1
|
Gui M, Lv L, Hu S, Qin L, Wang C. Sarcopenia in Parkinson's disease: from pathogenesis to interventions. Metabolism 2025; 169:156272. [PMID: 40258411 DOI: 10.1016/j.metabol.2025.156272] [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: 01/19/2025] [Revised: 04/04/2025] [Accepted: 04/17/2025] [Indexed: 04/23/2025]
Abstract
Parkinson's disease (PD) and sarcopenia are prevalent age-related conditions that often coexist in affected individuals. Sarcopenia is particularly common among PD patients, with severe cases affecting approximately one in five individuals with the disease. Furthermore, sarcopenia is closely linked to the accelerated progression of PD, diminished quality of life, greater susceptibility to falls and fractures, and increased mortality risk. Although the precise mechanisms remain unclear, numerous studies suggest that factors such as the accumulation of α-Synuclein in skeletal muscle, loss of motor neurons, inflammation, phosphate toxicity, hormonal dysregulation, vitamin D deficiency, intestinal flora imbalances, and dysfunction of the gut-muscle-brain axis contribute to sarcopenia in PD. Understanding these mechanisms provides valuable insights into the relationship between PD and sarcopenia and establishes a foundation for future research and therapeutic strategies. This review examines the mechanisms underlying sarcopenia in PD, methods for its screening and assessment, and potential avenues for future research, including strategies for risk reduction and treatment.
Collapse
Affiliation(s)
- Meilin Gui
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Lingling Lv
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Shenglan Hu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Lixia Qin
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China; Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Chunyu Wang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China; Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, China; Department of Medical Genetics, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha 410011, China.
| |
Collapse
|
2
|
Tari AR, Walker TL, Huuha AM, Sando SB, Wisloff U. Neuroprotective mechanisms of exercise and the importance of fitness for healthy brain ageing. Lancet 2025; 405:1093-1118. [PMID: 40157803 DOI: 10.1016/s0140-6736(25)00184-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 04/01/2025]
Abstract
Ageing is a scientifically fascinating and complex biological occurrence characterised by morphological and functional changes due to accumulated molecular and cellular damage impairing tissue and organ function. Ageing is often accompanied by cognitive decline but is also the biggest known risk factor for Alzheimer's disease, the most common form of dementia. Emerging evidence suggests that sedentary and unhealthy lifestyles accelerate brain ageing, while regular physical activity, high cardiorespiratory fitness (CRF), or a combination of both, can mitigate cognitive impairment and reduce dementia risk. The purpose of this Review is to explore the neuroprotective mechanisms of endurance exercise and highlight the importance of CRF in promoting healthy brain ageing. Key findings show how CRF mediates the neuroprotective effects of exercise via mechanisms such as improved cerebral blood flow, reduced inflammation, and enhanced neuroplasticity. We summarise evidence supporting the integration of endurance exercise that enhances CRF into public health initiatives as a preventive measure against age-related cognitive decline. Additionally, we address important challenges such as lack of long-term studies with harmonised study designs across preclinical and clinical settings, employing carefully controlled and repeatable exercise protocols, and outline directions for future research.
Collapse
Affiliation(s)
- Atefe R Tari
- The Cardiac Exercise Research Group at the Faculty of Medicine and Health Sciences, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology and Clinical Neurophysiology, St Olavs University Hospital, Trondheim, Norway
| | - Tara L Walker
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Aleksi M Huuha
- The Cardiac Exercise Research Group at the Faculty of Medicine and Health Sciences, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology and Clinical Neurophysiology, St Olavs University Hospital, Trondheim, Norway
| | - Sigrid B Sando
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology and Clinical Neurophysiology, St Olavs University Hospital, Trondheim, Norway
| | - Ulrik Wisloff
- The Cardiac Exercise Research Group at the Faculty of Medicine and Health Sciences, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
3
|
Orioli L, Thissen JP. Myokines as potential mediators of changes in glucose homeostasis and muscle mass after bariatric surgery. Front Endocrinol (Lausanne) 2025; 16:1554617. [PMID: 40171198 PMCID: PMC11958187 DOI: 10.3389/fendo.2025.1554617] [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: 01/02/2025] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Myokines are bioactive peptides released by skeletal muscle. Myokines exert auto-, para-, or endocrine effects, enabling them to regulate many aspects of metabolism in various tissues. However, the contribution of myokines to the dramatic changes in glucose homeostasis and muscle mass induced by bariatric surgery has not been established. Our review highlights that myokines such as brain-derived neurotrophic factor (BDNF), meteorin-like protein (Metrnl), secreted protein acidic and rich in cysteine (SPARC), apelin (APLN) and myostatin (MSTN) may mediate changes in glucose homeostasis and muscle mass after bariatric surgery. Our review also identifies myonectin as an interesting candidate for future studies, as this myokine may regulate lipid metabolism and muscle mass after bariatric surgery. These myokines may provide novel therapeutic targets and biomarkers for obesity, type 2 diabetes and sarcopenia.
Collapse
Affiliation(s)
- Laura Orioli
- Research Laboratory of Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Research Laboratory of Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
4
|
Li F, Liu X, Bai N, Li Y, Hou M, Hou Y, Liu Y, Wang X, He Q, Li J. Irisin attenuates liver fibrosis by regulating energy metabolism and HMGB1/β-catenin signaling in hepatic stellate cells. Eur J Pharmacol 2025; 998:177519. [PMID: 40101858 DOI: 10.1016/j.ejphar.2025.177519] [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: 11/25/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Liver fibrosis is characterized by excessive extracellular matrix accumulation during chronic liver disease progression. Hepatic stellate cell (HSC) activation involves metabolic reprogramming, while both HMGB1 and β-catenin pathways have been implicated in HSC activation and liver fibrosis progression. Given irisin's established role in metabolic regulation and emerging evidence of its anti-fibrotic properties, we investigated its effects on HSC activation and liver fibrosis, focusing on potential metabolic regulation through the HMGB1/β-catenin pathway. Using both in vitro HSC-T6 cell culture and in vivo CCl4-induced rat liver fibrosis model, we analyzed irisin's impact on HSC metabolism and fibrosis progression. Our results demonstrated that irisin dose-dependently suppressed HSC-T6 cell viability and glycolytic metabolism, significantly reducing ATP levels, glucose consumption, and lactate production at concentrations of 80-100 nmol/L. Irisin treatment markedly inhibited HSC-T6 cell proliferation and migration while inducing cellular senescence, as evidenced by increased H3K9me3, γ-H2AX, P16, and P21 expression. Mechanistically, irisin systematically downregulated key glycolytic enzymes (HK2, PFK1, PKM2, LDHA) and modulated the HMGB1/β-catenin pathway by reducing both cytoplasmic HMGB1 expression and β-catenin nuclear translocation. In the CCl4-induced rat model, irisin treatment significantly ameliorated liver fibrosis, as evidenced by reduced collagen deposition and α-SMA expression, while improving liver function indicators and decreasing serum fibrosis markers (HA, PIIIP, HMGB1), showing therapeutic effects comparable to colchicine. These findings reveal irisin's anti-fibrotic effects through metabolic regulation and HMGB1/β-catenin pathway modulation, suggesting its potential as a therapeutic agent for liver fibrosis.
Collapse
Affiliation(s)
- Fang Li
- Department of Infectious Diseases, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Xin Liu
- Department of Gastrointestinal Endoscopy, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Ning Bai
- Department of Gastrointestinal Endoscopy, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Ying Li
- Department of Gastrointestinal Endoscopy, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Minna Hou
- Department of Gastrointestinal Endoscopy, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Yali Hou
- Department of Gastrointestinal Endoscopy, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Yunting Liu
- Department of Gastrointestinal Endoscopy, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Xu Wang
- Department of Gastrointestinal Endoscopy, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Qi He
- Department of Gastrointestinal Endoscopy, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Jing Li
- Department of Gastrointestinal Endoscopy, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China.
| |
Collapse
|
5
|
Dai J, Zhao Y, Chen Y, Jiang Y, Sun R, Tang X, Cui Y, Mao H, Peng XG. Irisin reverses high-fat diet-induced metabolic dysfunction via activation of brown adipose tissue in mice. Int J Obes (Lond) 2025:10.1038/s41366-025-01739-z. [PMID: 40082597 DOI: 10.1038/s41366-025-01739-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 02/01/2025] [Accepted: 02/25/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND High-fat diet (HFD) induces negative effects on the activity of interscapular brown adipose tissue (iBAT) and systemic energy metabolism. Irisin, a small hormonal agent known to modulate metabolism has been used for intervening HFD-induced obesity. However, its mechanism of action on iBAT function remains to be fully elucidated. This study sought to investigate whether irisin intervention could restore the thermogenic function of iBAT in mice with HFD-induced obesity, thereby regulating systemic metabolism. METHODS Magnetic resonance imaging (MRI) and positron emission tomography/computed tomography (PET/CT) were used to monitor changes of thermogenic capacity of iBAT and systemic metabolism in mice with HFD-induced obesity and iBAT deficiency during 2-week or 4-week irisin intervention. Pathological and molecular biology analyses were performed on tissue and blood samples. RESULTS Prolonged HFD feeding in mice induced obesity and impaired the thermogenic capacity of iBAT. MRI results showed that irisin intervention for 4-week reduced lipid content in iBAT, increased uncoupling protein 1 (UCP 1) expression and enhanced glucose analogue uptake capacity. These improvements of functions in iBAT activity were accompanied by an improvement in systemic metabolism. The positive effects of irisin appears to be dependent on the length of intervention time. When iBAT was removed, the beneficial effects of irisin were partially suppressed, suggesting that irisin regulates metabolism through the restoration of the thermogenic function of iBAT. CONCLUSIONS HFD results in reduced thermogenic capacity of iBAT, while irisin intervention can effectively restore iBAT function, leading to improvement in overall glucose and lipid metabolism.
Collapse
Affiliation(s)
- Jingyue Dai
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yufei Zhao
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yue Chen
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yang Jiang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Rui Sun
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215002, China
| | - Xingzhe Tang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ying Cui
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Xin-Gui Peng
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
- Department of Radiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 211200, China.
| |
Collapse
|
6
|
Dong Q, Zhu Y, Zhang X, Li L, Yang Y, Liu C, Wen J. Phytochemicals Targeting Mitophagy to Treat Heart Diseases: Retrospective Insights and Prospective Directions. Phytother Res 2025; 39:1592-1614. [PMID: 39912509 DOI: 10.1002/ptr.8448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/07/2025] [Accepted: 01/19/2025] [Indexed: 02/07/2025]
Abstract
Mitophagy is a process by which cells selectively eliminate damaged or dysfunctional mitochondria through the autophagy-lysosome pathway, thereby maintaining mitochondrial quality and cellular homeostasis. This process is closely linked to the onset and progression of various heart diseases. Modern pharmacological research has demonstrated that phytochemicals can regulate mitochondrial homeostasis in cardiomyocytes through multiple mechanisms, influencing mitophagy and protecting cardiomyocytes, which in turn exerts anti-cardiovascular effects. However, the underlying mechanisms of these effects are not yet fully understood. This study summarizes the pharmacological effects and molecular mechanisms of mitophagy in heart diseases, aiming to provide reference for the research and treatment of phytochemicals targeting mitophagy against heart diseases. The results indicated that phytochemicals (such as Berberine, Ginsenoside Rg1, Quercetin, Resveratrol, Baicalein, and so on) can exert preventive and therapeutic effects on heart diseases (such as cardiac toxicity or damage, myocardial ischemia/reperfusion injury, heart failure, heart aging, cardiac hypertrophy, cardiomyopathy, and so on.) via regulating the PINK1/Parkin and FUNDC1-dependent mitophagy pathway. These compounds mainly exert their effects by regulating mitochondrial homeostasis, mitochondrial dynamics, mitochondrial oxidative stress, mitochondrial apoptosis, and mitochondrial energy metabolism. This study provides a reference that phytochemicals have effect on anti-cardiovascular effects by regulating mitophagy. However, further in-depth mechanistic and clinical research are needed in the future.
