1
|
Hoseini R, Hoseini Z, Kamangar A. Myogenic differentiation markers in muscle tissue after aerobic training. Heliyon 2025; 11:e41888. [PMID: 39897925 PMCID: PMC11787638 DOI: 10.1016/j.heliyon.2025.e41888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 11/21/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
Aerobic training induces a myriad of adaptations in muscle tissue, encompassing alterations in muscle fiber type composition, hypertrophy, and metabolic capacity. Understanding the potential role of myogenic differentiation markers (MDFs), such as Pax7, MyoD, Myogenin, and myosin heavy chain (MHC) isoforms, in mediating these adaptations is of paramount importance. The review delves into the intricate molecular mechanisms underlying the regulation of MDFs following aerobic training, elucidating the role of key signaling pathways including the MAPK/ERK, PI3K/Akt, and AMPK pathways, among others. These pathways play pivotal roles in orchestrating the expression and activity of MDFs, ultimately influencing muscle adaptation and regeneration. The comprehension of MDFs in the context of aerobic training is far-reaching, offering the potential for targeted interventions to optimize muscle adaptation and regeneration. This review identifies the need for further research to unveil the precise molecular mechanisms of the activation and interaction of myogenic differentiation markers with other signaling pathways, as well as to explore their potential as therapeutic targets for muscle-related conditions. This review article also provides a thorough analysis of MDFs in muscle tissue after aerobic training, highlighting their potential clinical implications and outlining future research directions in this area.
Collapse
Affiliation(s)
- Rastegar Hoseini
- Assistant Professor of Exercise Physiology, Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Zahra Hoseini
- PhD of Exercise Physiology, Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Ayob Kamangar
- PhD Student of Exercise Physiology, Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
2
|
Wang J, Jia D, Zhang Z, Wang D. Exerkines and Sarcopenia: Unveiling the Mechanism Behind Exercise-Induced Mitochondrial Homeostasis. Metabolites 2025; 15:59. [PMID: 39852400 PMCID: PMC11767263 DOI: 10.3390/metabo15010059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: Sarcopenia, characterized by the progressive loss of muscle mass and strength, is linked to physical disability, metabolic dysfunction, and an increased risk of mortality. Exercise therapy is currently acknowledged as a viable approach for addressing sarcopenia. Nevertheless, the molecular mechanisms behind exercise training or physical activity remain poorly understood. The disruption of mitochondrial homeostasis is implicated in the pathogenesis of sarcopenia. Exercise training effectively delays the onset of sarcopenia by significantly maintaining mitochondrial homeostasis, including promoting mitophagy, improving mitochondrial biogenesis, balancing mitochondrial dynamics, and maintaining mitochondrial redox. Exerkines (e.g., adipokines, myokines, hepatokines, and osteokines), signaling molecules released in response to exercise training, may potentially contribute to skeletal muscle metabolism through ameliorating mitochondrial homeostasis, reducing inflammation, and regulating protein synthesis as a defense against sarcopenia. Methods: In this review, we provide a detailed summary of exercise-induced exerkines and confer their benefit, with particular focus on their impact on mitochondrial homeostasis in the context of sarcopenia. Results: Exercise induces substantial adaptations in skeletal muscle, including increased muscle mass, improved muscle regeneration and hypertrophy, elevated hormone release, and enhanced mitochondrial function. An expanding body of research highlights that exerkines have the potential to regulate processes such as mitophagy, mitochondrial biogenesis, dynamics, autophagy, and redox balance. These mechanisms contribute to the maintenance of mitochondrial homeostasis, thereby supporting skeletal muscle metabolism and mitochondrial health. Conclusions: Through a comprehensive investigation of the molecular mechanisms within mitochondria, the context reveals new insights into the potential of exerkines as key exercise-protective sensors for combating sarcopenia.
Collapse
Affiliation(s)
- Jiayin Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (D.J.)
| | - Dandan Jia
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (D.J.)
| | - Zhiwang Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (J.W.); (D.J.)
| | - Dan Wang
- School of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
3
|
Wang T, Zhou D, Hong Z. Sarcopenia and cachexia: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2025; 6:e70030. [PMID: 39764565 PMCID: PMC11702502 DOI: 10.1002/mco2.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 03/17/2025] Open
Abstract
Sarcopenia is defined as a muscle-wasting syndrome that occurs with accelerated aging, while cachexia is a severe wasting syndrome associated with conditions such as cancer and immunodeficiency disorders, which cannot be fully addressed through conventional nutritional supplementation. Sarcopenia can be considered a component of cachexia, with the bidirectional interplay between adipose tissue and skeletal muscle potentially serving as a molecular mechanism for both conditions. However, the underlying mechanisms differ. Recognizing the interplay and distinctions between these disorders is essential for advancing both basic and translational research in this area, enhancing diagnostic accuracy and ultimately achieving effective therapeutic solutions for affected patients. This review discusses the muscle microenvironment's changes contributing to these conditions, recent therapeutic approaches like lifestyle modifications, small molecules, and nutritional interventions, and emerging strategies such as gene editing, stem cell therapy, and gut microbiome modulation. We also address the challenges and opportunities of multimodal interventions, aiming to provide insights into the pathogenesis and molecular mechanisms of sarcopenia and cachexia, ultimately aiding in innovative strategy development and improved treatments.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Dong Zhou
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Zhen Hong
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| |
Collapse
|
4
|
Samant V, Prabhu A. Exercise, exerkines and exercise mimetic drugs: Molecular mechanisms and therapeutics. Life Sci 2024; 359:123225. [PMID: 39522716 DOI: 10.1016/j.lfs.2024.123225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/09/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Chronic diseases linked with sedentary lifestyles and poor dietary habits are increasingly common in modern society. Exercise is widely acknowledged to have a plethora of health benefits, including its role in primary prevention of various chronic conditions like type 2 diabetes mellitus, obesity, cardiovascular disease, and several musculoskeletal as well as degenerative disorders. Regular physical activity induces numerous physiological adaptations that contribute to these positive effects, primarily observed in skeletal muscle but also impacting other tissues. There is a growing interest among researchers in developing pharmaceutical interventions that mimic the beneficial effects of exercise for therapeutic applications. Exercise mimetic medications have the potential to be helpful aids in enhancing functional outcomes for patients with metabolic dysfunction, neuromuscular and musculoskeletal disorders. Some of the potential targets for exercise mimetics include pathways involved in metabolism, mitochondrial function, inflammation, and tissue regeneration. The present review aims to provide an exhaustive overview of the current understanding of exercise physiology, the role of exerkines and biomolecular pathways, and the potential applications of exercise mimetic drugs for the treatment of several diseases.
Collapse
Affiliation(s)
- Vedant Samant
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Arati Prabhu
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India.
| |
Collapse
|
5
|
Ji RC. The emerging importance of lymphangiogenesis in aging and aging-associated diseases. Mech Ageing Dev 2024; 221:111975. [PMID: 39089499 DOI: 10.1016/j.mad.2024.111975] [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: 05/15/2024] [Revised: 07/17/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Lymphatic aging represented by cellular and functional changes, is involved in increased geriatric disorders, but the intersection between aging and lymphatic modulation is less clear. Lymphatic vessels play an essential role in maintaining tissue fluid homeostasis, regulating immune function, and promoting macromolecular transport. Lymphangiogenesis and lymphatic remodeling following cellular senescence and organ deterioration are crosslinked with the progression of some lymphatic-associated diseases, e.g., atherosclerosis, inflammation, lymphoedema, and cancer. Age-related detrimental tissue changes may occur in lymphatic vessels with diverse etiologies, and gradually shift towards chronic low-grade inflammation, so-called inflammaging, and lead to decreased immune response. The investigation of the relationship between advanced age and organ deterioration is becoming an area of rapidly increasing significance in lymphatic biology and medicine. Here we highlight the emerging importance of lymphangiogenesis and lymphatic remodeling in the regulation of aging-related pathological processes, which will help to find new avenues for effective intervention to promote healthy aging.
Collapse
Affiliation(s)
- Rui-Cheng Ji
- Faculty of Welfare and Health Science, Oita University, Oita 870-1192, Japan.
| |
Collapse
|
6
|
Chen Z, Sun J, Shi T, Song C, Wu C, Wu Z, Lin J. Causal roles of circulating cytokines in sarcopenia-related traits: a Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1370985. [PMID: 39345889 PMCID: PMC11427268 DOI: 10.3389/fendo.2024.1370985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/28/2024] [Indexed: 10/01/2024] Open
Abstract
Background Epidemiological and experimental evidence suggests that chronic inflammation plays an important role in the onset and progression of sarcopenia. However, there is inconsistent data on the inflammatory cytokines involved in the pathogenesis of sarcopenia. Therefore, we performed a two-sample Mendelian randomization (MR) analysis to explore the causal relationship between circulating cytokines and sarcopenia-related traits. Methods The MR analysis utilized genetic data from genome-wide association study that included genetic variations in 41 circulating cytokines and genetic variant data for appendicular lean mass (ALM), hand grip strength, and usual walking pace. Causal associations were primarily explored using the inverse variance-weighted (IVW) method, supplemented by MR-Egger, simple mode, weighted median, and weighted mode analyses. Additionally, sensitivity analyses were also performed to ensure the reliability and stability of the results. Results Three cytokines [hepatocyte growth factor (HGF), interferon gamma-induced protein 10 (IP-10), and macrophage colony-stimulating factor (M-CSF)] were positively associated with ALM (β: 0.0221, 95% confidence interval (CI): 0.0071, 0.0372, P= 0.0039 for HGF; β: 0.0096, 95%CI: 4e-04, 0.0189, P= 0.0419 for IP-10; and β: 0.0100, 95%CI: 0.0035, 0.0165, P= 0.0025 for M-CSF). Conversely, higher levels of interleukin-7 (IL-7), monocyte chemotactic protein 3 (MCP-3), and regulated on activation, normal T cell expressed and secreted (RANTES) were associated with decreased hand grip strength (β: -0.0071, 95%CI: -0.0127, -0.0014, P= 0.0140 for IL-7; β: -0.0064, 95%CI: -0.0123, -6e-04, P= 0.0313 for MCP-3; and β: -0.0082, 95%CI: -0.0164, -1e-04, P= 0.0480 for RANTES). Similarly, interleukin 1 receptor antagonist (IL-1RA) was negatively correlated with usual walking pace (β: -0.0104, 95%CI: -0.0195, -0.0013, P= 0.0254). Sensitivity analysis confirmed the robustness of these findings. Conclusions Our study provides additional insights into the pivotal role of specific inflammatory cytokines in the pathogenesis of sarcopenia. Further research is required to determine whether these cytokines can be used as targets for the prevention and treatment of sarcopenia.
Collapse
Affiliation(s)
- Zhi Chen
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jun Sun
- Department of Emergency, Zhaotong Traditional Chinese Medicine Hospital, Zhaotong, Yunnan, China
| | - Tengbin Shi
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chenyang Song
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chengjian Wu
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Zhengru Wu
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jiajun Lin
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
7
|
Feng F, Luo K, Yuan X, Lan T, Wang S, Xu X, Lu Z. Aerobic Exercise Protects against Cardiotoxin-Induced Skeletal Muscle Injury in a DDAH1-Dependent Manner. Antioxidants (Basel) 2024; 13:1069. [PMID: 39334728 PMCID: PMC11428882 DOI: 10.3390/antiox13091069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Dimethylarginine dimethylaminohydrolase 1 (DDAH1) is a critical enzyme that regulates nitric oxide (NO) signaling through the degradation of asymmetric dimethylarginine (ADMA). Previous studies have revealed a link between the beneficial effects of aerobic exercise and the upregulation of DDAH1 in bones and hearts. We previously reported that skeletal muscle DDAH1 plays a protective role in cardiotoxin (CTX)-induced skeletal muscle injury and regeneration. To determine the effects of aerobic exercise on CTX-induced skeletal muscle injury and the role of DDAH1 in this process, wild-type (WT) mice and skeletal muscle-specific Ddah1-knockout (Ddah1MKO) mice were subjected to swimming training for 8 weeks and then injected with CTX. In WT mice, swimming training for 8 weeks significantly promoted skeletal muscle regeneration and attenuated inflammation, oxidative stress, and apoptosis in the gastrocnemius (GA) muscle after CTX injection. These phenomena were associated with increases in the protein expression of PAX7, myogenin, MEF2A, eNOS, SOD2, and peroxiredoxin 5 and decreases in iNOS expression in GA muscles. Swimming training also decreased serum ADMA levels and increased serum nitrate/nitrite (NOx) levels and skeletal muscle DDAH1 expression. Interestingly, swimming training in Ddah1MKO mice had no obvious effect on CTX-induced skeletal muscle injury or regeneration and did not repress the CTX-induced inflammatory response, superoxide generation, or apoptosis. In summary, our data suggest that DDAH1 is important for the protective effect of aerobic exercise on skeletal muscle injury and regeneration.
