of total OxPHOS and myoglobin proteins. Taken together, AR enhances myogenic differentiation and improves muscle mass and function, suggesting that AR has a promising potential as a nutraceutical remedy to intervene in muscle weakness and atrophy.
Collapse
Affiliation(s)
- Chae-Eun Yun
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Hyun-Kyung So
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea.,Research Institute of Aging Related Disease, AniMusCure Inc., Suwon, South Korea
| | - Tuan Anh Vuong
- Research Institute of Aging Related Disease, AniMusCure Inc., Suwon, South Korea
| | - Myung Woo Na
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Subin Anh
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Hyo-Keun Lee
- Gyeonwoo Korean Medical Center, Seoul, South Korea
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Gyu-Un Bae
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, South Korea
| | - Sang-Jin Lee
- Research Institute of Aging Related Disease, AniMusCure Inc., Suwon, South Korea
| |
Collapse
|