Collapse
Affiliation(s)
- Qin Dong
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yichan Zhu
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Xinghai Zhang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Lu Li
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yi Yang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Chuan Liu
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Jianxia Wen
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| |
Collapse
|
7
|
Zhang QH, Jin LM, Lin MS, Wang MX, Cui YQ, Ye JX, Xiong YQ, Luo W, Zhu WW, Liang G. FNDC5/Irisin exacerbates APAP-induced acute liver injury through activating JNK/NF-κB and inflammatory response. Acta Pharmacol Sin 2025:10.1038/s41401-025-01509-7. [PMID: 40016523 DOI: 10.1038/s41401-025-01509-7] [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: 09/26/2024] [Accepted: 02/10/2025] [Indexed: 03/01/2025]
Abstract
Acute liver injury (ALI) is associated with high mortality rates. Despite its severity, there are currently no effective interventions, underscoring the urgent need for research on the mechanisms driving ALI progression. Irisin, a hormone derived from its precursor FNDC5, has been shown to play a critical role in some chronic liver diseases. In this study we investigated the role of hepatic FNDC5/Irisin in a mouse model of AILI induced by acetaminophen (APAP, 400 mg/kg, i.p.). The mice were euthanized at 6, 12 and 24 h after APAP injection, then the blood and liver tissues were collected for analyses. By conducting transcriptome sequencing, we identified that both the expression and release of FNDC5/Irisin were significantly increased and highly correlated with AILI. We showed that knockout of Irisin significantly improved APAP-induced tissue damage and hepatocyte death in mouse liver. Conversely, preinjection of recombinant Irisin protein (1 mg·kg-1·d-1, i.p., for 3 days) exacerbated the AILI in FNDC5 knockout mice. RNA-seq analysis revealed that knockout of FNDC5/Irisin reduced inflammatory responses and JNK/NF-κB activation in APAP-treated mouse liver, while exogenous Irisin administration aggravated JNK/NF-κB-mediated inflammation. In primary mouse hepatocytes treated with APAP (15 mM), application of Irisin (100 ng/mL) activated the integrin αV/JNK/NF-κB axis, driving inflammation and oxidative stress. In summary, this study highlights Irisin as a critical regulator in AILI progression. Circulating Irisin could be a novel biomarker for AILI diagnosis, and targeting FNDC5/Irisin could hold promise for the development of novel treatments for AILI.
Collapse
Affiliation(s)
- Qian-Hui Zhang
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, 133002, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Lei-Ming Jin
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Meng-Sha Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Min-Xiu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ya-Qian Cui
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jia-Xi Ye
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yong-Qiang Xiong
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Wu Luo
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
- The Affiliated Cangnan Hospital, Wenzhou Medical University, Wenzhou, 325800, China.
| | - Wei-Wei Zhu
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
- The Affiliated Cangnan Hospital, Wenzhou Medical University, Wenzhou, 325800, China.
| | - Guang Liang
- Department of Cardiology and Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
8
|
Zhao Y, Li J, Ma A, Wang Z, Ni Y, Wu D, Zhou Y, Zhang N, Zhang L, Chang Y, Wang Q. Irisin alleviates hepatic steatosis by activating the autophagic SIRT3 pathway. Chin Med J (Engl) 2025:00029330-990000000-01430. [PMID: 39965865 DOI: 10.1097/cm9.0000000000003427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Disruption of hepatic lipid homeostasis leads to excessive hepatic triglyceride accumulation and the development of metabolic dysfunction-associated steatotic liver disease (MASLD). Autophagy, a critical process in liver lipid metabolism, is impaired in MASLD pathogenesis. Irisin, a skeletal muscle-driven myokine, regulates lipid metabolism, but its impact on hepatic lipid metabolism is not well understood. Here, we aimed to explore the role of irisin in hepatic steatosis and the underlying mechanisms involved. METHODS A high-fat diet (HFD)-induced MASLD mouse model was used, and the recombinant irisin protein, herein referred to as "Irisin", was intraperitoneally administered for 4 weeks to evaluate the effects of irisin on hepatic lipid accumulation. Liver tissues were stained with Oil red O (ORO), and triglyceride (TG) and total cholesterol (TC) contents were measured in serum and liver homogenates. The expression of the autophagosome marker microtubule-associated protein 1 light chain 3 (LC3), the autophagy receptor protein sequestosome-1 (SQSTM1/p62), autophagy initiation complex unc-51-like kinase 1 (ULK1) and the lysosomal functional protein cathepsin B was measured via Western blotting, and the expression of the transcription factor EB (TFEB) was analyzed via immunofluorescence to explore autophagic changes. The effect of irisin on autophagic flux was further evaluated in palmitic acid-induced HepG2 cells by measuring autophagic degradation with chloroquine (CQ), and analyzing the colocalization of LC3 and lysosome-associated protein 1 (LAMP1). The possible mechanism was examined by measuring the expression of the autophagic sirtuin 3 (SIRT3) pathway and further validated using overexpression of SIRT3 with plasmid transfection or siRNA-mediated knockdown. Student's t-test was utilized for statistical analysis. RESULTS Irisin significantly reduces hepatic lipid accumulation in mice fed with HFD, accompanied by enhanced hepatocyte autophagy and upregulation of the SIRT3 pathway. In HepG2 cells, Irisin attenuated palmitic acid-induced lipid accumulation, which was partially dependent on SIRT3 levels. Mechanistically, Irisin treatment upregulated SIRT3 and phosphorylated AMP-activated protein kinase (AMPK), inhibited mammalian target of rapamycin (mTOR) activity, promoted TFEB nucleus translocation, increased cathepsin B expression, enhanced autophagic degradation, and alleviated hepatic steatosis. No significant changes in phosphorylation of ULK1 in the hepatocytes were observed. However, when siRNA was used to knock down SIRT3, the changes of those protein were partially reversed, and hepatic steatosis was further exacerbated. CONCLUSIONS Our findings highlight irisin as a potential therapeutic for hepatic steatosis by modulating autophagy and lipid metabolism, potentially providing a novel therapeutic target for the management of MASLD. Further research is needed to elucidate the underlying mechanisms and explore the potential clinical applications of this approach in the treatment of MASLD.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jia Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Anran Ma
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhihong Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yunzhi Ni
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Di Wu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yue Zhou
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Na Zhang
- Shanghai Innogen Pharmaceutical Co., Ltd., Shanghai 201203, China
| | - Li Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yongsheng Chang
- Key Laboratory of Immune Microenvironment and Disease, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
- Shanghai Innogen Pharmaceutical Co., Ltd., Shanghai 201203, China
| |
Collapse
|
9
|
Huang J, Xu M, Zhang Y, Han J, Zhou H, Wang K. Missense Mutations in FDNC5 Associated with Morphometric Traits and Meat Quality in Hainan Black Goats. Animals (Basel) 2025; 15:565. [PMID: 40003046 PMCID: PMC11851529 DOI: 10.3390/ani15040565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Goats are widely recognized for their adaptability and resource efficiency, making them an excellent choice for sustainable farming. However, the Hainan Black goat (HNBG), a vital breed in southern China's tropical regions, faces significant challenges that threaten its productivity and economic viability. Specifically, young HNBGs exhibit stunted growth and poor muscle development, indicating the breed may have more genetic defects that cause the poor phenotypes. The FNDC5 gene, which encodes the protein irisin, plays a key role in promoting mitochondrial biogenesis and oxidative metabolism by activating critical signaling molecules such as PGC-1α, thereby enhancing muscle endurance and metabolic efficiency. This study aimed to investigate the impact of missense mutations in the FNDC5 gene on growth and meat quality traits in HNBGs. We sequenced a population of HNBGs and identified three SNPs that could lead to amino acid substitutions. Notably, SNP1 (p.119A/V) and SNP2 (p.135R/H) showed strong linkage. Predictions on the structural effects of these mutations indicated that SNP1 (p.119A/V) and SNP3 (p.170W/G) could alter the secondary structure of the FNDC5 protein. Association analyses revealed that SNP1 (p.119A/V) and SNP2 (p.135R/H) were significantly associated with morphometric traits and meat quality. The phenotypic values of SNP1 and SNP2 co-mutants were significantly lower than those of other combined genotypes. Furthermore, gene expression levels of FNDC5 varied notably across individuals with different SNP1 genotypes. These findings suggest that FNDC5-SNP1 (p.119A/V) could serve as a promising genetic marker for selecting HNBGs with improved growth and muscle development, offering a potential pathway for enhancing key economic traits in this breed.
Collapse
Affiliation(s)
- Jing Huang
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China; (J.H.); (M.X.); (J.H.); (H.Z.)
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Mengning Xu
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China; (J.H.); (M.X.); (J.H.); (H.Z.)
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuelang Zhang
- Hainan Institute of Zhejiang University, Sanya 572024, China;
| | - Jiancheng Han
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China; (J.H.); (M.X.); (J.H.); (H.Z.)
| | - Hanlin Zhou
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China; (J.H.); (M.X.); (J.H.); (H.Z.)
- Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Sanya 572024, China
| | - Ke Wang
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China; (J.H.); (M.X.); (J.H.); (H.Z.)
- Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Sanya 572024, China
| |
Collapse
|
10
|
Lee K, Kim M. Evolutionary Insights into Irisin/FNDC5: Roles in Aging and Disease from Drosophila to Mammals. Biomolecules 2025; 15:261. [PMID: 40001564 PMCID: PMC11853655 DOI: 10.3390/biom15020261] [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: 01/14/2025] [Revised: 02/03/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
The Irisin/FNDC5 protein family has emerged as a pivotal link between exercise and the prevention of age-associated diseases. Irisin is highly expressed during exercise from skeletal and cardiac muscle cells, playing a critical role in mediating systemic health benefits through its actions on various tissues. However, Irisin levels decline with age, correlating with a heightened incidence of diseases such as muscle weakness, cardiovascular disorders, and neurodegeneration. Notably, the administration of Irisin has shown significant potential in both preventing and treating these conditions. Recently, an Irisin/FNDC5 homolog was identified in an invertebrate Drosophila model, providing valuable insights into its conserved role in exercise physiology. Importantly, Irisin/FNDC5 has been demonstrated to regulate autophagy-a process essential for clearing excessive nutrients, toxic aggregates, and dysfunctional organelles-in both flies and mammals. Dysregulated autophagy is often implicated in age-related diseases, highlighting its relevance to Irisin/FNDC5's functions. These findings deepen our understanding of Irisin/FNDC5's roles and its potential as a therapeutic target for mitigating aging-related health decline. Further studies are needed to elucidate the precise mechanisms by which Irisin regulates autophagy and its broader impact on physiological aging and related diseases.