Collapse
Affiliation(s)
- Fei Feng
- Sport and Health Science Department, Nanjing Sport Institute, Nanjing 210000, China
| | - Kai Luo
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyi Yuan
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ting Lan
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siyu Wang
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
8
|
Chen J, Jia S, Guo C, Fan Z, Yan W, Dong K. Research Progress on the Effect and Mechanism of Exercise Intervention on Sarcopenia Obesity. Clin Interv Aging 2024; 19:1407-1422. [PMID: 39139211 PMCID: PMC11319865 DOI: 10.2147/cia.s473083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
With the increasingly severe situation of obesity and population aging, there is growing concern about sarcopenia obesity (SO). SO refers to the coexistence of obesity and sarcopenia, which imposes a heavier burden on individuals and society compared to obesity or sarcopenia alone. Therefore, comprehending the pathogenesis of SO and implementing effective clinical interventions are vital for its prevention and treatment. This review uses a comprehensive literature search and analysis of PubMed, Web of Science, and CNKI databases, with search terms including "Sarcopenic obesity", "exercise", "cytokines", "inflammation", "mitochondrial quality control", and "microRNA", covering relevant studies published up to July 2024. The results indicate that the pathogenesis of SO is complex, involving mechanisms like age-related changes in body composition, hormonal alterations, inflammation, mitochondrial dysfunction, and genetic and epigenetic factors. Regarding exercise interventions for SO, aerobic exercise can reduce fat mass, resistance exercise can increase skeletal muscle mass and strength, and combined exercise can achieve both, making it the optimal intervention for SO. The potential mechanisms by which exercise may prevent and treat SO include regulating cytokine secretion, inhibiting inflammatory pathways, improving mitochondrial quality, and mediating microRNA expression. This review emphasizes the effectiveness of exercise interventions in mitigating sarcopenic obesity through comprehensive analysis of its multifactorial pathogenesis and the mechanistic insights into exercise's therapeutic effects. Understanding these mechanisms informs targeted therapeutic strategies aimed at alleviating the societal and individual burdens associated with SO.
Collapse
Affiliation(s)
- Jun Chen
- School of Graduate, Wuhan Sport University, Wuhan, 430079, People’s Republic of China
| | - Shaohui Jia
- School of Sports Medicine, Wuhan Sport University, Wuhan, 430079, People’s Republic of China
| | - Chenggen Guo
- School of Sports Training, Wuhan Sport University, Wuhan, 430079, People’s Republic of China
| | - Zhiwei Fan
- School of Graduate, Wuhan Sport University, Wuhan, 430079, People’s Republic of China
| | - Weiyi Yan
- School of Graduate, Wuhan Sport University, Wuhan, 430079, People’s Republic of China
| | - Kunwei Dong
- School of Arts, Wuhan Sport University, Wuhan, 430079, People’s Republic of China
| |
Collapse
|
9
|
Jin X, Yue X, Huang Z, Meng X, Xu S, Wu Y, Wan Y, Inoue A, Narisawa M, Hu L, Shi GP, Umegaki H, Murohara T, Lei Y, Kuzuya M, Cheng XW. Cathepsin K deficiency prevented stress-related thrombosis in a mouse FeCl 3 model. Cell Mol Life Sci 2024; 81:205. [PMID: 38703204 PMCID: PMC11069486 DOI: 10.1007/s00018-024-05240-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Exposure to chronic psychological stress (CPS) is a risk factor for thrombotic cardiocerebrovascular diseases (CCVDs). The expression and activity of the cysteine cathepsin K (CTSK) are upregulated in stressed cardiovascular tissues, and we investigated whether CTSK is involved in chronic stress-related thrombosis, focusing on stress serum-induced endothelial apoptosis. METHODS AND RESULTS Eight-week-old wild-type male mice (CTSK+/+) randomly divided to non-stress and 3-week restraint stress groups received a left carotid artery iron chloride3 (FeCl3)-induced thrombosis injury for biological and morphological evaluations at specific timepoints. On day 21 post-stress/injury, the stress had enhanced the arterial thrombi weights and lengths, in addition to harmful alterations of plasma ADAMTS13, von Willebrand factor, and plasminogen activation inhibitor-1, plus injured-artery endothelial loss and CTSK protein/mRNA expression. The stressed CTSK+/+ mice had increased levels of injured arterial cleaved Notch1, Hes1, cleaved caspase8, matrix metalloproteinase-9/-2, angiotensin type 1 receptor, galactin3, p16IN4A, p22phox, gp91phox, intracellular adhesion molecule-1, TNF-α, MCP-1, and TLR-4 proteins and/or genes. Pharmacological and genetic inhibitions of CTSK ameliorated the stress-induced thrombus formation and the observed molecular and morphological changes. In cultured HUVECs, CTSK overexpression and silencing respectively increased and mitigated stressed-serum- and H2O2-induced apoptosis associated with apoptosis-related protein changes. Recombinant human CTSK degraded γ-secretase substrate in a dose-dependent manor and activated Notch1 and Hes1 expression upregulation. CONCLUSIONS CTSK appeared to contribute to stress-related thrombosis in mice subjected to FeCl3 stress, possibly via the modulation of vascular inflammation, oxidative production and apoptosis, suggesting that CTSK could be an effective therapeutic target for CPS-related thrombotic events in patients with CCVDs.
Collapse
Affiliation(s)
- Xueying Jin
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Xueling Yue
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
| | - Zhe Huang
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Department of Neurology, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Xiangkun Meng
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Shengnan Xu
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Yuna Wu
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China
| | - Ying Wan
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Aiko Inoue
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University Institute of Innovation for Future Society, Nagoya University, Nagoya, Aichi-Ken, 466-8550, Japan
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Lina Hu
- Department of Public Health, Guilin Medical College, Guilin, 541199, Guangxi, People's Republic of China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Hiroyuki Umegaki
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University Institute of Innovation for Future Society, Nagoya University, Nagoya, Aichi-Ken, 466-8550, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Yanna Lei
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
- Department of Intensive Care, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
| | - Masafumi Kuzuya
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
- Meitetsu Hospital, Nagoya, Aichi, 451-8511, Japan
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
- Department of Community Health Care and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, Jilin, People's Republic of China.
| |
Collapse
|
10
|
Axelrod CL, Dantas WS, Kirwan JP. Sarcopenic obesity: emerging mechanisms and therapeutic potential. Metabolism 2023; 146:155639. [PMID: 37380015 PMCID: PMC11448314 DOI: 10.1016/j.metabol.2023.155639] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/08/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
Sarcopenic obesity, or the loss of muscle mass and function associated with excess adiposity, is a largely untreatable medical condition associated with diminished quality of life and increased risk of mortality. To date, it remains somewhat paradoxical and mechanistically undefined as to why a subset of adults with obesity develop muscular decline, an anabolic stimulus generally associated with retention of lean mass. Here, we review evidence surrounding the definition, etiology, and treatment of sarcopenic obesity with an emphasis on emerging regulatory nodes with therapeutic potential. We review the available clinical evidence largely focused on diet, lifestyle, and behavioral interventions to improve quality of life in patients with sarcopenic obesity. Based upon available evidence, relieving consequences of energy burden, such as oxidative stress, myosteatosis, and/or mitochondrial dysfunction, is a promising area for therapeutic development in the treatment and management of sarcopenic obesity.
Collapse
Affiliation(s)
- Christopher L Axelrod
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Wagner S Dantas
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - John P Kirwan
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| |
Collapse
|
11
|
Wan Y, Piao L, Xu S, Meng X, Huang Z, Inoue A, Wang H, Yue X, Jin X, Nan Y, Shi GP, Murohara T, Umegaki H, Kuzuya M, Cheng XW. Cathepsin S activity controls chronic stress-induced muscle atrophy and dysfunction in mice. Cell Mol Life Sci 2023; 80:254. [PMID: 37589754 PMCID: PMC10435624 DOI: 10.1007/s00018-023-04888-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/06/2023] [Accepted: 07/17/2023] [Indexed: 08/18/2023]
Abstract
Exposure to chronic psychological stress (CPS) is an intractable risk factor for inflammatory and metabolic diseases. Lysosomal cysteinyl cathepsins play an important role in human pathobiology. Given that cathepsin S (CTSS) is upregulated in the stressed vascular and adipose tissues, we investigated whether CTSS participates in chronic stress-induced skeletal muscle mass loss and dysfunction, with a special focus on muscle protein metabolic imbalance and apoptosis. Eight-week-old male wildtype (CTSS+/+) and CTSS-knockout (CTSS-/-) mice were randomly assigned to non-stress and variable-stress groups. CTSS+/+ stressed mice showed significant losses of muscle mass, dysfunction, and fiber area, plus significant mitochondrial damage. In this setting, stressed muscle in CTSS+/+ mice presented harmful alterations in the levels of insulin receptor substrate 2 protein content (IRS-2), phospho-phosphatidylinositol 3-kinase, phospho-protein kinase B, and phospho-mammalian target of rapamycin, forkhead box-1, muscle RING-finger protein-1 protein, mitochondrial biogenesis-related peroxisome proliferator-activated receptor-γ coactivator-α, and apoptosis-related B-cell lymphoma 2 and cleaved caspase-3; these alterations were prevented by CTSS deletion. Pharmacological CTSS inhibition mimics its genetic deficiency-mediated muscle benefits. In C2C12 cells, CTSS silencing prevented stressed serum- and oxidative stress-induced IRS-2 protein reduction, loss of the myotube myosin heavy chain content, and apoptosis accompanied by a rectification of investigated molecular harmful changes; these changes were accelerated by CTSS overexpression. These findings demonstrated that CTSS plays a role in IRS-2-related protein anabolism and catabolism and cell apoptosis in stress-induced muscle wasting, suggesting a novel therapeutic strategy for the control of chronic stress-related muscle disease in mice under our experimental conditions by regulating CTSS activity.
Collapse
Affiliation(s)
- Ying Wan
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Limei Piao
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
| | - Shengnan Xu
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Xiangkun Meng
- Department of Vascular Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Zhe Huang
- Department of Neurology, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Aiko Inoue
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, 4660855, Japan
| | - Hailong Wang
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Xueling Yue
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Xueying Jin
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Yongshan Nan
- Department of Anesthesiology, Yanbian University Hospital, 1327 Juzijie, Yanji, 133000, Jilin, People's Republic of China.
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
| | - Hiroyuki Umegaki
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, 4660855, Japan
- Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
| | - Masafumi Kuzuya
- Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
- Meitetsu Hospital, Nagoya, Aichi, 451-8511, Japan
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
- Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
| |
Collapse
|
12
|
Lu W, Feng W, Lai J, Yuan D, Xiao W, Li Y. Role of adipokines in sarcopenia. Chin Med J (Engl) 2023; 136:1794-1804. [PMID: 37442757 PMCID: PMC10406092 DOI: 10.1097/cm9.0000000000002255] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Indexed: 07/15/2023] Open
Abstract
ABSTRACT Sarcopenia is an age-related disease that mainly involves decreases in muscle mass, muscle strength and muscle function. At the same time, the body fat content increases with aging, especially the visceral fat content. Adipose tissue is an endocrine organ that secretes biologically active factors called adipokines, which act on local and distant tissues. Studies have revealed that some adipokines exert regulatory effects on muscle, such as higher serum leptin levels causing a decrease in muscle function and adiponectin inhibits the transcriptional activity of Forkhead box O3 (FoxO3) by activating peroxisome proliferators-activated receptor-γ coactivator -1α (PGC-1α) and sensitizing cells to insulin, thereby repressing atrophy-related genes (atrogin-1 and muscle RING finger 1 [MuRF1]) to prevent the loss of muscle mass. Here, we describe the effects on muscle of adipokines produced by adipose tissue, such as leptin, adiponectin, resistin, mucin and lipocalin-2, and discuss the importance of these adipokines for understanding the development of sarcopenia.
Collapse
Affiliation(s)
- Wenhao Lu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Wenjie Feng
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jieyu Lai
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Dongliang Yuan
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Wenfeng Xiao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yusheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
13
|
Ozaki Y, Ohashi K, Otaka N, Kawanishi H, Takikawa T, Fang L, Takahara K, Tatsumi M, Ishihama S, Takefuji M, Kato K, Shimizu Y, Bando YK, Inoue A, Kuzuya M, Miura S, Murohara T, Ouchi N. Myonectin protects against skeletal muscle dysfunction in male mice through activation of AMPK/PGC1α pathway. Nat Commun 2023; 14:4675. [PMID: 37542026 PMCID: PMC10403505 DOI: 10.1038/s41467-023-40435-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 07/28/2023] [Indexed: 08/06/2023] Open
Abstract
To maintain and restore skeletal muscle mass and function is essential for healthy aging. We have found that myonectin acts as a cardioprotective myokine. Here, we investigate the effect of myonectin on skeletal muscle atrophy in various male mouse models of muscle dysfunction. Disruption of myonectin exacerbates skeletal muscle atrophy in age-associated, sciatic denervation-induced or dexamethasone (DEX)-induced muscle atrophy models. Myonectin deficiency also contributes to exacerbated mitochondrial dysfunction and reduces expression of mitochondrial biogenesis-associated genes including PGC1α in denervated muscle. Myonectin supplementation attenuates denervation-induced muscle atrophy via activation of AMPK. Myonectin also reverses DEX-induced atrophy of cultured myotubes through the AMPK/PGC1α signaling. Furthermore, myonectin treatment suppresses muscle atrophy in senescence-accelerated mouse prone (SAMP) 8 mouse model of accelerated aging or mdx mouse model of Duchenne muscular dystrophy. These data indicate that myonectin can ameliorate skeletal muscle dysfunction through AMPK/PGC1α-dependent mechanisms, suggesting that myonectin could represent a therapeutic target of muscle atrophy.