Collapse
Affiliation(s)
| | - Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
11
|
Zhang L, Peng Y, Kong Y, Zhang X, Li Z, Jia H. Circulating irisin levels in patients with sarcopenia: a systematic review and meta-analysis. Eur Geriatr Med 2025; 16:5-13. [PMID: 39562482 DOI: 10.1007/s41999-024-01097-5] [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: 03/14/2024] [Accepted: 10/24/2024] [Indexed: 11/21/2024]
Abstract
OBJECTIVE During the aging process, a decrease in irisin levels is associated with numerous bone and muscle diseases. This study aims to provide evidence of circulating irisin levels in patients with sarcopenia. METHODS This systematic review was conducted based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) standard and the recommendations of the Cochrane Collaboration. A comprehensive search was conducted in PubMed, Embase, Web of Science databases, and other sources from their establishment until August 2023. The Review Manager software version 5.4 was used to calculate the standard mean difference (SMD). I2 statistics measured heterogeneity. RESULTS 12 studies involving 2133 participants who met the inclusion criteria were analyzed. We found that irisin levels were significantly lower in patients with sarcopenia (SMD: - 1.28; 95% CI - 1.65, - 0.90; I2 = 92% P < 0.001). Sensitivity analysis confirmed the robustness of this result. The correlation results showed that there was a positive correlation between the levels of circulating irisin and muscle mass (r value 0.62, 95% CI 0.31, 0.81; P < 0.001) and strength (r value 0.47, 95% CI 0.23, 0.66; P < 0.001), but no statistical correlation between irisin and muscle function (The P-values for gait speed and chair test time are 0.5523 and 0.1467, respectively). CONCLUSION No matter the study area, study design, blood samples, or diagnostic criteria, the concentration of circulating irisin in patients with sarcopenia was lower than that in the control group.
Collapse
Affiliation(s)
- Liangchuan Zhang
- School of Public Health, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Yating Peng
- School of Public Health, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Yuan Kong
- School of Public Health, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Xue Zhang
- School of Public Health, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Zetian Li
- School of Public Health, Southwest Medical University, Luzhou City, Sichuan Province, China
| | - Hong Jia
- School of Public Health, Southwest Medical University, Luzhou City, Sichuan Province, China.
- Collaborating Center of the National Institute of Health Data Sciences of China, Southwest Medical University, Luzhou, Sichuan Province, China.
| |
Collapse
|
12
|
Luo X, Wang J, Ju Q, Li T, Bi X. Molecular mechanisms and potential interventions during aging-associated sarcopenia. Mech Ageing Dev 2025; 223:112020. [PMID: 39667622 DOI: 10.1016/j.mad.2024.112020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024]
Abstract
Sarcopenia, a common condition observed in the elderly, presenting a significant public health challenge due to its high prevalence, insidious onset and diverse systemic effects. Despite ongoing research, the precise etiology of sarcopenia remains elusive. Aging-related processes, which included inflammation, oxidative stress, compromised mitochondrial function and apoptosis, have been implicated in its development. Notably, effective pharmacological treatments for sarcopenia are currently lacking, highlighting the necessity for a deeper understanding of its pathogenesis and causative factors to enable proactive interventions. This article is aimed to provide an extensive overview of the pathogenesis of sarcopenia, along with a summary of current treatment and prevention strategies.
Collapse
Affiliation(s)
- Xiaoqin Luo
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Jin Wang
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Qingqing Ju
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Tianyu Li
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Xiuli Bi
- College of Life Science, Liaoning University, Shenyang 110036, China; Key Laboratory for Chronic Diseases Molecular Mechanism Research and Nutritional Intervention of Shenyang, Shenyang 110036, China.
| |
Collapse
|
13
|
Tang N, Li W, Shang H, Yang Z, Chen Z, Shi G. Irisin-mediated KEAP1 degradation alleviates oxidative stress and ameliorates pancreatitis. Immunol Res 2025; 73:37. [PMID: 39821708 DOI: 10.1007/s12026-024-09588-0] [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: 10/16/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025]
Abstract
Oxidative stress (OS) injury is pivotal in acute pancreatitis (AP) pathogenesis, contributing to inflammatory cascades. Irisin, a ubiquitous cytokine, exhibits antioxidant properties. However, the role of irisin in AP remains inconclusive. Our study aims to elucidate irisin expression in AP patients and investigate its mechanism of action to propose a novel treatment strategy for AP. Serum irisin levels in 65 AP patients were quantified using an enzyme-linked immunosorbent assay and correlated with disease severity scores. Core genes implicated in AP-related oxidative stress were identified and screened via bioinformatics analysis. The therapeutic efficacy of irisin in AP was confirmed using a murine cerulein-induced AP model. The intrinsic mechanism of irisin's antioxidative stress action was investigated and verified in pancreatic AR42J cells (Supplementary Fig. 1). Common targets shared by irisin and AP were further validated using a molecular docking model which was constructed for virtual docking analysis. This study investigated alterations in redox status in AP and found a significant reduction in serum irisin levels, correlating inversely with AP severity. In a murine AP model, we showed that irisin triggers an antioxidative stress program via the KEAP1 gene; this process helps reestablish redox balance by decreasing the buildup of reactive oxygen species (ROS) and suppressing the secretion of inflammatory mediators within pancreatic tissues Notably, increased KEAP1 expression counteracted the antioxidative effects of irisin. Our findings unveil a novel therapeutic mechanism for AP, wherein irisin inhibits KEAP1 to alleviate OS. Increasing irisin levels in vivo presents a promising strategy for AP treatment.
Collapse
Affiliation(s)
- Nan Tang
- Dalian Medical University, Dalian, Liaoning, China
- Department of Hepatobiliary Surgery, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, China
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
- First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
| | - Wendi Li
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Hezhen Shang
- Department of Hepatobiliary Surgery, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, China
| | - Zhen Yang
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Zengyin Chen
- Department of Hepatobiliary Surgery, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, China
| | - Guangjun Shi
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China.
- First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
14
|
Plácido AI, Azevedo D, Herdeiro MT, Morgado M, Roque F. Understanding the Role of Irisin in Longevity and Aging: A Narrative Review. EPIDEMIOLOGIA 2025; 6:1. [PMID: 39846531 PMCID: PMC11755480 DOI: 10.3390/epidemiologia6010001] [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: 06/25/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 01/24/2025] Open
Abstract
Irisin is a protein resulting from a proteolytic cleavage of fibronectin type III domain-containing protein 5 (FND5). The ability of irisin to modulate adipocyte and control glucose metabolism in human metabolic diseases gave rise to the hypothesis that irisin could have a pivotal role in aging-related diseases. Although in animal models, increased levels of irisin have been positively associated with better health outcomes, in humans, its role remains controversial. To provide an overview of the main finding on irisin in older adults, a comprehensive search was performed through the MEDLINE-PubMed, Web of Science, Scopus, and Cochrane databases for studies conducted in older adults (≥60 years) published since 2012. After grouping and analyzing the articles based on diseases associated with older adults, the main conclusion of this narrative review is that the included studies did not yield consistent evidence regarding the association between irisin and health or disease in older adults. Further studies are necessary to clarify the effective role of this protein in promoting health and longevity.
Collapse
Affiliation(s)
- Ana I. Plácido
- Biotechnology Research, Innovation and Design for Health Products (BRIDGES), Research Laboratory on Epidemiology and Population Health, Polytechnic of Guarda Av. Dr. Francisco Sá Carneiro 50, 6300-559 Guarda, Portugal; (D.A.); (F.R.)
| | - Daniela Azevedo
- Biotechnology Research, Innovation and Design for Health Products (BRIDGES), Research Laboratory on Epidemiology and Population Health, Polytechnic of Guarda Av. Dr. Francisco Sá Carneiro 50, 6300-559 Guarda, Portugal; (D.A.); (F.R.)
| | - Maria Teresa Herdeiro
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Manuel Morgado
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), 6200-506 Covilhã, Portugal;
- Pharmaceutical Services of Local Health Unit of Cova da Beira (ULS Cova da Beira), 6200-251 Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior (FCS-UBI), 6200-506 Covilhã, Portugal
| | - Fátima Roque
- Biotechnology Research, Innovation and Design for Health Products (BRIDGES), Research Laboratory on Epidemiology and Population Health, Polytechnic of Guarda Av. Dr. Francisco Sá Carneiro 50, 6300-559 Guarda, Portugal; (D.A.); (F.R.)
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), 6200-506 Covilhã, Portugal;
| |
Collapse
|
15
|
Xing S, Ma Y, Song B, Bai M, Wang K, Song W, Cao T, Guo C, Zhang Y, Wang Z, Wang Y. Irisin reshapes bone metabolic homeostasis to delay age-related osteoporosis by regulating the multipotent differentiation of BMSCs via Wnt pathway. Front Mol Biosci 2025; 11:1524978. [PMID: 39840074 PMCID: PMC11746060 DOI: 10.3389/fmolb.2024.1524978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction Bone aging is linked to changes in the lineage differentiation of bone marrow stem cells (BMSCs), which show a heightened tendency to differentiate into adipocytes instead of osteoblasts. The therapeutic potential of irisin in addressing age-related diseases has garnered significant attention. More significantly, irisin has the capacity to enhance bone mass recovery and sustain overall bone health. Its mechanism of action in preventing osteoporosis has generated considerable interest within the research community. Nonetheless, the targeting effect of irisin on age-related osteoporosis and its underlying molecular biological mechanisms remain unclear. Methods The specific role of irisin in osteogenic-adipogenic differentiation in young or aging BMSCs was evaluated by multiple cells staining and quantitative real-time PCR (RT-qPCR) analysis. RNA-seq and protein Western blotting excavated and validated the key pathway by which irisin influences the fate determination of aging BMSCs. The macroscopic and microscopic changes of bone tissue in aging mice were examined using Micro-computed tomography (Micro-CT) and morphological staining. Results It was noted that irisin affected the multilineage differentiation of BMSCs in a manner dependent on the dosage. Simultaneously, the Wnt signaling pathway might be a crucial mechanism through which irisin sustains the bone-fat balance in aging BMSCs and mitigates the decline in pluripotency. In vivo, irisin reduced bone marrow fat deposition in aging mice and effectively alleviating the occurrence of bone loss. Conclusion Irisin mediates the Wnt signaling pathway, thereby influencing the fate determination of BMSCs. In addition, it is essential for preserving metabolic equilibrium in the bone marrow microenvironment and significantly contributes to overall bone health. The findings provide new evidence for the use of iris extract in the treatment of age-related osteoporosis.
Collapse
Affiliation(s)
- Shangman Xing
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yifan Ma
- Division of Cancer Biology, Laboratory Animal Center, Fourth Military Medical University, Xi’an, China
| | - Bing Song
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
- Medicine Research and Experimental center, Gansu University of Chinese Medicine, Lanzhou, China
| | - Min Bai
- Ningxia Medical University College of Traditional Chinese Medicine, Yinchuan, China
| | - Kexin Wang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Wenjing Song
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Tingting Cao
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Chao Guo
- Medicine Research and Experimental center, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yanying Zhang
- Medicine Research and Experimental center, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhandong Wang
- Clinical College of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yongfeng Wang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
- Gansu Medical University School of Basic Medicine, Pingliang, China
| |
Collapse
|
16
|
Chen Q, Wang J, Li K, Luan JQ, Li JM, Wang YT. Irisin in thyroid diseases. Clin Chim Acta 2025; 564:119929. [PMID: 39154700 DOI: 10.1016/j.cca.2024.119929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Irisin, a hormone-like adipo-myokine, has garnered considerable attention in recent years for its potential impact in metabolic diseases. Its physiological effects are similar to those of thyroid hormones, prompting numerous investigations into potential correlations and interactions between irisin and thyroid function through various in vitro and animal experiments. However, existing studies suggest that the relationship between irisin and thyroid diseases is highly complex and multifaceted. In this paper, we have summarized the research results on serum irisin and thyroid function, providing an overview of advancements and constraints in current research on irisin and thyroid hormones. The aim is to offer insights and directions for future clinical trials in this field.
Collapse
Affiliation(s)
- Qi Chen
- Department of outpatient, Shijiazhuang No.8 Retired Cadre Retirement Home of Hebei Military Region, Shijiazhuang 050000, China
| | - Jing Wang
- Department of Cardionephrology, Hospital affiliated to NCO School of Army Military Medical University, Shijiazhuang 050000, China
| | - Kang Li
- Department of oncology, hematology and endocrinology, Hospital affiliated to NCO School of Army Military Medical University, Shijiazhuang 050000, China
| | - Jun-Qin Luan
- Clinical laboratory, Hospital affiliated to NCO School of Army Military Medical University, Shijiazhuang 050000, China
| | - Jing-Mei Li
- Department of oncology, hematology and endocrinology, Hospital affiliated to NCO School of Army Military Medical University, Shijiazhuang 050000, China
| | - Ya-Ting Wang
- Department of oncology, hematology and endocrinology, Hospital affiliated to NCO School of Army Military Medical University, Shijiazhuang 050000, China.
| |
Collapse
|
17
|
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
|
18
|
Berezina TA, Berezin OO, Lichtenauer M, Berezin AE. Predictors for Irreversibility of Contrast-Induced Acute Kidney Injury in Patients with Obesity After Contrast-Enhanced Computed Tomography Coronary Angiography. Adv Ther 2025; 42:293-309. [PMID: 39527336 DOI: 10.1007/s12325-024-03036-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Although contrast-induced (CI) acute kidney injury (AKI) is a common complication in high-risk individuals requiring evaluation with contrast-enhanced angiography, the possible predictors of CI-AKI in patients with obesity are not fully understood. The aim of this study was to elucidate plausible factors associated with the irreversibility of CI-AKI in individuals with obesity undergoing contrast-enhanced computed tomography coronary angiography. METHODS A total of 96 adult patients with obesity and the KDIGO criteria of CI-AKI (increase of serum levels of creatinine ≥ 25% or ≥ 500 µmol/L at 48 h after procedure) were retrospectively screened from the cohort of 1833 patients who underwent iodine contrast medium (ICM)-enhanced computed tomography coronary angiography, and were included in the study. The patients were divided into two cohorts: 96 adult patients with obesity and recovery of CI-AKI in 7 days after initiating of the event, and 57 individuals with irreversibility of CI-AKI. Serum concentrations of conventional biochemistry and urine biomarkers [i.e., hemoglobin, creatinine, high-sensitivity C-reactive protein, urinary albumin/creatinine ratio (UACR)] as well as natriuretic peptide, adropin, apelin, irisin, tumor necrosis factor-alpha (TNF-alpha), were determined at baseline. The levels of creatinine were measured at baseline, at the event, and in 7 days after the event. RESULTS We identified 12 variables, which were associated with irreversibility of CI-AKI: age > 75 years [odds ratio (OR) = 1.22. P = 0.001], male gender (OR = 1.03, P = 0.042), stable coronary artery disease (OR = 1.06, P = 0.048), chronic kidney disease (CKD) 1-3 grade (OR = 1.60, P = 0.001), heart failure with preserved ejection fraction (HFpEF) (OR = 1.07, P = 0.046), baseline estimated GFR < 80 mL/min/1.73 m2 (OR = 1.10, P = 0.040), UACR > 17.5 mg/g Cr (OR = 1.05, P = 0.048), TNF-alpha > 3.11 pg/mL (OR = 1.12, P = 0.001), and adropin < 2.43 ng/mL (OR = 1.18, P = 0.001). After adjustment for CKD and UACR > 17.5 mg/g Cr, only HFpEF (OR = 1.06, P = 0.042) and adropin < 2.43 ng/mL (OR = 1.11, P = 0.001) remained independent predictors of CI-AKI irreversibility. Yet, adropin < 2.43 ng/mL at baseline exerted sufficiently better predictive ability than both HFpEF and preexisting CKD 1-3 grade. CONCLUSION In a multivariate prediction model adjusted for CKD and urinary albumin/creatinine ratio > 17.5 mg/g Cr, low levels of adropin (< 2.43 ng/mL) in individuals with non-morbid obesity together with the presence of HFpEF were independent predictors of CI-AKI irreversibility after ICM-enhanced computed tomography coronary angiography.
Collapse
Affiliation(s)
- Tetiana A Berezina
- Department of Internal Medicine and Nephrology, VitaCenter, Zaporozhye, 69000, Ukraine
| | - Oleksandr O Berezin
- Department of Alter Psychiatry, Luzerne Psychiatry AG, 4915 St., Urban, Switzerland
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020, Salzburg, Austria
| | - Alexander E Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, 5020, Salzburg, Austria.
| |
Collapse
|
19
|
Wu Y, Wu Y, Yu J, Zhang Y, Dai X, Chen J, Sun Y, Yang Y, Zhao K, Xiao Q. Irisin alters D-galactose-induced apoptosis by increasing caveolin-1 expression in C2C12 myoblasts and skeletal muscle fibroblasts. Mol Cell Biochem 2025; 480:577-588. [PMID: 38581552 DOI: 10.1007/s11010-024-04990-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/15/2024] [Indexed: 04/08/2024]
Abstract
Muscle atrophy and skeletal muscle fibrosis are significant pathological manifestations of primary sarcopenia. The regulation of C2C12 myoblast and skeletal muscle fibroblast apoptosis is associated with these pathological changes. Previous studies have indicated that irisin, the cleaved form of fibronectin type III domain-containing protein 5 (FNDC5), can alleviate primary sarcopenia. However, the mechanisms of the effect of irisin in age-related apoptosis remain unknown. Our present research aimed to explore the effect of irisin and the underlying mechanism of D-galactose (D-gal)-induced apoptosis in skeletal muscle fibroblasts and C2C12 myoblasts. We found the opposite effects of D-gal on C2C12 myoblasts and fibroblasts. We also found that irisin suppressed C2C12 cell apoptosis and promoted fibroblast apoptosis. Mechanistically, irisin altered D-gal-induced apoptosis by increasing caveolin-1 expression. Taken together, these findings further demonstrated that irisin is a potential agent that can treat aged-relative muscle atrophy and fibrosis.
Collapse
Affiliation(s)
- Yaoxuan Wu
- Geriatric Diseases Institute of Chengdu, Department of Geriatrics, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611137, China
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
| | - Yongxin Wu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
| | - Jing Yu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
| | - Yingxiao Zhang
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
| | - Xin Dai
- Department of General Practice, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400010, China
| | - Jinliang Chen
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China
| | - Yue Sun
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
| | - Yongxue Yang
- Geriatric Diseases Institute of Chengdu, Department of Geriatrics, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611137, China.
| | - Kexiang Zhao
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China.
| | - Qian Xiao
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
| |
Collapse
|
20
|
Paoletti I, Coccurello R. Irisin: A Multifaceted Hormone Bridging Exercise and Disease Pathophysiology. Int J Mol Sci 2024; 25:13480. [PMID: 39769243 PMCID: PMC11676223 DOI: 10.3390/ijms252413480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
The fibronectin domain-containing protein 5 (FNDC5), or irisin, is an adipo-myokine hormone produced during exercise, which shows therapeutic potential for conditions like metabolic disorders, osteoporosis, sarcopenia, obesity, type 2 diabetes, and neurodegenerative diseases, including Alzheimer's disease (AD). This review explores its potential across various pathophysiological processes that are often considered independent. Elevated in healthy states but reduced in diseases, irisin improves muscle-adipose communication, insulin sensitivity, and metabolic balance by enhancing mitochondrial function and reducing oxidative stress. It promotes osteogenesis and mitigates bone loss in osteoporosis and sarcopenia. Irisin exhibits anti-inflammatory effects by inhibiting NF-κB signaling and countering insulin resistance. In the brain, it reduces amyloid-β toxicity, inflammation, and oxidative stress, enhancing brain-derived neurotrophic factor (BDNF) signaling, which improves cognition and synaptic health in AD models. It also regulates dopamine pathways, potentially alleviating neuropsychiatric symptoms like depression and apathy. By linking physical activity to systemic health, irisin emphasizes its role in the muscle-bone-brain axis. Its multifaceted benefits highlight its potential as a therapeutic target for AD and related disorders, with applications in prevention, in treatment, and as a complement to exercise strategies.
Collapse
Affiliation(s)
- Ilaria Paoletti
- IRCSS Santa Lucia Foundation, European Center for Brain Research, 00143 Rome, Italy;
| | - Roberto Coccurello
- IRCSS Santa Lucia Foundation, European Center for Brain Research, 00143 Rome, Italy;
- Institute for Complex Systems (ISC), National Research Council (C.N.R.), 00185 Rome, Italy
| |
Collapse
|
21
|
Timofte DV, Tudor RC, Mocanu V, Labusca L. Obesity, Osteoarthritis, and Myokines: Balancing Weight Management Strategies, Myokine Regulation, and Muscle Health. Nutrients 2024; 16:4231. [PMID: 39683624 PMCID: PMC11644804 DOI: 10.3390/nu16234231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Obesity and osteoarthritis (OA) are increasingly prevalent conditions that are intricately linked, with each exacerbating the other's pathogenesis and worsening patient outcomes. This review explores the dual impact of obesity on OA, highlighting the role of excessive weight in aggravating joint degeneration and the limitations OA imposes on physical activity, which further perpetuates obesity. The role of muscle tissue, particularly the release of myokines during physical activity, is examined in the context of OA and obesity. Myokines such as irisin, IL-6, and myostatin are discussed for their roles in metabolic regulation, inflammation, and tissue repair, offering insights into their potential therapeutic targets. This review emphasizes the importance of supervised weight management methods in parallel with muscle rehabilitation in improving joint health and metabolic balance. The potential for myokine modulation through targeted exercise and weight loss interventions to mitigate the adverse effects of obesity and OA is also discussed, suggesting avenues for future research and therapy development to reduce the burden of these chronic conditions.
Collapse
Affiliation(s)
- Daniel Vasile Timofte
- Department of Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (D.V.T.); (R.C.T.)
| | - Razvan Cosmin Tudor
- Department of Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (D.V.T.); (R.C.T.)
- Dr. Iacob Czihac Military Emergency Hospital Iasi, General Henri Mathias Berthelot Str. 7-9, 700483 Iași, Romania
| | - Veronica Mocanu
- Department of Morpho-Functional Sciences II (Pathophysiology), Center for Obesity BioBehavioral Experimental Research, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Luminita Labusca
- Department of Orthopedics and Traumatology, “Sf. Spiridon” Emergency Clinical Hospital, 700111 Iasi, Romania;
- National Institute of Research and Development in Technical Physics Iasi, 700050 Iasi, Romania
| |
Collapse
|
22
|
Huang N, Ge M, Liu X, Tian X, Yin P, Bao Z, Cao F, Shyh-Chang N, Dong B, Dai L, Gan Z, Hu P, Qu J, Wang S, Wang H, Xiao Q, Yue R, Yue J, Zhang L, Zhang Y, Zhang H, Zhang W, Liu GH, Pei G, Liu Y, Zhu D, Dong B. A framework of biomarkers for skeletal muscle aging: a consensus statement by the Aging Biomarker Consortium. LIFE MEDICINE 2024; 3:lnaf001. [PMID: 40008206 PMCID: PMC11851484 DOI: 10.1093/lifemedi/lnaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025]
Abstract
The skeletal muscle is an important organ for movement and metabolism in human body, and its physiological aging underlies the occurrence of muscle atrophy and sarcopenia. China has the largest aging population in the world and is facing a grand challenge with how to prevent and treat skeletal muscle aging-related diseases. To address this difficult problem, the Aging Biomarker Consortium (ABC) of China has reached an expert consensus on biomarkers of skeletal muscle aging by synthesizing literatures and insights from scientists and clinicians. This consensus attempts to provide a comprehensive assessment of biomarkers associated with skeletal muscle aging, and proposes a systematic framework to classify them into three dimensions: functional, structural, and humoral. Within each dimension, the experts recommend clinically relevant biomarkers for skeletal muscle aging. This consensus aims to lay the foundation for future research on skeletal muscle aging, facilitating precise prediction, diagnosis, and treatment of skeletal muscle aging and sarcopenia. It is anticipated to make significant contributions to healthy aging of skeletal muscle in the elderly population in China and around the world as well.