Collapse
Affiliation(s)
- Yuta Ozaki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koji Ohashi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Naoya Otaka
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Kawanishi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomonobu Takikawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Lixin Fang
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kunihiko Takahara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Minako Tatsumi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sohta Ishihama
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikito Takefuji
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Katsuhiro Kato
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuko K Bando
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Aiko Inoue
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masafumi Kuzuya
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinji Miura
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Noriyuki Ouchi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
14
|
Soh S, Han S, Ka HI, Mun SH, Kim W, Oh G, Yang Y. Adiponectin affects the migration ability of bone marrow-derived mesenchymal stem cells via the regulation of hypoxia inducible factor 1α. Cell Commun Signal 2023; 21:158. [PMID: 37370133 PMCID: PMC10294307 DOI: 10.1186/s12964-023-01143-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/22/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Bone marrow (BM) is progressively filled with adipocytes during aging process. Thus, BM adipocytes-derived adiponectin (APN) affects the function of bone marrow-derived mesenchymal stem cells (BMSCs). However, little is known about the effect of APN on migration ability of BMSCs cultured under hypoxic conditions, which is similar to the BM microenvironment. RESULTS We found that the population and migration ability of BMSCs from APN KO mice was higher than that of WT mice due to increased stability of hypoxia inducible factor 1α (HIF1α). Stem cell factor (SCF)-activated STAT3 stimulated the induction of HIF1α which further stimulated SCF production, indicating that the SCF/STAT3/HIF1α positive loop was highly activated in the absence of APN. It implies that APN negatively regulated this positive loop by stimulating HIF1α degradation via the inactivation of GSK3β. Furthermore, APN KO BMSCs were highly migratory toward EL-4 lymphoma, and the interaction between CD44 in BMSCs and hyaluronic acid (HA) from EL-4 enhanced the migration of BMSCs. On the other hand, the migrated BMSCs recruited CD8+ T cells into the EL-4 tumor tissue, resulting in the retardation of tumor growth. Additionally, gradually increased APN in BM on the aging process affects migration and related functions of BMSCs, thus aged APN KO mice showed more significant suppression of EL-4 growth than young APN KO mice due to higher migration and recruitment of CD8+ T cells. CONCLUSION APN deficiency enhances CD44-mediated migration ability of BMSCs in the hypoxic conditions by the SCF/STAT3/HIF1α positive loop and influences the migration ability of BMSCs for a longer time depending on the aging process. Video Abstract.
Collapse
Affiliation(s)
- Sujung Soh
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Sora Han
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Hye In Ka
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Se Hwan Mun
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Woojung Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Gaeun Oh
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Young Yang
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
15
|
Liu C, Wu X, Vulugundam G, Gokulnath P, Li G, Xiao J. Exercise Promotes Tissue Regeneration: Mechanisms Involved and Therapeutic Scope. SPORTS MEDICINE - OPEN 2023; 9:27. [PMID: 37149504 PMCID: PMC10164224 DOI: 10.1186/s40798-023-00573-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 04/23/2023] [Indexed: 05/08/2023]
Abstract
Exercise has well-recognized beneficial effects on the whole body. Previous studies suggest that exercise could promote tissue regeneration and repair in various organs. In this review, we have summarized the major effects of exercise on tissue regeneration primarily mediated by stem cells and progenitor cells in skeletal muscle, nervous system, and vascular system. The protective function of exercise-induced stem cell activation under pathological conditions and aging in different organs have also been discussed in detail. Moreover, we have described the primary molecular mechanisms involved in exercise-induced tissue regeneration, including the roles of growth factors, signaling pathways, oxidative stress, metabolic factors, and non-coding RNAs. We have also summarized therapeutic approaches that target crucial signaling pathways and molecules responsible for exercise-induced tissue regeneration, such as IGF1, PI3K, and microRNAs. Collectively, the comprehensive understanding of exercise-induced tissue regeneration will facilitate the discovery of novel drug targets and therapeutic strategies.
Collapse
Affiliation(s)
- Chang Liu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Xinying Wu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| | | | - Priyanka Gokulnath
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China.
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
16
|
Zhang Y, Wang L, Kang H, Lin CY, Fan Y. Unlocking the Therapeutic Potential of Irisin: Harnessing Its Function in Degenerative Disorders and Tissue Regeneration. Int J Mol Sci 2023; 24:ijms24076551. [PMID: 37047523 PMCID: PMC10095399 DOI: 10.3390/ijms24076551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Physical activity is well-established as an important protective factor against degenerative conditions and a promoter of tissue growth and renewal. The discovery of Fibronectin domain-containing protein 5 (FNDC5) as the precursor of Irisin in 2012 sparked significant interest in its potential as a diagnostic biomarker and a therapeutic agent for various diseases. Clinical studies have examined the correlation between plasma Irisin levels and pathological conditions using a range of assays, but the lack of reliable measurements for endogenous Irisin has led to uncertainty about its prognostic/diagnostic potential as an exercise surrogate. Animal and tissue-engineering models have shown the protective effects of Irisin treatment in reversing functional impairment and potentially permanent damage, but dosage ambiguities remain unresolved. This review provides a comprehensive examination of the clinical and basic studies of Irisin in the context of degenerative conditions and explores its potential as a therapeutic approach in the physiological processes involved in tissue repair/regeneration.
Collapse
Affiliation(s)
- Yuwei Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Lizhen Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Correspondence:
| | - Hongyan Kang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Chia-Ying Lin
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Department of Biomedical, Chemical & Environmental Engineering, University of Cincinnati, Cincinnati, OH 45267, USA
- Department of Orthopaedic Surgery, University of Cincinnati, Cincinnati, OH 45267, USA
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- School of Engineering Medicine, Beihang University, Beijing 100083, China
| |
Collapse
|
17
|
Liu X, Yang Y, Shao H, Liu S, Niu Y, Fu L. Globular adiponectin ameliorates insulin resistance in skeletal muscle by enhancing the LKB1-mediated AMPK activation via SESN2. SPORTS MEDICINE AND HEALTH SCIENCE 2023; 5:34-41. [PMID: 36994173 PMCID: PMC10040333 DOI: 10.1016/j.smhs.2022.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/13/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Adiponectin has been demonstrated to be a mediator of insulin sensitivity; however, the underlined mechanisms remain unclear. SESN2 is a stress-inducible protein that phosphorylates AMPK in different tissues. In this study, we aimed to validate the amelioration of insulin resistance by globular adiponectin (gAd) and to reveal the role of SESN2 in the improvement of glucose metabolism by gAd. We used a high-fat diet-induced wild-type and SESN2-/- C57BL/6J insulin resistance mice model to study the effects of six-week aerobic exercise or gAd administration on insulin resistance. In vitro study, C2C12 myotubes were used to determine the potential mechanism by overexpressing or inhibiting SESN2. Similar to exercise, six-week gAd administration decreased fasting glucose, triglyceride and insulin levels, reduced lipid deposition in skeletal muscle and reversed whole-body insulin resistance in mice fed on a high-fat diet. Moreover, gAd enhanced skeletal muscle glucose uptake by activating insulin signaling. However, these effects were diminished in SESN2-/- mice. We found that gAd administration increased the expression of SESN2 and Liver kinase B1 (LKB1) and increased AMPK-T172 phosphorylation in skeletal muscle of wild-type mice, while in SESN2-/- mice, LKB1 expression was also increased but the pAMPK-T172 was unchanged. At the cellular level, gAd increased cellular SESN2 and pAMPK-T172 expression. Immunoprecipitation experiment suggested that SESN2 promoted the formation of complexes of AMPK and LKB1 and hence phosphorylated AMPK. In conclusion, our results revealed that SESN2 played a critical role in gAd-induced AMPK phosphorylation, activation of insulin signaling and skeletal muscle insulin sensitization in mice with insulin resistance.
Collapse
Affiliation(s)
- Xinmeng Liu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| | - Yang Yang
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| | - Heng Shao
- Department of Anatomy and Histology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Sujuan Liu
- Department of Anatomy and Histology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Yanmei Niu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| | - Li Fu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
18
|
Selvais CM, Davis-López de Carrizosa MA, Nachit M, Versele R, Dubuisson N, Noel L, Gillard J, Leclercq IA, Brichard SM, Abou-Samra M. AdipoRon enhances healthspan in middle-aged obese mice: striking alleviation of myosteatosis and muscle degenerative markers. J Cachexia Sarcopenia Muscle 2023; 14:464-478. [PMID: 36513619 PMCID: PMC9891981 DOI: 10.1002/jcsm.13148] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/16/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Obesity among older adults has increased tremendously. Obesity accelerates ageing and predisposes to age-related conditions and diseases, such as loss of endurance capacity, insulin resistance and features of the metabolic syndrome. Namely, ectopic lipids play a key role in the development of nonalcoholic fatty liver disease (NAFLD) and myosteatosis, two severe burdens of ageing and metabolic diseases. Adiponectin (ApN) is a hormone, mainly secreted by adipocytes, which exerts insulin-sensitizing and fat-burning properties in several tissues including the liver and the muscle. Its overexpression also increases lifespan in mice. In this study, we investigated whether an ApN receptor agonist, AdipoRon (AR), could slow muscle dysfunction, myosteatosis and degenerative muscle markers in middle-aged obese mice. The effects on myosteatosis were compared with those on NAFLD. METHODS Three groups of mice were studied up to 62 weeks of age: One group received normal diet (ND), another, high-fat diet (HFD); and the last, HFD combined with AR given orally for almost 1 year. An additional group of young mice under an ND was used. Treadmill tests and micro-computed tomography (CT) were carried out in vivo. Histological, biochemical and molecular analyses were performed on tissues ex vivo. Bodipy staining was used to assess intramyocellular lipid (IMCL) and lipid droplet morphology. RESULTS AR did not markedly alter diet-induced obesity. Yet, this treatment rescued exercise endurance in obese mice (up to 2.4-fold, P < 0.05), an event that preceded the improvement of insulin sensitivity. Dorsal muscles and liver densities, measured by CT, were reduced in obese mice (-42% and -109%, respectively, P < 0.0001), suggesting fatty infiltration. This reduction tended to be attenuated by AR. Accordingly, AR significantly mitigated steatosis and cellular ballooning at liver histology, thereby decreasing the NALFD activity score (-30%, P < 0.05). AR also strikingly reversed IMCL accumulation either due to ageing in oxidative fibres (types 1/2a, soleus) or to HFD in glycolytic ones (types 2x/2b, extensor digitorum longus) (-50% to -85%, P < 0.05 or less). Size of subsarcolemmal lipid droplets, known to be associated with adverse metabolic outcomes, was reduced as well. Alleviation of myosteatosis resulted from improved mitochondrial function and lipid oxidation. Meanwhile, AR halved aged-related accumulation of dysfunctional proteins identified as tubular aggregates and cylindrical spirals by electron microscopy (P < 0.05). CONCLUSIONS Long-term AdipoRon treatment promotes 'healthy ageing' in obese middle-aged mice by enhancing endurance and protecting skeletal muscle and liver against the adverse metabolic and degenerative effects of ageing and caloric excess.
Collapse
Affiliation(s)
- Camille M Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - María A Davis-López de Carrizosa
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium.,Department of Physiology, Faculty of Biology, University of Seville, Seville, Spain
| | - Maxime Nachit
- Hepato-Gastroenterology Unit, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Romain Versele
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Nicolas Dubuisson
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Justine Gillard
- Hepato-Gastroenterology Unit, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Isabelle A Leclercq
- Hepato-Gastroenterology Unit, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Sonia M Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| |
Collapse
|
19
|
Ladang A, Beaudart C, Reginster JY, Al-Daghri N, Bruyère O, Burlet N, Cesari M, Cherubini A, da Silva MC, Cooper C, Cruz-Jentoft AJ, Landi F, Laslop A, Maggi S, Mobasheri A, Ormarsdottir S, Radermecker R, Visser M, Yerro MCP, Rizzoli R, Cavalier E. Biochemical Markers of Musculoskeletal Health and Aging to be Assessed in Clinical Trials of Drugs Aiming at the Treatment of Sarcopenia: Consensus Paper from an Expert Group Meeting Organized by the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO) and the Centre Académique de Recherche et d'Expérimentation en Santé (CARES SPRL), Under the Auspices of the World Health Organization Collaborating Center for the Epidemiology of Musculoskeletal Conditions and Aging. Calcif Tissue Int 2023; 112:197-217. [PMID: 36633611 PMCID: PMC9859913 DOI: 10.1007/s00223-022-01054-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/18/2022] [Indexed: 01/13/2023]
Abstract
In clinical trials, biochemical markers provide useful information on the drug's mode of action, therapeutic response and side effect monitoring and can act as surrogate endpoints. In pharmacological intervention development for sarcopenia management, there is an urgent need to identify biomarkers to measure in clinical trials and that could be used in the future in clinical practice. The objective of the current consensus paper is to provide a clear list of biochemical markers of musculoskeletal health and aging that can be recommended to be measured in Phase II and Phase III clinical trials evaluating new chemical entities for sarcopenia treatment. A working group of the European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO) proposed classifying biochemical markers into 2 series: biochemical markers evaluating musculoskeletal status and biochemical markers evaluating causal factors. For series 1, the group agreed on 4 biochemical markers that should be assessed in Phase II or Phase III trials (i.e., Myostatin-Follistatin, Brain Derived Neurotrophic Factor, N-terminal Type III Procollagen and Serum Creatinine to Serum Cystatin C Ratio - or the Sarcopenia Index). For series 2, the group agreed on 6 biochemical markers that should be assessed in Phase II trials (i.e., the hormones insulin-like growth factor-1 (IGF-I), dehydroepiandrosterone sulphate, and cortisol, and the inflammatory markers C-reactive protein (CRP), interleukin-6 and tumor necrosis factor-α), and 2 in Phase III trials (i.e., IGF-I and CRP). The group also proposed optional biochemical markers that may provide insights into the mode of action of pharmacological therapies. Further research and development of new methods for biochemical marker assays may lead to the evolution of these recommendations.
Collapse
Affiliation(s)
- Aurélie Ladang
- Department of Clinical Chemistry, CHU de Liège, University of Liège, Liège, Belgium.
| | - Charlotte Beaudart
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
| | - Jean-Yves Reginster
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
- Biochemistry Department, College of Science, Chair for Biomarkers of Chronic Diseases, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Nasser Al-Daghri
- Biochemistry Department, College of Science, Chair for Biomarkers of Chronic Diseases, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Olivier Bruyère
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
| | - Nansa Burlet
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
| | - Matteo Cesari
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonio Cherubini
- Geriatric Unit, IRCCS Istituti Clinici Scientifici Maugeri, Milan, Italy
| | | | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | | | - Francesco Landi
- Department of Geriatrics, Neurosciences and Orthopedics, Catholic University of the Sacred Heart, Rome, Italy
| | - Andrea Laslop
- Scientific Office, Federal Office for Safety in Health Care, Vienna, Austria
| | | | - Ali Mobasheri
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Center for Public Health Aspects of Musculo-Skeletal Health and Ageing,, University of Liège, Liège, Belgium
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | | | - Régis Radermecker
- Department of Diabetes, Nutrition and Metabolic Disorders, Clinical Pharmacology, University of Liege, CHU de Liège, Liège, Belgium
| | - Marjolein Visser
- Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - René Rizzoli
- Faculty of Medicine, Service of Bone Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Etienne Cavalier
- Department of Clinical Chemistry, CHU de Liège, University of Liège, Liège, Belgium
| |
Collapse
|
20
|
Yakabe M, Hosoi T, Sasakawa H, Akishita M, Ogawa S. Kampo formula hochu-ekki-to (Bu-Zhong-Yi-Qi-Tang, TJ-41) ameliorates muscle atrophy by modulating atrogenes and AMPK in vivo and in vitro. BMC Complement Med Ther 2022; 22:341. [PMID: 36578084 PMCID: PMC9795672 DOI: 10.1186/s12906-022-03812-w] [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: 12/06/2020] [Accepted: 11/28/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Muscle disuse results in loss of skeletal muscle mass and function. Hochu-ekki-to (TJ-41; Bu-Zhong-Yi-Qi-Tang in Chinese) is an herbal medicinal formulation used to treat patients with frailty, fatigue and appetite loss. It has been suggested that two atrogenes, atrogin-1 and muscle Ring finger 1 (MuRF1), are ubiquitin ligases involved in disuse-induced muscle atrophy and that 5' adenosine monophosphate-activated protein kinase (AMPK) is involved in skeletal muscle metabolism. Effects of TJ-41 on disuse-induced muscle atrophy are unclear. METHODS We subjected differentiated C2C12 myotubes to serum starvation, then examined the effects of TJ-41 on atrogenes expression, AMPK activity and the morphology of the myotubes. Male C57BL/6J mice were subjected to tail-suspension to induce hindlimb atrophy. We administered TJ-41 by gavage to the control group and the tail-suspended group, then examined the effects of TJ-41 on atrogene expression, AMPK activity, and the muscle weight. RESULTS Serum starvation induced the expression of atrogin-1 and MuRF1 in C2C12 myotubes, and TJ-41 significantly downregulated the expression of atrogin-1. Tail-suspension of the mice induced the expression of atrogin-1 and MuRF1 in skeletal muscle as well as its muscle atrophy, whereas TJ-41 treatment significantly downregulated the expression of atrogin-1 and ameliorated the loss of the muscle weight. In addition, TJ-41 also activated AMPK and inactivated Akt and mTOR in skeletal muscle in vivo. CONCLUSION TJ-41 inhibited atrogenes in an Akt-independent manner as well as activating AMPK in skeletal muscles in vivo, further implying the therapeutic potential of TJ-41 against disuse-induced muscle atrophy and other atrogenes-dependent atrophic conditions.
Collapse
Affiliation(s)
- Mitsutaka Yakabe
- grid.26999.3d0000 0001 2151 536XDepartment of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7- 3-1, Hongo, Bunkyo-ku, 113-8655 Tokyo, Japan
| | - Tatsuya Hosoi
- grid.26999.3d0000 0001 2151 536XDepartment of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7- 3-1, Hongo, Bunkyo-ku, 113-8655 Tokyo, Japan
| | - Hiroko Sasakawa
- grid.26999.3d0000 0001 2151 536XDepartment of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7- 3-1, Hongo, Bunkyo-ku, 113-8655 Tokyo, Japan
| | - Masahiro Akishita
- grid.26999.3d0000 0001 2151 536XDepartment of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7- 3-1, Hongo, Bunkyo-ku, 113-8655 Tokyo, Japan
| | - Sumito Ogawa
- grid.26999.3d0000 0001 2151 536XDepartment of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, 7- 3-1, Hongo, Bunkyo-ku, 113-8655 Tokyo, Japan
| |
Collapse
|
21
|
Liu S, Liu H, Liu Y, Zhang J, Liu Z, Zheng Z, Luo E. Adiponectin receptors activation performs dual effects on regulating myogenesis and adipogenesis of young and aged muscle satellite cells. Cell Prolif 2022; 56:e13370. [PMID: 36484401 PMCID: PMC9977665 DOI: 10.1111/cpr.13370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Skeletal muscle mass and function deteriorate with ageing. Adiponectin receptors (APNrs), mainly activated by adiponectin, participate in various physiological activities and have varying signalling pathways at different ages. This study aimed to explore whether discrepant performance exists in APNr activation regulating young and aged muscle satellite cells (MUSCs) and whether age-related muscle dysfunction could be alleviated upon APNr activation. METHODS The gastrocnemius muscle phenotype was observed in male mice aged 2 and 18 months. An APNr agonist (AdipoRon) was used in vitro and in vivo to investigate the changes in cell biological behaviours and whether muscle dysfunction could be retarded after APNr activation. RESULTS Aged mice exhibited decreased muscle mass and increased fat infiltration. APNr activation inhibited C2C12 cells and young MUSCs (YMUSCs) proliferation but showed no obvious effect on aged MUSCs (AMUSCs). Moreover, APNr activation inhibited the migration of both YMUSCs and AMUSCs. Interestingly, APNr activation hampered the myogenic differentiation but advanced the adipogenic differentiation of YMUSCs, yet exact opposite results were presented in AMUSCs. It was demonstrated that Wnt and PI3K signalling pathways may mediate the phenotypic differences. Furthermore, in vivo experiments verified that APNr activation ameliorated age-related muscle atrophy and excessive fat infiltration. CONCLUSIONS APNr activation exerted dual effects on the regulation of myogenesis and adipogenesis of YMUSCs and AMUSCs and rescued age-related skeletal muscle dysfunction.
Collapse
Affiliation(s)
- Shibo Liu
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral DiseasesWest China Hospital of Stomatology, Sichuan UniversityChengduChina,Department of Oral Maxillofacial SurgeryWest China Hospital of Stomatology, Sichuan UniversityChengduChina
| | - Hanghang Liu
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral DiseasesWest China Hospital of Stomatology, Sichuan UniversityChengduChina,Emergency DepartmentWest China Hospital of Stomatology, Sichuan UniversityChengduChina,Maine Medical Center Research InstituteScarboroughMaineUSA
| | - Yao Liu
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral DiseasesWest China Hospital of Stomatology, Sichuan UniversityChengduChina,Department of Oral Maxillofacial SurgeryWest China Hospital of Stomatology, Sichuan UniversityChengduChina
| | - Ju Zhang
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral DiseasesWest China Hospital of Stomatology, Sichuan UniversityChengduChina,Department of Oral Maxillofacial SurgeryWest China Hospital of Stomatology, Sichuan UniversityChengduChina
| | - Zhikai Liu
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral DiseasesWest China Hospital of Stomatology, Sichuan UniversityChengduChina,Department of Oral Maxillofacial SurgeryWest China Hospital of Stomatology, Sichuan UniversityChengduChina
| | - Zizhuo Zheng
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral DiseasesWest China Hospital of Stomatology, Sichuan UniversityChengduChina,Department of Oral Maxillofacial SurgeryWest China Hospital of Stomatology, Sichuan UniversityChengduChina
| | - En Luo
- State Key Laboratory of Oral Disease, National Clinical Research Center for Oral DiseasesWest China Hospital of Stomatology, Sichuan UniversityChengduChina,Department of Oral Maxillofacial SurgeryWest China Hospital of Stomatology, Sichuan UniversityChengduChina
| |
Collapse
|
22
|
Mariné-Casadó R, Domenech-Coca C, Crescenti A, Rodríguez Gómez MÁ, Del Bas JM, Arola L, Boqué N, Caimari A. Maternal Supplementation with a Cocoa Extract during Lactation Deeply Modulates Dams' Metabolism, Increases Adiponectin Circulating Levels and Improves the Inflammatory Profile in Obese Rat Offspring. Nutrients 2022; 14:nu14235134. [PMID: 36501173 PMCID: PMC9738144 DOI: 10.3390/nu14235134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/07/2022] Open
Abstract
High-flavonoid cocoa consumption has been associated with beneficial properties. However, there are scarce data concerning the effects of maternal cocoa intake on dams and in their progeny. Here, we evaluated in rats whether maternal supplementation with a high-flavan-3-ol cocoa extract (CCX) during lactation (200 mg.kg-1.day-1) produced beneficial effects on dams and in their normoweight (STD-CCX group) and cafeteria-fed obese (CAF-CCX group) adult male offspring. Maternal intake of CCX significantly increased the circulating levels of adiponectin and decreased the mammary gland lipid content of dams. These effects were accompanied by increased energy expenditure and circulating free fatty acids, as well as by a higher expression of lipogenic and adiponectin-related genes in their mammary glands, which could be related to a compensatory mechanism to ensure enough lipid supply to the pups. CCX consumption programmed both offspring groups towards increased plasma total adiponectin levels, and decreased liver weight and lean/fat ratio. Furthermore, CAF-CCX progeny showed an improvement of the inflammatory profile, evidenced by the significant decrease of the monocyte chemoattractant protein-1 (MCP-1) circulating levels and the mRNA levels of the gene encoding the major histocompatibility complex, class II invariant chain (Cd74), a marker of M1 macrophage phenotype, in the epididymal white adipose tissue. Although further studies are needed, these findings can pave the way for using CCX as a nutraceutical supplement during lactation.
Collapse
Affiliation(s)
- Roger Mariné-Casadó
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, 43204 Reus, Spain
| | - Cristina Domenech-Coca
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, 43204 Reus, Spain
| | - Anna Crescenti
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, 43204 Reus, Spain
| | - Miguel Ángel Rodríguez Gómez
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT, 43204 Reus, Spain
| | - Josep Maria Del Bas
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Reus, Spain
| | - Lluís Arola
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | - Noemí Boqué
- Eurecat, Centre Tecnològic de Catalunya, Technological Unit of Nutrition and Health, 43204 Reus, Spain
- Correspondence: (N.B.); (A.C.)
| | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Reus, Spain
- Correspondence: (N.B.); (A.C.)
| |
Collapse
|
23
|
Inoue A, Piao L, Yue X, Huang Z, Hu L, Wu H, Meng X, Xu W, Yu C, Sasaki T, Itakura K, Umegaki H, Kuzuya M, Cheng XW. Young bone marrow transplantation prevents aging-related muscle atrophy in a senescence-accelerated mouse prone 10 model. J Cachexia Sarcopenia Muscle 2022; 13:3078-3090. [PMID: 36058630 PMCID: PMC9745469 DOI: 10.1002/jcsm.13058] [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: 01/08/2022] [Revised: 06/22/2022] [Accepted: 07/04/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Young bone marrow transplantation (YBMT) has been shown to stimulate vascular regeneration in pathological conditions, including ageing. Here, we investigated the benefits and mechanisms of the preventive effects of YBMT on loss of muscle mass and function in a senescence-associated mouse prone 10 (SAMP10) model, with a special focus on the role of growth differentiation factor 11 (GDF-11). METHODS Nine-week-old male SAMP10 mice were randomly assigned to a non-YBMT group (n = 6) and a YBMT group (n = 7) that received the bone marrow of 8-week-old C57BL/6 mice. RESULTS Compared to the non-YBMT mice, the YBMT mice showed the following significant increases (all P < 0.05 in 6-7 mice): endurance capacity (>61.3%); grip strength (>37.9%), percentage of slow myosin heavy chain fibres (>14.9-15.9%). The YBMT also increased the amounts of proteins or mRNAs for insulin receptor substrate 1, p-Akt, p-extracellular signal-regulated protein kinase1/2, p-mammalian target of rapamycin, Bcl-2, peroxisom proliferator-activated receptor-γ coactivator (PGC-1α), plus cytochrome c oxidase IV and the numbers of proliferating cells (n = 5-7, P < 0.05) and CD34+/integrin-α7+ muscle stem cells (n = 5-6, P < 0.05). The YMBT significantly decreased the levels of gp91phox, caspase-9 proteins and apoptotic cells (n = 5-7, P < 0.05) in both muscles; these beneficial changes were diminished by the blocking of GDF-11 (n = 5-6, P < 0.05). An administration of mouse recombinant GDF-11 improved the YBMT-mediated muscle benefits (n = 5-6, P < 0.05). Cell therapy with young bone marrow from green fluorescent protein (GFP) transgenic mice exhibited GFP+ myofibres in aged muscle tissues. CONCLUSIONS These findings suggest that YBMT can prevent muscle wasting and dysfunction by mitigating apoptosis and proliferation via a modulation of GDF-11 signalling and mitochondrial dysfunction in SAMP10 mice.