Collapse
Affiliation(s)
| | - Ning Huang
- The Center of Gerontology and Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Meiling Ge
- The Center of Gerontology and Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaolei Liu
- The Center of Gerontology and Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xu Tian
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Pengbin Yin
- Department of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Zhijun Bao
- Department of Geriatrics, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Feng Cao
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Ng Shyh-Chang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Biao Dong
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Sichuan Real and Best Biotech Co., Ltd., Chengdu 610041, China
| | - Lunzhi Dai
- National Clinical Research Center for Geriatrics and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhenji Gan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Department of Spine Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing University, Nanjing 210061, China
| | - Ping Hu
- Spine Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200072, China
- Guangzhou Laboratory, Guangzhou 510005, China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510005, China
- The Tenth People’s Hospital Affiliated to Tongji University, Shanghai 200072, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Institute for Stem Cell and Regenerative Medicine, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Si Wang
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Huating Wang
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Department of Orthopedics and Traumatology, Prince of Wales Hospital, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Qian Xiao
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rui Yue
- Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University, Shanghai 200092, China
| | - Jirong Yue
- The Center of Gerontology and Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Licheng Zhang
- Department of Orthopaedic Trauma, the Fourth Medical Center, National Clinical Research Center for Orthopaedics & Sports Rehabilitation in China, Chinese PLA General Hospital, Beijing 100853, China
| | - Yong Zhang
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Hongbo Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
- The SYSU-YSG Joint Laboratory for Skin Health Research, Sun Yat-sen University, Guangzhou 510080, China
- Advanced Medical Technology Center, The First Afiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Gang Pei
- The Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200070, China
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences; TaiKang Center for Life and Medical Sciences; the Institute for Advanced Studies; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China
| | - Dahai Zhu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Birong Dong
- The Center of Gerontology and Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
23
|
Wang YT, Zheng SY, Jiang SD, Luo Y, Wu YX, Naranmandakh S, Li YS, Liu SG, Xiao WF. Irisin in degenerative musculoskeletal diseases: Functions in system and potential in therapy. Pharmacol Res 2024; 210:107480. [PMID: 39490914 DOI: 10.1016/j.phrs.2024.107480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Degenerative musculoskeletal diseases are a class of diseases related to the gradual structural and functional deterioration of muscles, joints, and bones, including osteoarthritis (OA), osteoporosis (OP), sarcopenia (SP), and intervertebral disc degeneration (IDD). As the proportion of aging people around the world increases, degenerative musculoskeletal diseases not only have a multifaceted impact on patients, but also impose a huge burden on the medical industry in various countries. Therefore, it is crucial to find key regulatory factors and potential therapeutic targets. Recent studies have shown that irisin plays an important role in degenerative musculoskeletal diseases, suggesting that it may become a key molecule in the prevention and treatment of degenerative diseases of the musculoskeletal system. Therefore, this review provides a comprehensive description of the release and basic functions of irisin, and summarizes the role of irisin in OA, OP, SP, and IDD from a cellular and tissue perspective, providing comprehensive basis for clinical application. In addition, we summarized the many roles of irisin as a key information molecule in bone-muscle-adipose crosstalk and a regulatory molecule involved in inflammation, senescence, and cell death, and proposed the interesting possibility of irisin in degenerative musculoskeletal diseases.
Collapse
Affiliation(s)
- Yu-Tong Wang
- 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; Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Sheng-Yuan Zheng
- 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
| | - Shi-de Jiang
- The Central Hospital of Yongzhou, Yongzhou 425000, China
| | - Yan Luo
- 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; Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Yu-Xiang Wu
- School of Kinesiology, Jianghan University, Wuhan, Hubei, China
| | - Shinen Naranmandakh
- Department of chemistry, School of Arts and Sciences, National University of Mongolia, Ulaanbaatar 14201, Mongolia
| | - 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.
| | - Shu-Guang Liu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 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.
| |
Collapse
|
24
|
Saponaro F, Bertolini A, Baragatti R, Galfo L, Chiellini G, Saba A, D'Urso G. Myokines and Microbiota: New Perspectives in the Endocrine Muscle-Gut Axis. Nutrients 2024; 16:4032. [PMID: 39683426 DOI: 10.3390/nu16234032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
This review explores the dual role of skeletal muscle as both a mechanical and endocrine organ, highlighting its contributions to overall health and its adaptability to various inputs such as nutrition, hormones, exercise, and injuries. In addition to its role in metabolism and energy conversion, skeletal muscle secretes signalling molecules called myokines (at rest) and exerkines (during/after physical exercise), which communicate with other organs like the brain, the cardiovascular system, and the immune system. Key molecules such as interleukins, irisin, and myostatin are discussed for their roles in mediating muscle health and inter-organ communication. This work also focuses on the muscle-gut axis, emphasising the bidirectional interaction between skeletal muscle and the gut microbiota, a complex ecosystem influencing immune defence, digestion, and metabolism. Muscle activity, particularly exercise, alters the gut microbial composition, promoting beneficial species, while gut-derived metabolites like short-chain fatty acids (SCFAs) impact muscle metabolism, mitochondrial function, and insulin sensitivity. Dysbiosis, or an imbalanced microbiota, can lead to muscle atrophy, inflammation, and metabolic dysfunction. This evidence highlights emerging research into myokines and exerkines as potential therapeutic targets for managing conditions like muscle decline, ageing, and metabolic diseases through muscle-gut interactions.
Collapse
Affiliation(s)
- Federica Saponaro
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| | - Andrea Bertolini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| | - Riccardo Baragatti
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| | - Leonardo Galfo
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| | - Grazia Chiellini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| | - Alessandro Saba
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| | - Giuseppina D'Urso
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy
| |
Collapse
|
25
|
Qiu R, Sun W, Su Y, Sun Z, Fan K, Liang Y, Lin X, Zhang Y. Irisin's emerging role in Parkinson's disease research: A review from molecular mechanisms to therapeutic prospects. Life Sci 2024; 357:123088. [PMID: 39357796 DOI: 10.1016/j.lfs.2024.123088] [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: 06/24/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
Parkinson's disease (PD), a neurodegenerative disorder characterized by impaired motor function, is typically treated with medications and surgery. However, recent studies have validated physical exercise as an effective adjunct therapy, significantly improving both motor and non-motor symptoms in PD patients. Irisin, a myokine, has garnered increasing attention for its beneficial effects on the nervous system. Research has shown that irisin plays a crucial role in regulating metabolic balance, optimizing autophagy, maintaining mitochondrial quality, alleviating oxidative stress and neuroinflammation, and regulating cell death-all processes intricately linked to the pathogenesis of PD. This review examines the mechanisms through which irisin may counteract PD, provides insights into its biological effects, and considers its potential as a target for therapeutic strategies.
Collapse
Affiliation(s)
- Ruqing Qiu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Weilu Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yana Su
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Zhihui Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Kangli Fan
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yue Liang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xiaoyue Lin
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Ying Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
26
|
Chen Y, Sha W, Zhang Y, Kou W, Yang L, Guo R, Li C, Zhao J, Wang Z. Irisin-regulated lncRNAs and their potential regulatory functions in chondrogenic differentiation of human mesenchymal stem cells. Open Med (Wars) 2024; 19:20241073. [PMID: 39588384 PMCID: PMC11587921 DOI: 10.1515/med-2024-1073] [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: 04/08/2024] [Revised: 09/20/2024] [Accepted: 10/01/2024] [Indexed: 11/27/2024] Open
Abstract
Objective Dysregulation of chondrogenic differentiation is associated with osteoarthritis (OA). The myokine irisin is beneficial in OA treatment; yet, the underlying mechanism is not fully understood. Long noncoding RNAs (lncRNAs) act as important regulators of chondrocyte differentiation. This study was conducted to address the role of lncRNAs in mediating irisin-induced chondrocyte differentiation. Methods We investigated the irisin-regulated lncRNA profile change in human mesenchymal stem cells (MSCs) using published whole transcriptome sequencing data. We predicted their potential targets and competitive endogenous RNA (ceRNA) prediction and analyzed their molecular functions using functional enrichment analysis. Results More differentially expressed lncRNAs (DElncRNAs) were observed in irisin-treated samples. The top irisin-induced lncRNAs were associated with OA or chondrogenic differentiation, including XIST, PAX8-AS1, CASC15, LINC01618, and DLX6-AS1. The DEGs co-expressed with DElncRNAs were enriched in skeletal system development, extracellular matrix (ECM) organization, cell adhesion, and inflammation associated pathways. Several lncRNAs likely acted as ceRNAs to regulate downstream mRNAs including ROR2 and SORBS1 in in OA or chondrogenic differentiation. Conclusions We demonstrate the global regulation of lncRNAs by irisin during chondrogenic differentiation of human MSCs. Further study is required to characterize the key irisin-regulated lncRNAs in chondrogenic differentiation.
Collapse
Affiliation(s)
- Yijie Chen
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Shaanxi, 710004, People’s Republic of China
| | - Wenqi Sha
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Shaanxi, 710004, People’s Republic of China
| | - Yifan Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Shaanxi, 710004, People’s Republic of China
| | - Wanyi Kou
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Shaanxi, 710004, People’s Republic of China
| | - Liu Yang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Shaanxi, 710004, People’s Republic of China
| | - Ruixin Guo
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Shaanxi, 710004, People’s Republic of China
| | - Chenyang Li
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Shaanxi, 710004, People’s Republic of China
| | - Junjie Zhao
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Shaanxi, 710004, People’s Republic of China
| | - Zhenghui Wang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, No.157 Xi Wu Road, Xi’anShaanxi, 710004, People’s Republic of China
| |
Collapse
|
27
|
Heo J, Koh HS, Nam CH, Lee DC, Baek JH, Ahn HS, Lee SC. Changes in Muscle Mass and Bone Density and Their Relationship After Total Knee Arthroplasty. J Clin Med 2024; 13:6700. [PMID: 39597845 PMCID: PMC11594649 DOI: 10.3390/jcm13226700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Purpose: To investigate the effects of total knee arthroplasty (TKA) on muscle mass and bone density in end-stage knee osteoarthritis (OA). Methods: This prospective study was conducted on 111 patients with Kellgren-Lawrence grade 4 knee OA who underwent TKA after failing to respond to conservative treatment for more than 3 months at a single institution from June 2022 to May 2023. Appendicular lean mass index (ALMI) and bone mineral density (BMD) were measured using dual-energy X-ray absorptiometry before and every 6 months after surgery. The average follow-up period was 15.5 ± 2.31 months (range, 11.6-24 months). Results: During the follow-up period after TKA, the ALMI increased relatively continuously and consistently. The BMD of the L-spine and proximal femur did not change significantly until 12 months after TKA surgery but began to increase steeply after 12 months and slowed down after 18 months. The increase in muscle mass showed a significant positive correlation with the increase in BMD. Conclusions: Muscle mass gradually increased after TKA for end-stage knee OA, whereas bone density also increased but not until 12 months after surgery. The significant positive correlation between the increase in muscle mass and bone density suggests that the increase in muscle mass after TKA may be one of the causes of the increase in bone density.