Collapse
Affiliation(s)
- Aiko Inoue
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, Japan.,Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, Japan
| | - Limei Piao
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, PR China
| | - Xueling Yue
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, PR China
| | - Zhe Huang
- Department of Human Cord Applied Cell Therapy, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, Japan
| | - Lina Hu
- Department of Public Health, Guilin Medical College, Guilin, Guangxi, PR China
| | - Hongxian Wu
- Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Xiangkun Meng
- Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, Japan
| | - Wenhu Xu
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, PR China
| | - Chenglin Yu
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, PR China
| | - Takeshi Sasaki
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine, Hamamatsu, Shizuokaken, Japan
| | - Kohji Itakura
- Division for Medical Research Engineering, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, Japan
| | - Hiroyuki Umegaki
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, Japan.,Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, Japan
| | - Masafumi Kuzuya
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, Japan.,Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichiken, Japan
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular Disease, Yanbian University Hospital, Yanji, Jilin, PR China
| |
Collapse
|
24
|
Molecular mechanisms of exercise contributing to tissue regeneration. Signal Transduct Target Ther 2022; 7:383. [PMID: 36446784 PMCID: PMC9709153 DOI: 10.1038/s41392-022-01233-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/03/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2022] Open
Abstract
Physical activity has been known as an essential element to promote human health for centuries. Thus, exercise intervention is encouraged to battle against sedentary lifestyle. Recent rapid advances in molecular biotechnology have demonstrated that both endurance and resistance exercise training, two traditional types of exercise, trigger a series of physiological responses, unraveling the mechanisms of exercise regulating on the human body. Therefore, exercise has been expected as a candidate approach of alleviating a wide range of diseases, such as metabolic diseases, neurodegenerative disorders, tumors, and cardiovascular diseases. In particular, the capacity of exercise to promote tissue regeneration has attracted the attention of many researchers in recent decades. Since most adult human organs have a weak regenerative capacity, it is currently a key challenge in regenerative medicine to improve the efficiency of tissue regeneration. As research progresses, exercise-induced tissue regeneration seems to provide a novel approach for fighting against injury or senescence, establishing strong theoretical basis for more and more "exercise mimetics." These drugs are acting as the pharmaceutical alternatives of those individuals who cannot experience the benefits of exercise. Here, we comprehensively provide a description of the benefits of exercise on tissue regeneration in diverse organs, mainly focusing on musculoskeletal system, cardiovascular system, and nervous system. We also discuss the underlying molecular mechanisms associated with the regenerative effects of exercise and emerging therapeutic exercise mimetics for regeneration, as well as the associated opportunities and challenges. We aim to describe an integrated perspective on the current advances of distinct physiological mechanisms associated with exercise-induced tissue regeneration on various organs and facilitate the development of drugs that mimics the benefits of exercise.
Collapse
|
25
|
Liang Z, Zhang T, Liu H, Li Z, Peng L, Wang C, Wang T. Inflammaging: The ground for sarcopenia? Exp Gerontol 2022; 168:111931. [PMID: 35985553 DOI: 10.1016/j.exger.2022.111931] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/02/2022] [Accepted: 08/14/2022] [Indexed: 12/15/2022]
Abstract
Sarcopenia is a progressive skeletal muscle disease that occurs most commonly in the elderly population, contributing to increased costs and hospitalization. Exercise and nutritional therapy have been proven to be effective for sarcopenia, and some drugs can also alleviate declines in muscle mass and function due to sarcopenia. However, there is no specific pharmacological treatment for sarcopenia at present. This review will mainly discuss the relationship between inflammaging and sarcopenia. The increased secretion of proinflammatory cytokines with aging may be because of cellular senescence, immunosenescence, alterations in adipose tissue, damage-associated molecular patterns (DAMPs), and gut microbes due to aging. These sources of inflammaging can impact the sarcopenia process through direct or indirect pathways. Conversely, sarcopenia can also aggravate the process of inflammaging, creating a vicious cycle. Targeting sources of inflammaging can influence muscle function, which could be considered a therapeutic target for sarcopenia. Moreover, not only proinflammatory cytokines but also anti-inflammatory cytokines can influence muscle and inflammation and participate in the progression of sarcopenia. This review focuses on the effects of TNF-α, IL-6, and IL-10, which can be detected in plasma. Therefore, clearing chronic inflammation by targeting proinflammatory cytokines (TNF-α, IL-1, IL-6) and the inflammatory pathway (JAK/STAT, autophagy, NF-κB) may be effective in treating sarcopenia.
Collapse
Affiliation(s)
- Zejun Liang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Tianxiao Zhang
- School of Healthcare Sciences, Cardiff University, Health Park, CF14 4XN Wales, UK
| | - Honghong Liu
- West China School of Nursing/West China Hospital, Sichuan University, NO.37 Alley, Chengdu 610041, Sichuan, PR China
| | - Zhenlin Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lihong Peng
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, PR China
| | - Changyi Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Tiantian Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
26
|
Collins KH, Gui C, Ely EV, Lenz KL, Harris CA, Guilak F, Meyer GA. Leptin mediates the regulation of muscle mass and strength by adipose tissue. J Physiol 2022; 600:3795-3817. [PMID: 35844058 PMCID: PMC9378542 DOI: 10.1113/jp283034] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Adipose tissue secretes numerous cytokines (termed 'adipokines') that have known or hypothesized actions on skeletal muscle. The majority of adipokines have been implicated in the pathological link between excess adipose and muscle insulin resistance, but approximately half also have documented in vitro effects on myogenesis and/or hypertrophy. This complexity suggests a potential dual role for adipokines in the regulation of muscle mass in homeostasis and the development of pathology. In this study, we used lipodystrophic 'fat-free' mice to demonstrate that adipose tissue is indeed necessary for the development of normal muscle mass and strength. Fat-free mice had significantly reduced mass (∼15%) and peak contractile tension (∼20%) of fast-twitch muscles, a slowing of contractile dynamics and decreased cross-sectional area of fast twitch fibres compared to wild-type littermates. These deficits in mass and contractile tension were fully rescued by reconstitution of ∼10% of normal adipose mass, indicating that this phenotype is the direct consequence of absent adipose. We then showed that the rescue is solely mediated by the adipokine leptin, as similar reconstitution of adipose from leptin-knockout mice fails to rescue mass or strength. Together, these data indicate that the development of muscle mass and strength in wild-type mice is dependent on adipose-secreted leptin. This finding extends our current understanding of the multiple roles of adipokines in physiology as well as disease pathophysiology to include a critical role for the adipokine leptin in muscle homeostasis. KEY POINTS: Adipose-derived cytokines (adipokines) have long been implicated in the pathogenesis of insulin resistance in obesity but likely have other under-appreciated roles in muscle physiology. Here we use a fat-free mouse to show that adipose tissue is necessary for the normal development of muscle mass and strength. Through add-back of genetically modified adipose tissue we show that leptin is the key adipokine mediating this regulation. This expands our understanding of leptin's role in adipose-muscle signalling to include development and homeostasis and adds the surprising finding that leptin is the sole mediator of the maintenance of muscle mass and strength by adipose tissue.
Collapse
Affiliation(s)
- Kelsey H. Collins
- Department of Orthopaedic SurgeryWashington University in St. LouisMOUSA,Shriners Hospitals for ChildrenSt LouisMOUSA,Center of Regenerative MedicineWashington University in St. LouisMOUSA
| | - Chang Gui
- Department of Biomedical EngineeringWashington University in St. LouisMOUSA,Program in Physical TherapyWashington UniversitySt LouisMOUSA
| | - Erica V. Ely
- Department of Orthopaedic SurgeryWashington University in St. LouisMOUSA,Shriners Hospitals for ChildrenSt LouisMOUSA,Center of Regenerative MedicineWashington University in St. LouisMOUSA,Department of Biomedical EngineeringWashington University in St. LouisMOUSA
| | - Kristin L. Lenz
- Department of Orthopaedic SurgeryWashington University in St. LouisMOUSA,Shriners Hospitals for ChildrenSt LouisMOUSA,Center of Regenerative MedicineWashington University in St. LouisMOUSA
| | - Charles A. Harris
- Division of EndocrinologyMetabolism & Lipid ResearchWashington UniversitySt LouisMissouriUSA
| | - Farshid Guilak
- Department of Orthopaedic SurgeryWashington University in St. LouisMOUSA,Shriners Hospitals for ChildrenSt LouisMOUSA,Center of Regenerative MedicineWashington University in St. LouisMOUSA,Department of Biomedical EngineeringWashington University in St. LouisMOUSA
| | - Gretchen A. Meyer
- Department of Orthopaedic SurgeryWashington University in St. LouisMOUSA,Center of Regenerative MedicineWashington University in St. LouisMOUSA,Department of Biomedical EngineeringWashington University in St. LouisMOUSA,Program in Physical TherapyWashington UniversitySt LouisMOUSA,Department of NeurologyWashington University in St. LouisSt LouisMOUSA
| |
Collapse
|
27
|
Piao L, Huang Z, Inoue A, Kuzuya M, Cheng XW. Human umbilical cord-derived mesenchymal stromal cells ameliorate aging-associated skeletal muscle atrophy and dysfunction by modulating apoptosis and mitochondrial damage in SAMP10 mice. Stem Cell Res Ther 2022; 13:226. [PMID: 35659361 PMCID: PMC9166592 DOI: 10.1186/s13287-022-02895-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background Skeletal muscle mass and function losses in aging individuals are associated with quality of life deterioration and disability. Mesenchymal stromal cells exert immunomodulatory and anti-inflammatory effects and could yield beneficial effects in aging-related degenerative disease. Methods and results We investigated the efficacy of umbilical cord-derived mesenchymal stromal cells (UC-MSCs) on sarcopenia-related skeletal muscle atrophy and dysfunction in senescence-accelerated mouse prone 10 (SAMP10) mice. We randomly assigned 24-week-old male SAMP10 mice to a UC-MSC treatment group and control group. At 12 weeks post-injection, the UC-MSC treatment had ameliorated sarcopenia-related muscle changes in performance, morphological structures, and mitochondria biogenesis, and it enhanced the amounts of proteins or mRNAs for myosin heavy chain, phospho-AMP-activated protein kinase, phospho-mammalian target of rapamycin, phospho-extracellular signal-regulated kinase1/2, peroxisome proliferator-activated receptor-γ coactivator, GLUT-4, COX-IV, and hepatocyte growth factor in both gastrocnemius and soleus muscles, and it reduced the levels of proteins or mRNAs for cathepsin K, cleaved caspase-3/-8, tumor necrosis factor-α, monocyte chemoattractant protein-1, and gp91phox mRNAs. The UC-MSC treatment retarded mitochondria damage, cell apoptosis, and macrophage infiltrations, and it enhanced desmin/laminin expression and proliferating and CD34+/Integrin α7+ cells in both types of skeletal muscle of the SAMP10 mice. In vitro, we observed increased levels of HGF, PAX-7, and MoyD mRNAs at the 4th passage of UC-MSCs. Conclusions Our results suggest that UC-MSCs can improve sarcopenia-related skeletal muscle atrophy and dysfunction via anti-apoptosis, anti-inflammatory, and mitochondrial biogenesis mechanisms that might be mediated by an AMPK-PGC1-α axis, indicating that UC-MSCs may provide a promising treatment for sarcopenia/muscle diseases.
Collapse
Affiliation(s)
- Limei Piao
- Department of Human Life Cord Applied Cell Therapy, Graduate School of Medicine, Nagoya University, Nagoya, Aichi-ken, 466-8550, Japan.,Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China
| | - Zhe Huang
- Department of Human Life Cord Applied Cell Therapy, Graduate School of Medicine, Nagoya University, Nagoya, Aichi-ken, 466-8550, Japan.
| | - Aiko Inoue
- Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
| | - Masafumi Kuzuya
- Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan.,Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Aichi-ken, 466-8550, Japan
| | - Xian Wu Cheng
- Department of Human Life Cord Applied Cell Therapy, Graduate School of Medicine, Nagoya University, Nagoya, Aichi-ken, 466-8550, Japan. .,Department of Cardiology and Hypertension, Yanbian University Hospital, Yanji, 133000, Jilin, People's Republic of China.
| |
Collapse
|
28
|
Wang T. Searching for the link between inflammaging and sarcopenia. Ageing Res Rev 2022; 77:101611. [PMID: 35307560 DOI: 10.1016/j.arr.2022.101611] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/14/2022] [Accepted: 03/15/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Tiantian Wang
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
29
|
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: 2.3] [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
|
30
|
Alizadeh Pahlavani H. Exercise Therapy for People With Sarcopenic Obesity: Myokines and Adipokines as Effective Actors. Front Endocrinol (Lausanne) 2022; 13:811751. [PMID: 35250869 PMCID: PMC8892203 DOI: 10.3389/fendo.2022.811751] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
Sarcopenic obesity is defined as a multifactorial disease in aging with decreased body muscle, decreased muscle strength, decreased independence, increased fat mass, due to decreased physical activity, changes in adipokines and myokines, and decreased satellite cells. People with sarcopenic obesity cause harmful changes in myokines and adipokines. These changes are due to a decrease interleukin-10 (IL-10), interleukin-15 (IL-15), insulin-like growth factor hormone (IGF-1), irisin, leukemia inhibitory factor (LIF), fibroblast growth factor-21 (FGF-21), adiponectin, and apelin. While factors such as myostatin, leptin, interleukin-6 (IL-6), interleukin-8 (IL-8), and resistin increase. The consequences of these changes are an increase in inflammatory factors, increased degradation of muscle proteins, increased fat mass, and decreased muscle tissue, which exacerbates sarcopenia obesity. In contrast, exercise, especially strength training, reverses this process, which includes increasing muscle protein synthesis, increasing myogenesis, increasing mitochondrial biogenesis, increasing brown fat, reducing white fat, reducing inflammatory factors, and reducing muscle atrophy. Since some people with chronic diseases are not able to do high-intensity strength training, exercises with blood flow restriction (BFR) are newly recommended. Numerous studies have shown that low-intensity BFR training produces the same increase in hypertrophy and muscle strength such as high-intensity strength training. Therefore, it seems that exercise interventions with BFR can be an effective way to prevent the exacerbation of sarcopenia obesity. However, due to limited studies on adipokines and exercises with BFR in people with sarcopenic obesity, more research is needed.