Collapse
Affiliation(s)
- Juneyoung Heo
- Joint & Arthritis Research, Department of Neurosurgery, Himchan Hospital, Seoul 07999, Republic of Korea; (J.H.); (H.-S.K.); (D.-C.L.)
| | - Han-Seung Koh
- Joint & Arthritis Research, Department of Neurosurgery, Himchan Hospital, Seoul 07999, Republic of Korea; (J.H.); (H.-S.K.); (D.-C.L.)
| | - Chang Hyun Nam
- Joint & Arthritis Research, Department of Orthopaedic Surgery, Himchan Hospital, Seoul 07999, Republic of Korea; (C.H.N.); (J.-H.B.); (H.S.A.)
| | - Dong-Chan Lee
- Joint & Arthritis Research, Department of Neurosurgery, Himchan Hospital, Seoul 07999, Republic of Korea; (J.H.); (H.-S.K.); (D.-C.L.)
| | - Ji-Hoon Baek
- Joint & Arthritis Research, Department of Orthopaedic Surgery, Himchan Hospital, Seoul 07999, Republic of Korea; (C.H.N.); (J.-H.B.); (H.S.A.)
| | - Hye Sun Ahn
- Joint & Arthritis Research, Department of Orthopaedic Surgery, Himchan Hospital, Seoul 07999, Republic of Korea; (C.H.N.); (J.-H.B.); (H.S.A.)
| | - Su Chan Lee
- Joint & Arthritis Research, Department of Orthopaedic Surgery, Himchan Hospital, Seoul 07999, Republic of Korea; (C.H.N.); (J.-H.B.); (H.S.A.)
| |
Collapse
|
28
|
Bucarey JL, Trujillo-González I, Paules EM, Espinosa A. Myokines and Their Potential Protective Role Against Oxidative Stress in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Antioxidants (Basel) 2024; 13:1363. [PMID: 39594505 PMCID: PMC11591161 DOI: 10.3390/antiox13111363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Myokines, bioactive peptides released by skeletal muscle, have emerged as crucial regulators of metabolic and protective pathways in peripheral tissues, particularly in combating oxidative stress and inflammation. Their plasma concentration significantly increases following exercise, offering valuable insights into the role of physical activity in preventing sarcopenia and mitigating metabolic diseases, including obesity, diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD). This review focuses on discussing the roles of specific myokines in activating intracellular signaling pathways within the liver, which confer protection against steatosis and lipid peroxidation. We detail the mechanism underlying lipid peroxidation and highlight the liver's antioxidant defenses, such as glutathione (GSH) and glutathione peroxidase 4 (GPX4), which are pivotal in reducing ferroptosis. Furthermore, we provide an in-depth analysis of key myokines, including myostatin, brain-derived neurotrophic factor (BDNF), and irisin, among others, and their potential impact on liver function. Finally, we discuss the molecular mechanisms through which these myokines influence oxidate stress and lipid metabolism, emphasizing their capacity to modulate antioxidant responses in the liver. Finally, we underscore the therapeutic potential of exercise as a non-pharmacological intervention to enhance myokine release, thereby preventing the progression of MASD through improved hepatic antioxidant defenses. This review represents a comprehensive perspective on the intersection of exercise, myokine biology, and liver health.
Collapse
Affiliation(s)
- José Luis Bucarey
- School of Medicine, Faculty of Medicine, Universidad de Valparaíso, San Felipe 2172972, Chile;
| | - Isis Trujillo-González
- Nutrition Research Institute, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.T.-G.); (E.M.P.)
| | - Evan M. Paules
- Nutrition Research Institute, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.T.-G.); (E.M.P.)
| | - Alejandra Espinosa
- School of Medicine, Faculty of Medicine, Universidad de Valparaíso, San Felipe 2172972, Chile;
- Center of Interdisciplinary Biomedical and Engineering Research for Health, Universidad de Valparaíso, San Felipe 2172972, Chile
| |
Collapse
|
29
|
Wang T, Zhou D, Hong Z. Adipose tissue in older individuals: a contributing factor to sarcopenia. Metabolism 2024; 160:155998. [PMID: 39128607 DOI: 10.1016/j.metabol.2024.155998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
Sarcopenia is a geriatric syndrome characterized by a functional decline in muscle. The prevalence of sarcopenia increases with natural aging, becoming a serious health problem among elderly individuals. Therefore, understanding the pathology of sarcopenia is critical for inhibiting age-related alterations and promoting health and longevity in elderly individuals. The development of sarcopenia may be influenced by interactions between visceral and subcutaneous adipose tissue and skeletal muscle, particularly under conditions of chronic low-grade inflammation and metabolic dysfunction. This hypothesis is supported by the following observations: (i) accumulation of senescent cells in both adipose tissue and skeletal muscle with age; (ii) gut dysbiosis, characterized by an imbalance in gut microbial communities as the main trigger for inflammation, sarcopenia, and aged adipose tissue; and (iii) microbial dysbiosis, which could impact the onset or progression of a senescent state. Moreover, adipose tissue acts as an endocrine organ, releasing molecules that participate in intricate communication networks between organs. Our discussion focuses on novel adipokines and their role in regulating adipose tissue and muscle, particularly those influenced by aging and obesity, emphasizing their contributions to disease development. On the basis of these findings, we propose that age-related adipose tissue and sarcopenia are disorders characterized by chronic inflammation and metabolic dysregulation. Finally, we explore new potential therapeutic strategies involving specialized proresolving mediator (SPM) G protein-coupled receptor (GPCR) agonists, non-SPM GPCR agonists, transient receptor potential (TRP) channels, antidiabetic drugs in conjunction with probiotics and prebiotics, and compounds designed to target senescent cells and mitigate their pro-inflammatory activity.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China.
| | - Dong Zhou
- Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China
| | - Zhen Hong
- Department of Neurology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China; Institute of Brain Science and Brain-inspired Technology of West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurology, Chengdu Shangjin Nanfu Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
30
|
Lin Y, Shi H, Yang R, Li S, Tang J, Li S. A transcriptomic analysis of incisional hernia based on high-throughput sequencing technology. Hernia 2024; 28:1899-1907. [PMID: 39073735 DOI: 10.1007/s10029-024-03116-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE Incisional hernia is a common postoperative complication; however, few transcriptomic studies have been conducted on it. In this study, we used second-generation high-throughput sequencing to explore the pathogenesis and potential therapeutic targets of incisional hernias. METHODS Superficial fasciae were collected from 15 patients without hernia and 21 patients with an incisional hernia. High-throughput sequencing of the fascia was performed to generate an expression matrix. We analyzed the matrix to identify differentially expressed genes (DEGs) and performed gene ontology and enrichment analyses of these DEGs. Additionally, an external dataset was utilized to identify key DEGs. RESULTS We identified 1,823 DEGs closely associated with extracellular matrix (ECM) imbalance, bacterial inflammatory response, and fibrillar collagen trimerization. TNNT3, CMAY5, ATP1B4, ASB5, CILP, SIX4, FBN1 and FNDC5 were identified as key DEGs at the intersection of the two expression matrices. Moreover, non-alcoholic fatty liver disease-related, TNF, and IL-17 signaling pathways were identified as key enrichment pathways. CONCLUSIONS We identified eight key DEGs and three pathways associated with incisional hernias. Our findings offer new insights into the pathogenesis of incisional hernias and highlight potential targets for their prevention and treatment.
Collapse
Affiliation(s)
- Yiming Lin
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Hekai Shi
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Rongduo Yang
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Shaochun Li
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Jianxiong Tang
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Shaojie Li
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China.
| |
Collapse
|
31
|
Zhao C, Wu Y, Zhu S, Liu H, Xu S. Irisin Protects Musculoskeletal Homeostasis via a Mitochondrial Quality Control Mechanism. Int J Mol Sci 2024; 25:10116. [PMID: 39337601 PMCID: PMC11431940 DOI: 10.3390/ijms251810116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Irisin, a myokine derived from fibronectin type III domain-containing 5 (FNDC5), is increasingly recognized for its protective role in musculoskeletal health through the modulation of mitochondrial quality control. This review synthesizes the current understanding of irisin's impact on mitochondrial biogenesis, dynamics, and autophagy in skeletal muscle, elucidating its capacity to bolster muscle strength, endurance, and resilience against oxidative-stress-induced muscle atrophy. The multifunctional nature of irisin extends to bone metabolism, where it promotes osteoblast proliferation and differentiation, offering a potential intervention for osteoporosis and other musculoskeletal disorders. Mitochondrial quality control is vital for cellular metabolism, particularly in energy-demanding tissues. Irisin's influence on this process is highlighted, suggesting its integral role in maintaining cellular homeostasis. The review also touches upon the regulatory mechanisms of irisin secretion, predominantly induced by exercise, and its systemic effects as an endocrine factor. While the therapeutic potential of irisin is promising, the need for standardized measurement techniques and further elucidation of its mechanisms in humans is acknowledged. The collective findings underscore the burgeoning interest in irisin as a keystone in musculoskeletal health and a candidate for future therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Haiying Liu
- Department of Spinal Surgery, Peking University People’s Hospital, Peking University, Beijing 100871, China
| | - Shuai Xu
- Department of Spinal Surgery, Peking University People’s Hospital, Peking University, Beijing 100871, China
| |
Collapse
|
32
|
Zhu Y, Chi K, Wang J. Mendelian randomization study on association between grip strength and BMD in different age groups. J Bone Miner Metab 2024; 42:564-581. [PMID: 38884649 PMCID: PMC11455795 DOI: 10.1007/s00774-024-01519-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 05/09/2024] [Indexed: 06/18/2024]
Abstract
INTRODUCTION This study aimed to use the Mendelian randomization study method to verify the causal relationship between grip strength and bone mineral density (BMD) in different ages and different parts of the body. MATERIALS AND METHODS The analysis was based on pooled data from genome-wide association studies (GWAS). Hand grip strength (right) was used as the exposure variable and total body bone mineral density (BMD) of different age groups was used as the outcome variable. Single-nucleotide polymorphisms highly correlated with exposure variables were used as instrumental variables. The inverse variance weighted (IVW) method was used as the primary analysis method, and the Mendelian randomization Egger (MR-Egger) regression and weighted median methods were used as supplementary evidence for the IVW results. Horizontal pleiotropy and heterogeneity tests were conducted to ensure the stability of the results. RESULTS Analyzing the GWAS data on osteoporosis as the outcome variable, the IVW analysis showed that osteoporosis risk was associated with decreased grip strength in the 45-60 age group and the risk of declining lumbar spine BMD was associated with decreased grip strength. However, there was no significant correlation between the risk of osteoporosis in other age groups and changes in grip strength. CONCLUSION A causal relationship exists between decreased grip strength and osteoporosis risk in people aged 45-60 years. The risk of BMD declining in the lumbar spine was associated with reduced grip strength.
Collapse
Affiliation(s)
- Yingying Zhu
- Department of Geriatric Medicine, Zhongshan Hospital of Traditional Chinese Medicine, Zhong Shan, 528400, Guangdong Province, China
| | - Kede Chi
- Department of Orthopedics, Zhongshan Hospital of Traditional Chinese Medicine, No.3, Kangxin Road, Zhong Shan, 528400, Guangdong Province, China.
| | - Jiaci Wang
- Department of Geriatric Medicine, Zhongshan Hospital of Traditional Chinese Medicine, Zhong Shan, 528400, Guangdong Province, China
| |
Collapse
|
33
|
Rathor R, Suryakumar G. Myokines: A central point in managing redox homeostasis and quality of life. Biofactors 2024; 50:885-909. [PMID: 38572958 DOI: 10.1002/biof.2054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 03/15/2024] [Indexed: 04/05/2024]
Abstract
Redox homeostasis is a crucial phenomenon that is obligatory for maintaining the healthy status of cells. However, the loss of redox homeostasis may lead to numerous diseases that ultimately result in a compromised quality of life. Skeletal muscle is an endocrine organ that secretes hundreds of myokines. Myokines are peptides and cytokines produced and released by muscle fibers. Skeletal muscle secreted myokines act as a robust modulator for regulating cellular metabolism and redox homeostasis which play a prime role in managing and improving metabolic function in multiple organs. Further, the secretory myokines maintain redox homeostasis not only in muscles but also in other organs of the body via stabilizing oxidants and antioxidant levels. Myokines are also engaged in maintaining mitochondrial dynamics as mitochondria is a central point for the generation of reactive oxygen species (ROS). Ergo, myokines also act as a central player in communicating signals to other organs, including the pancreas, gut, liver, bone, adipose tissue, brain, and skin via their autocrine, paracrine, or endocrine effects. The present review provides a comprehensive overview of skeletal muscle-secreted myokines in managing redox homeostasis and quality of life. Additionally, probable strategies will be discussed that provide a solution for a better quality of life.
Collapse
Affiliation(s)
- Richa Rathor
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Ministry of Defence, Delhi, India
| | - Geetha Suryakumar
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Ministry of Defence, Delhi, India
| |
Collapse
|
34
|
Cui Y, Yu L, Cong W, Jiang S, Qiu X, Wei C, Zheng G, Mao J, Liu R, Patzak A, Persson PB, Chen J, Zhao L, Lai EY. Irisin preserves mitochondrial integrity and function in tubular epithelial cells after ischemia-reperfusion-induced acute kidney injury. Acta Physiol (Oxf) 2024; 240:e14211. [PMID: 39073055 DOI: 10.1111/apha.14211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 06/15/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024]
Abstract
AIMS A myokine secreted by skeletal muscles during exercise called irisin mitigates ischemia-reperfusion (I/R) injury in epithelial cells of various organs by limiting damage to mitochondria. We test whether irisin may preserve the mitochondrial integrity and function in renal tubular epithelial cells and protect against ischemia-reperfusion-induced acute kidney injury (AKI). METHODS We correlated serum irisin levels with serum creatinine and BUN levels from both AKI patients and healthy individuals. In mice with irisin administration, various renal injury markers such as serum creatinine, BUN, kidney injury molecule-1 (Kim-1), and neutrophil gelatinase-associated lipocalin (NGAL), and renal histopathology were assessed after I/R. To identify the potential mechanisms of the protective of irisin's protective effect, we perfused proximal tubules under confocal microscopy and analyzed kidney tissues by qPCR, western blot, and immunohistochemistry. RESULTS Serum irisin correlated inversely with serum creatinine and BUN levels were significantly lower in AKI patients than in healthy subjects. Administering irisin to mice after I/R decreased biomarker levels for AKI including serum creatinine, BUN, Kim-1, NAGL and lessened histological changes. In kidney tissues of mice, irisin upregulated the mitochondrial autophagy marker protein microtubule-associated protein 1 light chain 3 (LC3), the mitochondrial autophagy pathway-related proteins PTEN-induced putative kinase 1 (PINK1) and Parkinson's disease 2 parkin (PARK2) and downregulated the reactive substrate protein sequestosome 1 (P62) and mitochondrial membrane proteins translocase of outer mitochondrial membrane 20 (TOM20) and translocase of inner mitochondrial membrane 23 (TIM23). CONCLUSION Irisin protects against renal I/R injury, which may involve the preservation of mitochondrial integrity and function.
Collapse
Affiliation(s)
- Yu Cui
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Lu Yu
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Wenqi Cong
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Shan Jiang
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingyu Qiu
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunchun Wei
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Gui Zheng
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Jianhua Mao
- Provincial Key Laboratory of Neonatal Diseases, Department of Nephrology, National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruisheng Liu
- Department of Molecular Pharmacology & Physiology, Hypertension and Kidney Research Center, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Andreas Patzak
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pontus B Persson
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jianghua Chen
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Liang Zhao
- Provincial Key Laboratory of Neonatal Diseases, Department of Nephrology, National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - En Yin Lai
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
35
|
Guo M, Shen F, Guo X, Zhang J, Ma Y, Wu X, Zuo H, Yao J, Hu Y, Wang D, Li Y, Li J, Qiu J, Yu J, Meng M, Zheng Y, Chen X, Gong M, Liu K, Jin L, Ren X, Zhang Q, Zhao Y, Gu X, Shen F, Li D, Gao L, Liu C, Zhou F, Li M, Wang J, Ding S, Ma X, Lu J, Xie C, Xiao J, Xu L. BMAL1/PGC1α4-FNDC5/irisin axis impacts distinct outcomes of time-of-day resistance exercise. JOURNAL OF SPORT AND HEALTH SCIENCE 2024; 14:100968. [PMID: 39187065 PMCID: PMC11863284 DOI: 10.1016/j.jshs.2024.100968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/21/2024] [Accepted: 05/15/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Resistance exercise leads to improved muscle function and metabolic homeostasis. Yet how circadian rhythm impacts exercise outcomes and its molecular transduction remains elusive. METHODS Human volunteers were subjected to 4 weeks of resistance training protocols at different times of day to assess training outcomes and their associations with myokine irisin. Based on rhythmicity of Fibronectin type III domain containing 5 (FNDC5/irisin), we trained wild type and FNDC5 knockout mice at late active phase (high FNDC5/irisin level) or late rest phase (low FNDC5/irisin level) to analyze exercise benefits on muscle function and metabolic homeostasis. Molecular analysis was performed to understand the regulatory mechanisms of FNDC5 rhythmicity and downstream signaling transduction in skeletal muscle. RESULTS In this study, we showed that regular resistance exercises performed at different times of day resulted in distinct training outcomes in humans, including exercise benefits and altered plasma metabolomics. We found that muscle FNDC5/irisin levels exhibit rhythmicity. Consistent with human data, compared to late rest phase (low irisin level), mice trained chronically at late active phase (high irisin level) gained more muscle capacity along with improved metabolic fitness and metabolomics/lipidomics profiles under a high-fat diet, whereas these differences were lost in FNDC5 knockout mice. Mechanistically, Basic helix-loop-helix ARNT like 1 (BMAL1) and Peroxisome proliferative activated receptor, gamma, coactivator 1 alpha 4 (PGC1α4) induce FNDC5/irisin transcription and rhythmicity, and the signaling is transduced via αV integrin in muscle. CONCLUSION Together, our results offered novel insights that exercise performed at distinct times of day determines training outcomes and metabolic benefits through the rhythmic regulation of the BMAL1/PGC1α4-FNDC5/irisin axis.
Collapse
Affiliation(s)
- Mingwei Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Fei Shen
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Institute of Physical Education, Jiangsu Normal University, Xuzhou 221116, China
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jun Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ying Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xia Wu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Hui Zuo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jing Yao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yepeng Hu
- Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Dongmei Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yu Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jin Li
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Jin Qiu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jian Yu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Meiyao Meng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ying Zheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xin Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mingkai Gong
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, China
| | - Kailin Liu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, China
| | - Ling Jin
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, China
| | - Xiangyu Ren
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, China
| | - Qiang Zhang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, China
| | - Yu Zhao
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, China
| | - Xuejiang Gu
- Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Feixia Shen
- Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Liangcai Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Chang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Fei Zhou
- Cambridge-Suda Genomic Resource Center, Medical College of Soochow University, Suzhou 215123, China
| | - Mian Li
- Department of Endocrinology and Metabolism, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuzhe Ding
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, China
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jian Lu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, College of Physical Education and Health, East China Normal University, Shanghai 200241, China.
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China.
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
36
|
Berezina TA, Berezin OO, Hoppe UC, Lichtenauer M, Berezin AE. Trajectory of Irisin as a Predictor of Kidney-Related Outcomes in Patients with Asymptomatic Heart Failure. Biomedicines 2024; 12:1827. [PMID: 39200291 PMCID: PMC11352030 DOI: 10.3390/biomedicines12081827] [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: 07/04/2024] [Revised: 07/27/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
The purpose of the study is to elucidate whether irisin is a promising predictive biomarker for kidney-related events in patients with T2DM and concomitant asymptomatic HF. We prospectively enrolled 146 T2DM patients who had either evidence of structural cardiac abnormality or elevated levels of N-terminal brain natriuretic pro-peptide (NT-proBNP) > 125 pmol/mL and followed them for 52 weeks. Structural cardiac abnormalities were used as the minimum from the following criteria: abnormal left ventricular (LV) global longitudinal strain (GLS) < -16%, LV hypertrophy, left atrial volume index > 34 mL/m2, abnormal ratio of early transmitral diastolic filling velocity/early mitral annular velocity ≥ 13 units. All the patients underwent echocardiographic and Doppler examinations by two blinded, highly experienced echocardiographers. NT-proBNP, irisin, TNF-alpha, and hs-CRP were quantified in the serum at baseline, at 26 weeks, and at the end of the study. The kidney-related outcomes consisted of an eGFR reduction by 40% from baseline, or end-stage kidney disease, or kidney replacement therapy. We found that levels of irisin at baseline < 4.15 ng/mL and/or its decrease > 20% from baseline in T2DM patients predicted kidney-related events better than baseline levels/dynamic NT-proBNP and the use of SGLT2 inhibitors. In conclusion, we established that a low baseline level of irisin and its 20% decrease correlated with newly kidney-related events in T2DM patients with asymptomatic HFpEF/HFmrEF.
Collapse
Affiliation(s)
- Tetiana A. Berezina
- Department of Internal Medicine and Nephrology, VitaCenter, 69000 Zaporozhye, Ukraine;
| | - Oleksandr O. Berezin
- Departament of Alter Psychiatrie, Luzerner Psychiatrie AG, 4915 St. Urban, Switzerland
| | - Uta C. Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, 5020 Salzburg, Austria; (U.C.H.); (M.L.)