Collapse
|
31
|
Proliferin-1 Ameliorates Cardiotoxin-Related Skeletal Muscle Repair in Mice. Stem Cells Int 2021; 2021:9202990. [PMID: 34950212 PMCID: PMC8692050 DOI: 10.1155/2021/9202990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/22/2021] [Accepted: 10/28/2021] [Indexed: 12/29/2022] Open
Abstract
Background We recently demonstrated that proliferin-1 (PLF-1) functions as an apoptotic cell-derived growth factor and plays an important role in vascular pathobiology. We therefore investigated its role in muscle regeneration in response to cardiotoxin injury. Methods and Results To determine the effects of PLF-1 on muscle regeneration, we used a CTX-induced skeletal muscle injury model in 9-week-old male mice that were administered with the recombinant PLF-1 (rPLF-1) or neutralizing PLF-1 antibody. The injured muscles exhibited increased levels of PLF-1 gene expression in a time-dependent manner. On day 14 after injury, rPLF-1 supplementation ameliorated CTX-induced alterations in muscle fiber size, interstitial fibrosis, muscle regeneration capacity, and muscle performance. On day 3 postinjury, rPLF-1 increased the levels of proteins or genes for p-Akt, p-mTOR, p-GSK3α/β, p-Erk1/2, p-p38MAPK, interleukin-10, Pax7, MyoD, and Cyclin B1, and it increased the numbers of CD34+/integrin-α7+ muscle stem cells and proliferating cells in the muscles and/or bone marrow of CTX mice. An enzyme-linked immunosorbent assay revealed that rPLF-1 suppressed the levels of plasma tumor necrosis factor-α and interleukin-1β in CTX mice. PLF-1 blocking accelerated CTX-related muscle damage and dysfunction. In C2C12 myoblasts, rPLF-1 increased the levels of proteins for p-Akt, p-mTOR, p-GSK3α/β, p-Erk1/2, and p-p38MAPK as well as cellular functions; and these effects were diminished by the depletion of PLF-1 or silencing of its mannose-6-phosphate receptor. Conclusions These findings demonstrated that PLF-1 can improve skeletal muscle repair in response to injury, possibly via the modulation of inflammation and proliferation and regeneration, suggesting a novel therapeutic strategy for the management of skeletal muscle diseases.
Collapse
|
32
|
An L, Wang Y. Potential Roles of miRNA-1245a Regulatory Networks in Sarcopenia. Int J Gen Med 2021; 14:6807-6813. [PMID: 34703286 PMCID: PMC8523505 DOI: 10.2147/ijgm.s334501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/14/2021] [Indexed: 12/25/2022] Open
Abstract
Objective Sarcopenia is a universal problem in elderly individuals. The molecular regulatory mechanisms in sarcopenia are not well understood. In the present study, we explored a possible molecular mechanism involved in the pathogenesis of sarcopenia. Methods Differentially expressed genes (DEGs) were identified using the Gene Expression Omnibus (GEO) database. Signaling pathways related to these DEGs were identified by gene set enrichment analysis (GSEA). Pearson correlation was calculated for all the pairwise comparisons of gene expression values between coding genes and DEGs. Interactions between the proteins encoded by the DEGs were identified using the STRING database. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) biological pathway analyses were performed to predict the functions of the DEGs. Results Three differentially expressed miRNAs and 5 differentially expressed mRNAs were identified in association with DEGs. We found that miRNA-1245a expression in patients with sarcopenia was higher than that in healthy controls. The GSEA showed that many pathways, such as the JAK-STAT signaling pathway and pathways related to glioma, gap junctions, and regulation of the actin cytoskeleton, were enriched in the high-miRNA-1245a-expression group. A total of 127 miRNA-1245a-related mRNAs were identified. The GO and KEGG analyses revealed that miRNA-1245a had a strong effect on a number of fundamental biological processes, such as kinase activity, that are related to the development of sarcopenia. Conclusion Our analyses indicate that miRNA-1245a may be a potential key molecule in the diagnosis and treatment of sarcopenia, which provides a basis for the research of miRNA in sarcopenia.
Collapse
Affiliation(s)
- Li An
- Department of Geriatrics, Zhongda Hospital Southeast University,, Nanjing City, People's Republic of China
| | - Yao Wang
- Department of Endocrine, Zhongda Hospital Southeast University, Nanjing City, People's Republic of China
| |
Collapse
|
33
|
Kamada Y, Toyama S, Arai Y, Inoue H, Nakagawa S, Fujii Y, Kaihara K, Kishida T, Mazda O, Takahashi K. Treadmill running prevents atrophy differently in fast- versus slow-twitch muscles in a rat model of rheumatoid arthritis. J Muscle Res Cell Motil 2021; 42:429-441. [PMID: 34687403 DOI: 10.1007/s10974-021-09610-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 10/13/2021] [Indexed: 10/20/2022]
Abstract
To investigate the effects of treadmill running on two different types of skeletal muscle, we established a rat model of collagen-induced arthritis (CIA). The skeletal muscles studied were the extensor digitorum longus (EDL), which is rich in fast-twitch muscle fibers, and the soleus, which is rich in slow-twitch muscle fibers. The histological and transcriptional changes in these muscles at 14 and 44 days after immunosensitization were compared between rats that were forced to exercise (CIA ex group) and free-reared CIA rats (CIA no group). Change in protein expression was examined on day 14 after a single bout of treadmill running. Treadmill running had different effects on the relative muscle weight and total and fiber cross-sectional areas in each muscle type. In the soleus, it prevented muscle atrophy. Transcriptional analysis revealed increased eukaryotic translation initiation factor 4E (Eif4e) expression on day 14 and increased Atrogin-1 and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) expression on day 44 in the soleus in the CIA ex group, suggesting an interaction between muscle type and exercise. A single bout of treadmill running increased the level of Eif4e and p70S6K and decreased that of Atrogin-1 in the soleus on day 14. Treadmill running prevented muscle atrophy in the soleus in a rat model of rheumatoid arthritis via activation of mitochondrial function, as evidenced by increased PGC-1α expression.
Collapse
Affiliation(s)
- Yoichiro Kamada
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Shogo Toyama
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Yuji Arai
- Department of Sports and Para-Sports Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan.
| | - Hiroaki Inoue
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Shuji Nakagawa
- Department of Sports and Para-Sports Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Yuta Fujii
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Kenta Kaihara
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Tsunao Kishida
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Kenji Takahashi
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| |
Collapse
|
34
|
Pan R, Chen Y. Management of Oxidative Stress: Crosstalk Between Brown/Beige Adipose Tissues and Skeletal Muscles. Front Physiol 2021; 12:712372. [PMID: 34603076 PMCID: PMC8481590 DOI: 10.3389/fphys.2021.712372] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/19/2021] [Indexed: 12/23/2022] Open
Abstract
Exercise plays an important role in the physiology, often depending on its intensity, duration, and frequency. It increases the production of reactive oxygen species (ROS). Meanwhile, it also increases antioxidant enzymes involved in the oxidative damage defense. Prolonged, acute, or strenuous exercise often leads to an increased radical production and a subsequent oxidative stress in the skeletal muscles, while chronic regular or moderate exercise results in a decrease in oxidative stress. Notably, under pathological state, such as obesity, aging, etc., ROS levels could be elevated in humans, which could be attenuated by proper exercise. Significantly, exercise stimulates the development of beige adipose tissue and potentially influence the function of brown adipose tissue (BAT), which is known to be conducive to a metabolic balance through non-shivering thermogenesis (NST) and may protect from oxidative stress. Exercise-related balance of the ROS levels is associated with a healthy metabolism in humans. In this review, we summarize the integrated effects of exercise on oxidative metabolism, and especially focus on the role of brown and beige adipose tissues in this process, providing more evidence and knowledge for a better management of exercise-induced oxidative stress.
Collapse
Affiliation(s)
- Ruping Pan
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Chen
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| |
Collapse
|
35
|
Xie WQ, He M, Yu DJ, Wu YX, Wang XH, Lv S, Xiao WF, Li YS. Mouse models of sarcopenia: classification and evaluation. J Cachexia Sarcopenia Muscle 2021; 12:538-554. [PMID: 33951340 PMCID: PMC8200444 DOI: 10.1002/jcsm.12709] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/09/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023] Open
Abstract
Sarcopenia is a progressive and widespread skeletal muscle disease that is related to an increased possibility of adverse consequences such as falls, fractures, physical disabilities and death, and its risk increases with age. With the deepening of the understanding of sarcopenia, the disease has become a major clinical disease of the elderly and a key challenge of healthy ageing. However, the exact molecular mechanism of this disease is still unclear, and the selection of treatment strategies and the evaluation of its effect are not the same. Most importantly, the early symptoms of this disease are not obvious and are easy to ignore. In addition, the clinical manifestations of each patient are not exactly the same, which makes it difficult to effectively study the progression of sarcopenia. Therefore, it is necessary to develop and use animal models to understand the pathophysiology of sarcopenia and develop therapeutic strategies. This paper reviews the mouse models that can be used in the study of sarcopenia, including ageing models, genetically engineered models, hindlimb suspension models, chemical induction models, denervation models, and immobilization models; analyses their advantages and disadvantages and application scope; and finally summarizes the evaluation of sarcopenia in mouse models.
Collapse
Affiliation(s)
- Wen-Qing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Miao He
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Deng-Jie Yu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu-Xiang Wu
- School of Kinesiology, Jianghan University, Wuhan, Hubei, China
| | - Xiu-Hua Wang
- Xiang Ya Nursing School, The Central South University, Changsha, Hunan, China
| | - Shan Lv
- Department of Geriatric Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wen-Feng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
36
|
Antiobesity effects of kimchi added with Jeju citrus concentrate on high-fat diet-induced obese mice. Nutr Res 2021; 86:50-59. [PMID: 33482598 DOI: 10.1016/j.nutres.2020.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 10/29/2020] [Accepted: 11/15/2020] [Indexed: 12/14/2022]
Abstract
Citrus fruits contain an abundance of nutrients, including vitamins C and B6 and hesperidin, which attribute to its beneficial health effects. Previously, kimchi with Jeju citrus concentrate (CK) elicited anti-obesity effects in 3T3-L1 adipocytes. Here, we aimed to investigate whether CK exhibits anti-obesity effects by reducing serum and hepatic lipid concentrations and anti-obesity-associated gene expression in high-fat diet (HFD)-induced obese C57BL/6N mice. Low-dose CK (LDCK, 50 mg/kg) and high-dose CK (HDCK, 200 mg/kg) were orally administered 3 times per week over 8 weeks with HFD diet. Body weight gain, food efficiency ratio, and tissue weight were measured. Serum glutamic oxaloacetic transaminase, glutamic pyruvic transaminase, fasting glucose, fasting insulin, homeostatic model assessment-insulin resistance, leptin, and adiponectin concentrations were also assessed. The effect of CK on the lipid profile and lipid accumulation was analyzed. Body and white adipose tissue masses were significantly lower in the LDCK and HDCK groups than in the HFD group. Orally administered CK significantly decreased serum lipid, fasting glucose, fasting insulin, homeostatic model assessment-insulin resistance, glutamic oxaloacetic transaminase, and glutamic pyruvic transaminase levels. Hepatic lipid content also decreased in the LDCK and HDCK groups. Serum leptin concentrations decreased, whereas serum adiponectin concentrations increased, confirming the anti-obesity effects of LDCK and HDCK. The decrease of hepatic vacuoles and stained lipid droplets indicated inhibition of lipid accumulation. These results support the hypothesis that CK exhibits anti-obesity effects in vivo by reducing lipid accumulation and by regulating anti-obesity-related genes.
Collapse
|
37
|
Komici K, Dello Iacono A, De Luca A, Perrotta F, Bencivenga L, Rengo G, Rocca A, Guerra G. Adiponectin and Sarcopenia: A Systematic Review With Meta-Analysis. Front Endocrinol (Lausanne) 2021; 12:576619. [PMID: 33935962 PMCID: PMC8082154 DOI: 10.3389/fendo.2021.576619] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Sarcopenia is a progressive loss of skeletal muscle mass whose pathophysiology has been proposed to possibly involve mechanisms of altered inflammatory status and endocrine function. Adiponectin has been shown to modulate inflammatory status and muscle metabolism. However, the possible association between adiponectin levels and sarcopenia is poorly understood. In order to fill this gap, in the present manuscript we aimed to summarize the current evidence with a systematic review and a meta-analysis of studies reporting serum adiponectin levels in patients with sarcopenia compared to non-sarcopenic controls. METHODS An electronic search through Medline/PubMed, Cochrane Library, and Science Direct was performed till March 1, 2020. From the included papers, meta-analysis of cross-sectional studies comparing serum levels of adiponectin between patients with sarcopenia and controls was performed. RESULTS Out of 1,370 initial studies, seven studies were meta-analyzed. Sarcopenic participants had significantly higher levels of adiponectin Hedges' g with 95% confidence interval (CI): 1.20 (0.19-2.22), p = 0.02 than controls. Subgroup analysis, performed in Asian population and focused on identification of the condition based on AWGS criteria, reported higher adiponectin levels in sarcopenic population (2.1 (0.17-4.03), p = 0.03 and I2 = 98.98%. Meta-regression analysis revealed female gender to significantly influence the results as demonstrated by beta = 0.14 (95% CI (0.010-0.280), p = 0.040). CONCLUSIONS Our meta-analysis found evidence that sarcopenia is associated with higher adiponectin levels. However, caution is warranted on the interpretation of these findings, and future longitudinal research is required to disentangle and better understand the topic.
Collapse
Affiliation(s)
- Klara Komici
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
- *Correspondence: Klara Komici,
| | - Antonio Dello Iacono
- School of Health and Life Sciences, University of the West of Scotland, Hamilton, United Kingdom
| | - Antonio De Luca
- Section of Human Anatomy, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Fabio Perrotta
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
- Istituti Clinici Scientifici Maugeri SpA Società Benefit (ICS Maugeri SpA SB), Telese Terme, Italy
| | - Aldo Rocca
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| |
Collapse
|
38
|
Muratsu J, Kamide K, Fujimoto T, Takeya Y, Sugimoto K, Taniyama Y, Morishima A, Sakaguchi K, Matsuzawa Y, Rakugi H. The Combination of High Levels of Adiponectin and Insulin Resistance Are Affected by Aging in Non-Obese Old Peoples. Front Endocrinol (Lausanne) 2021; 12:805244. [PMID: 35069451 PMCID: PMC8777034 DOI: 10.3389/fendo.2021.805244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/15/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Adipokine dysregulation is a key feature of insulin resistance and a metabolic syndrome associated with obesity. Low adiponectin levels are associated with higher risks of cardiovascular diseases (CVD). However, high adiponectin levels have also been associated with increased all-cause and cardiovascular mortality in the elderly. This adiponectin paradox has yet to be clarified, which has hindered our understanding of the biological role of adiponectin. Adipokine dysregulation and insulin resistance are also associated with energy-deprivation conditions, such as frailty in old age. The objective of this study was to investigate the association between plasma adiponectin and insulin resistance using the homeostasis model assessment for insulin resistance (HOMA-IR) classified by age. In particular, we sought to determine the factors of the subjects associated with both high adiponectin levels and HOMA-IR (H-adiponectin/H-HOMA) and high adiponectin levels and low HOMA-IR (H-adiponectin/L-HOMA). METHODS The eligible subjects in this cross-sectional study were 33,216 individuals who had undergone health checkups at the Physical Checkup Center of Sumitomo Hospital between April 2008 and December 2018. After excluding 26,371 individuals who were under 60 years old, 529 who had been taking medications for diabetes mellitus, and 690 with missing data, the present study included 5,673 (3,467 males, 2,206 females) subjects with no missing data. The relationship between serum adiponectin levels and HOMA-IR was assessed using logistic regression models adjusted by clinically relevant factors. RESULTS In the multivariable logistic regression analysis, age and low BMI were shown to positively correlate with the characteristics of H-adiponectin/H-HOMA. In females, systolic blood pressure was also shown to be an associated factor. CONCLUSION In conclusion, this study showed that aging or a low BMI may contribute to high adiponectin levels and insulin resistance.
Collapse
Affiliation(s)
- Jun Muratsu
- Department of Nephrology and Hypertension, Sumitomo Hospital, Osaka, Japan
- Department of Nephrology, Rinku General Medical Center, Izumisano City, Japan
- *Correspondence: Jun Muratsu,
| | - Kei Kamide
- Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takashi Fujimoto
- Department of Nephrology and Hypertension, Sumitomo Hospital, Osaka, Japan
| | - Yasushi Takeya
- Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ken Sugimoto
- Department of General and Geriatric Medicine, Kawasaki Medical School General Medical Center, Okayama, Japan
| | - Yoshiaki Taniyama
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Atsuyuki Morishima
- Department of Nephrology and Hypertension, Sumitomo Hospital, Osaka, Japan
| | | | - Yuji Matsuzawa
- Department of Nephrology and Hypertension, Sumitomo Hospital, Osaka, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
39
|
Liu Q, Gao J, Deng J, Xiao J. Current Studies and Future Directions of Exercise Therapy for Muscle Atrophy Induced by Heart Failure. Front Cardiovasc Med 2020; 7:593429. [PMID: 33195482 PMCID: PMC7644508 DOI: 10.3389/fcvm.2020.593429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Muscle atrophy is a common complication of heart failure. At present, there is no specific treatment to reverse the course of muscle atrophy. Exercise training, due to the safety and easy operation, is a recommended therapy for muscle atrophy induced by heart failure. However, the patients with muscle atrophy are weak in mobility and may not be able to train for a long time. Therefore, it is necessary to explore novel targets of exercise protection for muscle atrophy, so as to improve the quality of life and survival rate of patients with muscular atrophy induced by heart failure. This article aims to review latest studies, summarize the evidence and limitations, and provide a glimpse into the future of exercise for the treatment of muscle atrophy induced by heart failure. We wish to highlight some important findings about the essential roles of exercise sensors in muscle atrophy induced by heart failure, which might be helpful for searching potential therapeutic targets for muscle wasting induced by heart failure.
Collapse
Affiliation(s)
- Qi Liu
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Juan Gao
- School of Medicine, Shanghai University, Shanghai, China
| | - Jiali Deng
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China.,School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
40
|
Alcazar CA, Hu C, Rando TA, Huang NF, Nakayama KH. Transplantation of insulin-like growth factor-1 laden scaffolds combined with exercise promotes neuroregeneration and angiogenesis in a preclinical muscle injury model. Biomater Sci 2020; 8:5376-5389. [PMID: 32996916 DOI: 10.1039/d0bm00990c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The regeneration of skeletal muscle can be permanently impaired by traumatic injuries, despite the high regenerative capacity of native muscle. An attractive therapeutic approach for treating severe muscle inuries is the implantation of off-the-shelf engineered biomimetic scaffolds into the site of tissue damage to enhance muscle regeneration. Anisotropic nanofibrillar scaffolds provide spatial patterning cues to create organized myofibers, and growth factors such as insulin-like growth factor-1 (IGF-1) are potent inducers of both muscle regeneration as well as angiogenesis. The aim of this study was to test the therapeutic efficacy of anisotropic IGF-1-releasing collagen scaffolds combined with voluntary exercise for the treatment of acute volumetric muscle loss, with a focus on histomorphological effects. To enhance the angiogenic and regenerative potential of injured murine skeletal muscle, IGF-1-laden nanofibrillar scaffolds with aligned topography were fabricated using a shear-mediated extrusion approach, followed by growth factor adsorption. Individual scaffolds released a cumulative total of 1244 ng ± 153 ng of IGF-1 over the course of 21 days in vitro. To test the bioactivity of IGF-1-releasing scaffolds, the myotube formation capacity of murine myoblasts was quantified. On IGF-1-releasing scaffolds seeded with myoblasts, the resulting myotubes formed were 1.5-fold longer in length and contained 2-fold greater nuclei per myotube, when compared to scaffolds without IGF-1. When implanted into the ablated murine tibialis anterior muscle, the IGF-1-laden scaffolds, in conjunction with voluntary wheel running, significantly increased the density of perfused microvessels by greater than 3-fold, in comparison to treatment with scaffolds without IGF-1. Enhanced myogenesis was also observed in animals treated with the IGF-1-laden scaffolds combined with exercise, compared to control scaffolds transplanted into mice that did not receive exercise. Furthermore, the abundance of mature neuromuscular junctions was greater by approximately 2-fold in muscles treated with IGF-1-laden scaffolds, when paired with exercise, in comparison to the same treatment without exercise. These findings demonstrate that voluntary exercise improves the regenerative effect of growth factor-laden scaffolds by augmenting neurovascular regeneration, and have important translational implications in the design of off-the-shelf therapeutics for the treatment of traumatic muscle injury.
Collapse
Affiliation(s)
- Cynthia A Alcazar
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.
| | | | | | | | | |
Collapse
|
41
|
Low energy irradiation of narrow-range UV-LED prevents osteosarcopenia associated with vitamin D deficiency in senescence-accelerated mouse prone 6. Sci Rep 2020; 10:11892. [PMID: 32681041 PMCID: PMC7368004 DOI: 10.1038/s41598-020-68641-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/29/2020] [Indexed: 12/21/2022] Open
Abstract
Deficiency of vitamin D is an important cause of osteosarcopenia. The purpose of this study is to examine the effects of low energy narrow-range UV-LED on osteosarcopenia in animal models of senescence-accelerated mouse prone 6 (SAMP6). Preliminary experiments specified the minimum irradiance intensity and dose efficacy for vitamin D production (316 nm, 0.16 mW/cm2, 1,000 J/m2). we set a total of 4 groups (n = 8 per group); vitamin D-repletion without UV irradiation (Vit.D+UV-), vitamin D-repletion with UV irradiation (Vit.D+UV +), vitamin D-deficiency without UV irradiation, (Vit.D-UV-), and vitamin D-deficiency with UV irradiation (Vit.D-UV +). Serum levels of 25(OH)D at 28 and 36 weeks of age were increased in Vit.D-UV+ group as compared with Vit.D-UV- group. Trabecular bone mineral density on micro-CT was higher in Vit.D-UV+ group than in Vit.D-UV- group at 36 weeks of age. In the histological assay, fewer osteoclasts were observed in Vit.D-UV+ group than in Vit.D-UV- group. Grip strength and muscle mass were higher in Vit.D-UV+ group than in Vit.D-UV- group at 36 weeks of age. Signs of severe damage induced by UV irradiation was not found in skin histology. Low energy narrow-range UV irradiation may improve osteosarcopenia associated with vitamin D deficiency in SAMP6.
Collapse
|
42
|
Paris MT, Bell KE, Mourtzakis M. Myokines and adipokines in sarcopenia: understanding cross-talk between skeletal muscle and adipose tissue and the role of exercise. Curr Opin Pharmacol 2020; 52:61-66. [PMID: 32668398 DOI: 10.1016/j.coph.2020.06.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
Abstract
Detrimental age-associated changes in skeletal muscle and adipose tissue increase the risk of sarcopenia. Age-related changes in myokines, such as myostatin and irisin, as well as adipokines, such as leptin and adiponectin, contribute to cross-talk between muscle and adipose tissue. These age-related changes in myokines and adipokines have important implications for sarcopenia, however, recent literature highlights discrepancies in these relationships. Exercise may alter serum profiles and muscle receptor expression of these factors, but future work is needed to determine whether these changes in myokines and adipokines relate to improvements in muscle mass and function. Here, we describe myokine-mediated and adipokine-mediated interactions between muscle and adipose tissue, and discuss the fundamental importance of these cytokines to understanding the development of sarcopenia.
Collapse
Affiliation(s)
- Michael T Paris
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 1A3, Canada
| | - Kirsten E Bell
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 1A3, Canada
| | - Marina Mourtzakis
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 1A3, Canada.
| |
Collapse
|
43
|
Wang P, Liang Y, Chen K, Yau SY, Sun X, Cheng KKY, Xu A, So KF, Li A. Potential Involvement of Adiponectin Signaling in Regulating Physical Exercise-Elicited Hippocampal Neurogenesis and Dendritic Morphology in Stressed Mice. Front Cell Neurosci 2020; 14:189. [PMID: 32774242 PMCID: PMC7381385 DOI: 10.3389/fncel.2020.00189] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022] Open
Abstract
Adiponectin, a cytokine secreted by mature adipocytes, proves to be neuroprotective. We have previously reported that running triggers adiponectin up-regulation which subsequently promotes generation of hippocampal neurons and thereby alleviates depression-like behaviors in non-stressed mice. However, under the stressing condition, whether adiponectin could still exert antidepressant-like effects following exercise remained unexplored. In this study, by means of repeated corticosterone injections to mimic stress insult and voluntary wheel running as physical exercise intervention, we examined whether exercise-elicited antidepressive effects might involve adiponectin's regulation on hippocampal neurogenesis and dendritic plasticity in stressed mice. Here we show that repeated injections of corticosterone inhibited hippocampal neurogenesis and impaired dendritic morphology of neurons in the dentate gyrus of both wild-type and adiponectin-knockout mice comparably, which subsequently evoked depression-like behaviors. Voluntary wheel running attenuated corticosterone-suppressed neurogenesis and enhanced dendritic plasticity in the hippocampus, ultimately reducing depression-like behaviors in wild-type, but not adiponectin-knockout mice. We further demonstrate that such proneurogenic effects were potentially achieved through activation of the AMP-dependent kinase (AMPK) pathway. Our study provides the first evidence that adiponectin signaling is essential for physical exercise-triggered effects on stress-elicited depression by retaining the normal proliferation of neural progenitors and dendritic morphology of neurons in the hippocampal dentate gyrus, which may depend on activation of the AMPK pathway.
Collapse
Affiliation(s)
- Pingjie Wang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Jinan University, Guangzhou, China
| | - Yiyao Liang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Jinan University, Guangzhou, China
| | - Kai Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Jinan University, Guangzhou, China
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Xin Sun
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Jinan University, Guangzhou, China
| | - Kenneth King-Yip Cheng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Aimin Xu
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Pharmacy and Pharmacology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Jinan University, Guangzhou, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ang Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Jinan University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
44
|
The ratio of prematurely aging to non-prematurely aging mice cohabiting, conditions their behavior, immunity and lifespan. J Neuroimmunol 2020; 343:577240. [PMID: 32330742 DOI: 10.1016/j.jneuroim.2020.577240] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/09/2020] [Accepted: 04/09/2020] [Indexed: 12/15/2022]
Abstract
Adult prematurely aging mice (PAM) show behavioral deterioration, premature immunosenescence and increased oxidative stress, impairments that are associated with their shorter lifespan, compared to the corresponding exceptional non-prematurely aging mice (ENPAM). When PAM live in a predominantly ENPAM environment (2/5, respectively) they exhibit an improvement of immunity and redox state in their spleen and thymus leukocytes, and an increased lifespan. Nevertheless, it is unknown if other PAM/ENPAM ratios could affect behavioral and peritoneal leukocyte functions of PAM and change their lifespan. ENPAM and PAM were divided into the following groups: C-ENPAM (8 ENPAM in the cage); C-PAM (8 PAM in the cage); ENPAM>50% and PAM<50% (5 ENPAM/2 PAM in each cage); ENPAM = 50% and PAM = 50% (4 ENPAM/4 PAM in each cage), and PAM>50% and ENPAM<50% (5 PAM/2 ENPAM in each cage). After two months, mice were submitted to a battery of behavioral tests. Several functions and oxidative stress parameters were then assessed in their peritoneal leukocytes. Animals were maintained in these conditions to analyze their lifespan. The results showed that PAM>50%, PAM = 50% and PAM<50% exhibited better behavioral responses, immunity and redox states in their peritoneal leukocytes than C-PAM. This improvement was higher when the number of ENPAM in the cage was increased, with most of the parameters in PAM<50% reaching similar values to those in C-ENPAM, and an increased lifespan. However, ENPAM that cohabited with PAM showed, in general, an impairment of parameters studied. In conclusion, the PAM/ENPAM cohabitation ratio is relevant to behavior and immunity.
Collapse
|
45
|
Mankhong S, Kim S, Moon S, Kwak HB, Park DH, Kang JH. Experimental Models of Sarcopenia: Bridging Molecular Mechanism and Therapeutic Strategy. Cells 2020; 9:E1385. [PMID: 32498474 PMCID: PMC7348939 DOI: 10.3390/cells9061385] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
Sarcopenia has been defined as a progressive decline of skeletal muscle mass, strength, and functions in elderly people. It is accompanied by physical frailty, functional disability, falls, hospitalization, and mortality, and is becoming a major geriatric disorder owing to the increasing life expectancy and growing older population worldwide. Experimental models are critical to understand the pathophysiology of sarcopenia and develop therapeutic strategies. Although its etiologies remain to be further elucidated, several mechanisms of sarcopenia have been identified, including cellular senescence, proteostasis imbalance, oxidative stress, and "inflammaging." In this article, we address three main aspects. First, we describe the fundamental aging mechanisms. Next, we discuss both in vitro and in vivo experimental models based on molecular mechanisms that have the potential to elucidate the biochemical processes integral to sarcopenia. The use of appropriate models to reflect sarcopenia and/or its underlying pathways will enable researchers to understand sarcopenia and develop novel therapeutic strategies for sarcopenia. Lastly, we discuss the possible molecular targets and the current status of drug candidates for sarcopenia treatment. In conclusion, the development of experimental models for sarcopenia is essential to discover molecular targets that are valuable as biochemical biomarkers and/or therapeutic targets for sarcopenia.
Collapse
Affiliation(s)
- Sakulrat Mankhong
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.); (S.M.)
| | - Sujin Kim
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.); (S.M.)
- Department of Kinesiology, Inha University, Incheon 22212, Korea; (H.-B.K.); (D.-H.P.)
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| | - Sohee Moon
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.); (S.M.)
| | - Hyo-Bum Kwak
- Department of Kinesiology, Inha University, Incheon 22212, Korea; (H.-B.K.); (D.-H.P.)
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| | - Dong-Ho Park
- Department of Kinesiology, Inha University, Incheon 22212, Korea; (H.-B.K.); (D.-H.P.)
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| | - Ju-Hee Kang
- Department of Pharmacology, Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea; (S.M.); (S.K.); (S.M.)
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| |
Collapse
|
46
|
Abou-Samra M, Selvais CM, Dubuisson N, Brichard SM. Adiponectin and Its Mimics on Skeletal Muscle: Insulin Sensitizers, Fat Burners, Exercise Mimickers, Muscling Pills … or Everything Together? Int J Mol Sci 2020; 21:ijms21072620. [PMID: 32283840 PMCID: PMC7178193 DOI: 10.3390/ijms21072620] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 02/07/2023] Open
Abstract
Adiponectin (ApN) is a hormone abundantly secreted by adipocytes and it is known to be tightly linked to the metabolic syndrome. It promotes insulin-sensitizing, fat-burning, and anti-atherosclerotic actions, thereby effectively counteracting several metabolic disorders, including type 2 diabetes, obesity, and cardiovascular diseases. ApN is also known today to possess powerful anti-inflammatory/oxidative and pro-myogenic effects on skeletal muscles exposed to acute or chronic inflammation and injury, mainly through AdipoR1 (ApN specific muscle receptor) and AMP-activated protein kinase (AMPK) pathway, but also via T-cadherin. In this review, we will report all the beneficial and protective properties that ApN can exert, specifically on the skeletal muscle as a target tissue. We will highlight its effects and mechanisms of action, first in healthy skeletal muscle including exercised muscle, and second in diseased muscle from a variety of pathological conditions. In the end, we will go over some of AdipoRs agonists that can be easily produced and administered, and which can greatly mimic ApN. These interesting and newly identified molecules could pave the way towards future therapeutic approaches to potentially prevent or combat not only skeletal muscle disorders but also a plethora of other diseases with sterile inflammation or metabolic dysfunction.
Collapse
|
47
|
Abou-Samra M, Selvais CM, Boursereau R, Lecompte S, Noel L, Brichard SM. AdipoRon, a new therapeutic prospect for Duchenne muscular dystrophy. J Cachexia Sarcopenia Muscle 2020; 11:518-533. [PMID: 31965757 PMCID: PMC7113498 DOI: 10.1002/jcsm.12531] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 10/23/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Adiponectin (ApN) is a hormone known to exhibit insulin-sensitizing, fat-burning, and anti-inflammatory properties in several tissues, including the skeletal muscle. Duchenne muscular dystrophy (DMD) is a devastating disease characterized by dystrophin deficiency with subsequent chronic inflammation, myofiber necrosis, and impaired regeneration. Previously, we showed that transgenic up-regulation of ApN could significantly attenuate the dystrophic phenotype in mdx mice (model of DMD). Recently, an orally active ApN receptor agonist, AdipoRon, has been identified. This synthetic small molecule has the advantage of being more easily produced and administrable than ApN. The aim of this study was to investigate the potential effects of AdipoRon on the dystrophic muscle. METHODS Four-week-old mdx mice (n = 6-9 per group) were orally treated with AdipoRon (mdx-AR) for 8 weeks and compared with untreated (mdx) mice and to control (wild-type) mice. In vivo functional tests were carried out to measure the global force and endurance of mice. Ex vivo biochemical and molecular analyses were performed to evaluate the pathophysiology of the skeletal muscle. Finally, in vitro tests were conducted on primary cultures of healthy and DMD human myotubes. RESULTS AdipoRon treatment mitigated oxidative stress (-30% to 45% for 4-hydroxy-2-nonenal and peroxiredoxin 3, P < 0.0001) as well as inflammation in muscles of mdx mice (-35% to 65% for interleukin 1 beta, tumour necrosis factor alpha, and cluster of differentiation 68, a macrophage maker, P < 0.0001) while increasing the anti-inflammatory cytokine, interleukin 10 (~5-fold, P < 0.0001). AdipoRon also improved the myogenic programme as assessed by a ~2-fold rise in markers of muscle proliferation and differentiation (P < 0.01 or less vs. untreated mdx). Plasma lactate dehydrogenase and creatine kinase were reduced by 30-40% in mdx-AR mice, reflecting less sarcolemmal damage (P < 0.0001). When compared with untreated mdx mice, mdx-AR mice exhibited enhanced physical performance with an increase in both muscle force and endurance and a striking restoration of the running capacity during eccentric exercise. AdipoRon mainly acted through ApN receptor 1 by increasing AMP-activated protein kinase signalling, which led to repression of nuclear factor-kappa B, up-regulation of utrophin (a dystrophin analogue), and a switch towards an oxidative and more resistant fibre phenotype. The effects of AdipoRon were then recapitulated in human DMD myotubes. CONCLUSIONS These results demonstrate that AdipoRon exerts several beneficial effects on the dystrophic muscle. This molecule could offer promising therapeutic prospect for managing DMD or other muscle and inflammatory disorders.
Collapse
Affiliation(s)
- Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, Brussels, Belgium
| | - Camille M Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, Brussels, Belgium
| | - Raphael Boursereau
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, Brussels, Belgium
| | - Sophie Lecompte
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, Brussels, Belgium
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, Brussels, Belgium
| | - Sonia M Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
48
|
Loncar G, Cvetinovic N, Lainscak M, Isaković A, von Haehling S. Bone in heart failure. J Cachexia Sarcopenia Muscle 2020; 11:381-393. [PMID: 32087616 PMCID: PMC7113538 DOI: 10.1002/jcsm.12516] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 10/10/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022] Open
Abstract
There is an increasing interest in osteoporosis and reduced bone mineral density affecting not only post-menopausal women but also men, particularly with coexisting chronic diseases. Bone status in patients with stable chronic heart failure (HF) has been rarely studied so far. HF and osteoporosis are highly prevalent aging-related syndromes that exact a huge impact on society. Both disorders are common causes of loss of function and independence, and of prolonged hospitalizations, presenting a heavy burden on the health care system. The most devastating complication of osteoporosis is hip fracture, which is associated with high mortality risk and among those who survive, leads to a loss of function and independence often necessitating admission to long-term care. Current HF guidelines do not suggest screening methods or patient education in terms of osteoporosis or osteoporotic fracture. This review may serve as a solid base to discuss the need for bone health evaluation in HF patients.
Collapse
Affiliation(s)
- Goran Loncar
- Institute for Cardiovascular Diseases Dedinje, Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany
| | - Natasa Cvetinovic
- Department of Cardiology, University Clinical Hospital Center 'Dr. Dragisa Misovic-Dedinje', Belgrade, Serbia
| | - Mitja Lainscak
- Department of Internal Medicine, General Hospital Murska Sobota, Murska Sobota, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | - Stephan von Haehling
- Department of Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University, Goettingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Goettingen, Goettingen, Germany
| |
Collapse
|
49
|
Liang J, Zeng Z, Zhang Y, Chen N. Regulatory role of exercise-induced autophagy for sarcopenia. Exp Gerontol 2019; 130:110789. [PMID: 31765742 DOI: 10.1016/j.exger.2019.110789] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 10/24/2019] [Accepted: 11/20/2019] [Indexed: 12/21/2022]
Abstract
Sarcopenia is an aging-related disease, described as the progressive reduction in mass and strength of skeletal muscle. Sarcopenia is typically characterized as the accumulation of damaged products due to an imbalance between protein synthesis and protein degradation. This imbalance between protein synthesis and degradation is attributed to impaired autophagic signal pathways. Sarcopenia can predispose elderly patients to several complications that may significantly impact patient quality of life. Recent evidence indicates that autophagy is required for the control of skeletal muscle mass under catabolic conditions and plays a crucial role in maintaining the homeostasis and integrity of skeletal muscle, specifically at appropriate level of autophagy. Exercise may be considered as a stress stimulus that can substantially modulate cellular signaling to promote metabolic adaptations. Appropriate exercise can induce autophagy or regulate the functional status of autophagy. Additionally, exercise-induced autophagy is the most effective treatment available in slowing down sarcopenia, improving mitochondrial quality, and the number of quiescent satellite cells, as a process that depends on basal autophagy. The molecular mechanisms underpinning the development of sarcopenia, however, remained largely unknown. In this narrative review, the current molecular mechanisms of sarcopenia are discussed from the perspective of exercise-induced autophagy and the effect of different exercise modalities on this response. This narrative review will aim to provide the references for developing scientific and optimal intervention strategies including exercise intervention for the prevention and treatment of sarcopenia through regulating autophagic signal pathways.
Collapse
Affiliation(s)
- Jiling Liang
- Graduate School, Wuhan Sports University, Wuhan 430079, China
| | - Zhengzhong Zeng
- Graduate School, Wuhan Sports University, Wuhan 430079, China
| | - Ying Zhang
- Graduate School, Wuhan Sports University, Wuhan 430079, China
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan 430079, China.
| |
Collapse
|
50
|
Uno C, Maeda K, Wakabayashi H, Nishioka S, Ogawa N, Okamoto T, Hoyano K, Momosaki R. Nutritional status change and activities of daily living in elderly pneumonia patients admitted to acute care hospital: A retrospective cohort study from the Japan Rehabilitation Nutrition Database. Nutrition 2019; 71:110613. [PMID: 31837639 DOI: 10.1016/j.nut.2019.110613] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The aim of this study was to examine the effect of improved nutritional status on activities of daily living (ADLs) and dysphagia in elderly patients with pneumonia who were admitted to acute care hospitals. METHODS A retrospective cohort study was conducted using registry data from the Japan Rehabilitation Nutrition Database of patients with pneumonia who were admitted to acute care hospitals. Patients were divided into two groups based on the Mini Nutritional Assessment Short-Form (MNA-SF) status at discharge: Patients with no status change or with decreased status were allocated to the unimproved nutritional status (UN) group and those with increased status were assigned to the improved nutritional status (IN) group. The primary outcome was ADLs as assessed by Barthel Index (BI) score at hospital discharge. Secondary outcomes included dysphagia as assessed by the Food Intake Level Scale (FILS) at discharge. RESULTS The study included 143 patients with a mean age of 84.7 ± 7.8 y. Based on the MNA-SF categories at discharge, 127 (88.8%) patients were assigned to the UN group and 16 (11.2%) to the IN group. Patients in the IN group had significantly higher BI and FILS scores than those in the UN group. Multiple regression analysis indicated that improvement in nutritional status was independently associated with BI gain (B = 9.916; β = 0.153; 95% confidence interval [CI], 1.929-11.761; P = 0.017) and FILS gain (B = 1.259; β = 0.167; 95% CI, 1.224-2.814; P = 0.044). CONCLUSIONS Nutritional improvement is associated with improvements in ADL and dysphagia in patients with pneumonia and malnutrition.
Collapse
Affiliation(s)
- Chiharu Uno
- Institute of Innovation for Future Society, Nagoya University, Nagoya City, Aichi, Japan; Department of Community Healthcare and Geriatrics, Nagoya University Graduate School of Medicine, Nagoya City, Aichi, Japan.
| | - Keisuke Maeda
- Department of Palliative and Supportive Medicine, Graduate School of Medicine, Aichi Medical University, Nagakute City, Aichi, Japan
| | - Hidetaka Wakabayashi
- Department of Rehabilitation Medicine, Yokohama City University Medical Center. Yokohama City, Kanagawa, Japan
| | - Shinta Nishioka
- Department of Clinical Nutrition and Food Services, Nagasaki Rehabilitation Hospital, Nagasaki City, Nagasaki, Japan
| | - Nami Ogawa
- Department of Gerontology and Gerodontology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Takayuki Okamoto
- Department of Rehabilitation, Kurashiki Rehabilitation Hospital, Kurashiki City, Okayama, Japan
| | - Kengo Hoyano
- Fukui Health Science University, Faculty of Health Science Department of Rehabilitation Division of Speech-Language-Hearing Therapy, eiheiji-cho, Yoshida-gun, Fukui, Japan
| | - Ryo Momosaki
- Department of Rehabilitation Medicine, Teikyo University School of Medicine University Hospital, Mizonokuchi, Kawasaki city, Kanagawa, Japan
| |
Collapse
|