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, 5020 Salzburg, Austria; (U.C.H.); (M.L.)
| | - Alexander E. Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, 5020 Salzburg, Austria; (U.C.H.); (M.L.)
| |
Collapse
|
37
|
Shen X, Chen Y, Zhang J, Yang M, Huang L, Luo J, Xu L. The association between circulating irisin levels and osteoporosis in women: a systematic review and meta-analysis of observational studies. Front Endocrinol (Lausanne) 2024; 15:1388717. [PMID: 39175571 PMCID: PMC11338845 DOI: 10.3389/fendo.2024.1388717] [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: 02/20/2024] [Accepted: 07/09/2024] [Indexed: 08/24/2024] Open
Abstract
Objective This systematic review and meta-analysis aimed to investigate the association between circulating irisin levels and osteoporosis in women, exploring irisin's potential role in the pathophysiology and management of osteoporosis. Method We searched PubMed, Embase, Web of Science, Cochrane Library, CNKI, WanFang, and VIP databases up to January 2023. The inclusion criteria were observational studies reporting on circulating irisin levels in women. The standardized mean difference (SMD) and correlation coefficients with a 95% confidence interval (CI) were used as the main effect measures under a random-effects model. Heterogeneity was evaluated using the Cochrane Q statistic and the I2 statistics. Subgroup analysis and univariate meta-regression analysis were performed to identify the sources of heterogeneity. The quality of the included study was assessed by the Newcastle-Ottawa Score. The quality of evidence was evaluated using the GRADE system. Publication bias was assessed using Begg's and Egger's test, and the trim-and-fill method. Sensitivity analysis was performed to assess the stability of the results. Results Fifteen studies with a total of 2856 participants met the criteria. The analysis showed significantly lower irisin levels in postmenopausal osteoporotic women compared to non-osteoporotic controls (SMD = -1.66, 95% CI: -2.43 to -0.89, P < 0.0001; I2 = 98%, P < 0.00001) and in postmenopausal individuals with osteoporotic fractures than in non-fractures controls (SMD = -1.25, 95% CI: -2.15 to -0.34, P = 0.007; I2 = 97%, P < 0.00001). Correlation analysis revealed that irisin levels positively correlated with lumbar spine BMD (r = 0.37, 95% CI: 0.18 to 0.54), femoral BMD (r = 0.30, 95% CI: 0.18 to 0.42), and femoral neck BMD (r = 0.31, 95% CI: 0.14 to 0.47) in women. Despite significant heterogeneity, the robustness of the results was supported by using the random effects model and sensitivity analysis. Conclusion The current evidence suggests that lower irisin levels are significantly associated with osteoporosis and fracture in postmenopausal women, suggesting its utility as a potential biomarker for early detection of osteoporosis and therapeutic target. However, further high-quality prospective research controlling for confounding factors is needed to clarify the relationship between irisin levels and osteoporotic outcomes. Systematic review registration https://www.crd.york.ac.uk/PROSPERO, identifier CRD42023410264.
Collapse
Affiliation(s)
- Xiaoyang Shen
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Yan Chen
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Jing Zhang
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Meina Yang
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Lu Huang
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Jiaqi Luo
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Liangzhi Xu
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
38
|
Qiao R, Guo J, Zhang C, Wang S, Fang J, Geng R, Kang SG, Huang K, Tong T. Diabetes-induced muscle wasting: molecular mechanisms and promising therapeutic targets. Crit Rev Food Sci Nutr 2024:1-17. [PMID: 39049742 DOI: 10.1080/10408398.2024.2382348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Diabetes has become a serious public health crisis, presenting significant challenges to individuals worldwide. As the largest organ in the human body, skeletal muscle is a significant target of this chronic disease, yet muscle wasting as a complication of diabetes is still not fully understood and effective treatment methods have yet to be developed. Here, we discuss the targets involved in inducing muscle wasting under diabetic conditions, both validated targets and emerging targets. Diabetes-induced skeletal muscle wasting is known to involve changes in various signaling molecules and pathways, such as protein degradation pathways, protein synthesis pathways, mitochondrial function, and oxidative stress inflammation. Recent studies have shown that some of these present potential as promising therapeutic targets, including the neuregulin 1/epidermal growth factor receptor family, advanced glycation end-products, irisin, ferroptosis, growth differentiation factor 15 and more. This study's investigation and discussion of such pathways and their potential applications provides a theoretical basis for the development of clinical treatments for diabetes-induced muscle wasting and a foundation for continued focus on this disease.
Collapse
Affiliation(s)
- Ruixue Qiao
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Jingya Guo
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Chengmei Zhang
- Guizhou Academy of Testing and Analysis, Guiyang, The People's Republic of China
| | - Sirui Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Jingjing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Ruixuan Geng
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
| | - Seong-Gook Kang
- Department of Food Engineering and Solar Salt Research Center, Mokpo National University, Muangun, Republic of Korea
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, The People's Republic of China
- Beijing Laboratory for Food Quality and Safety, Beijing, The People's Republic of China
| | - Tao Tong
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, The People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, The People's Republic of China
- Beijing Laboratory for Food Quality and Safety, Beijing, The People's Republic of China
| |
Collapse
|
39
|
Chen L, Xuan Y, Zhu Y, Wang J, Tian W, Yang X, Chen W, Chen S, Wang S, Miao Q, Liu Y, Zhang R, Hu C, Zhang Y, Jin L, Yu H. Adipocyte secreted NRG4 ameliorates age-associated metabolic dysfunction. Biochem Pharmacol 2024; 225:116327. [PMID: 38823457 DOI: 10.1016/j.bcp.2024.116327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024]
Abstract
With the progressive aging of society, there is an increasing prevalence of age-related diseases that pose a threat to the elderly's quality of life. Adipose tissue, a vital energy reservoir with endocrine functions, is one of the most vulnerable tissues in aging, which in turn influences systematic aging process, including metabolic dysfunction. However, the underlying mechanism is still poorly understood. In this study, we found that NRG4, a novel adipokine, is obviously decreased in adipocyte tissues and serums during aging. Moreover, delivered recombinant NRG4 protein (rNRG4) into aged mice can ameliorate age-associated insulin resistance, glucose disorders and other metabolic disfunction. In addition, rNRG4 treatment alleviates age-associated hepatic steatosis and sarcopenia, accompanied with altered gene signatures. Together, these results indicate that NRG4 plays a key role in the aging process and is a therapeutic target for the treatment of age-associated metabolic dysfunction.
Collapse
Affiliation(s)
- Liwei Chen
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ye Xuan
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yangyang Zhu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute for Metabolic Disease, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 226001, China
| | - Jinghui Wang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Department of Endocrinology, Xihua Xian People's Hospital, Zhoukou 466000, China
| | - Wen Tian
- Department of Endocrinology, Jinzhou Medical University, Jinzhou 121001, China
| | - Xiaoyue Yang
- Department of Endocrinology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Wei Chen
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Si Chen
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Siyi Wang
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Qizeng Miao
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yahui Liu
- Department of Laboratory Medicine, Shanghai Post and Telecommunications Hospital, China
| | - Rong Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute for Metabolic Disease, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 226001, China.
| | - Yi Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Li Jin
- Department of Endocrinology and Metabolism, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, China.
| | - Haoyong Yu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Clinical Research Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
40
|
Niyonzima YB, Wanjiru DK, Kadokawa H. Exercise-induced muscle hormone "irisin" controls luteinizing hormone and follicle-stimulating hormone secretion by bovine gonadotrophs. Anim Reprod Sci 2024; 266:107516. [PMID: 38823233 DOI: 10.1016/j.anireprosci.2024.107516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Irisin is a hormone secreted by muscle in response to exercise. The irisin receptor (IrisinR) is a heterodimer of integrin alpha V (ITGAV) and integrin beta 5 (ITGB5) subunits. Since irisin may mediate some beneficial effects of exercise on animal reproduction, we tested the hypothesis that bovine gonadotrophs express IrisinR and irisin stimulates luteinizing hormone (LH) and follicle stimulating hormone (FSH) secretion by gonadotrophs. Reverse transcription polymerase chain reaction was used to detect the mRNA expression of both ITGAV and ITGB5 in the anterior pituitary glands (APs) of post pubertal heifers and mouse gonadotroph cell line "LβT2." Western blotting was used to detect protein expression in bovine APs. Immunofluorescence microscopy, utilizing the same antibody, visualized IrisinR on the plasma membrane of majority of gonadotrophs. We prepared AP cells from healthy postpubertal heifers, cultured them for 3.5 d, and treated them with increasing concentrations (0, 0.01, 0.1, 1, or 10 nM) of irisin for 5 min before either no treatment or gonadotropin-releasing hormone (GnRH) stimulation. After 2 h, media were harvested for LH and FSH assays. Irisin (0.1-10 nM) stimulated basal LH and FSH secretion, and these stimulatory effects were inhibited by the extracellular signal-regulated kinase or SMAD pathway inhibitors. In the presence of GnRH, irisin at 0.01-1 nM stimulated LH and FSH secretion. A higher dose of irisin (10 nM), however, suppressed the GnRH-induced LH and FSH levels. In conclusion, bovine gonadotrophs expressed IrisinR, and irisin controlled LH and FSH secretion from bovine gonadotrophs.
Collapse
Affiliation(s)
- Yvan Bienvenu Niyonzima
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken 1677-1, Japan
| | - Denis Karani Wanjiru
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken 1677-1, Japan
| | - Hiroya Kadokawa
- Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi-shi, Yamaguchi-ken 1677-1, Japan.
| |
Collapse
|
41
|
Jasper AA, Shah KH, Karim H, Gujral S, Miljkovic I, Rosano C, Barchowsky A, Sahu A. Regenerative rehabilitation measures to restore tissue function after arsenic exposure. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2024; 30:100529. [PMID: 40191583 PMCID: PMC11970924 DOI: 10.1016/j.cobme.2024.100529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Environmental exposure of arsenic impairs the cardiometabolic profile, skeletal muscle health, and neurological function. Such declining tissue health is observed as early as in one's childhood, where the exposure is prevalent, thereby accelerating the effect of time's arrow. Despite the known deleterious effects of arsenic exposure, there is a paucity of specific treatment plans for restoring tissue function in exposed individuals. In this review, we propose to harness the untapped potential of existing regenerative rehabilitation programs, such as stem cell therapeutics with rehabilitation, acellular therapeutics, and artificial intelligence/robotics technologies, to address this critical gap in environmental toxicology. With regenerative rehabilitation techniques showing promise in other injury paradigms, fostering collaboration between these scientific realms offers an effective means of mitigating the detrimental effects of arsenic on tissue function.
Collapse
Affiliation(s)
- Adam A Jasper
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, USA
| | - Kush H Shah
- The Lake Erie College of Osteopathic Medicine (LECOM), Erie, PA, USA
| | - Helmet Karim
- Department of Psychiatry, University of Pittsburgh, USA
- Department of Bioengineering, University of Pittsburgh, USA
| | - Swathi Gujral
- Department of Psychiatry, University of Pittsburgh, USA
| | - Iva Miljkovic
- Department of Epidemiology, University of Pittsburgh, USA
| | | | - Aaron Barchowsky
- Department of Environmental and Occupational Health, University of Pittsburgh, USA
| | - Amrita Sahu
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, USA
| |
Collapse
|
42
|
Sinder SB, Sharma SV, Shirvaikar IS, Pradhyumnan H, Patel SH, Cabeda Diaz I, Perez GG, Bramlett HM, Raval AP. Impact of menopause-associated frailty on traumatic brain injury. Neurochem Int 2024; 176:105741. [PMID: 38621511 DOI: 10.1016/j.neuint.2024.105741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024]
Abstract
Navigating menopause involves traversing a complex terrain of hormonal changes that extend far beyond reproductive consequences. Menopausal transition is characterized by a decrease in estradiol-17β (E2), and the impact of menopause resonates not only in the reproductive system but also through the central nervous system, musculoskeletal, and gastrointestinal domains. As women undergo menopausal transition, they become more susceptible to frailty, amplifying the risk and severity of injuries, including traumatic brain injury (TBI). Menopause triggers a cascade of changes leading to a decline in muscle mass, accompanied by diminished tone and excitability, thereby restricting the availability of irisin, a crucial hormone derived from muscles. Concurrently, bone mass undergoes reduction, culminating in the onset of osteoporosis and altering the dynamics of osteocalcin, a hormone originating from bones. The diminishing levels of E2 during menopause extend their influence on the gut microbiota, resulting in a reduction in the availability of tyrosine, tryptophan, and serotonin metabolites, affecting neurotransmitter synthesis and function. Understanding the interplay between menopause, frailty, E2 decline, and the intricate metabolisms of bone, gut, and muscle is imperative when unraveling the nuances of TBI after menopause. The current review underscores the significance of accounting for menopause-associated frailty in the incidence and consequences of TBI. The review also explores potential mechanisms to enhance gut, bone, and muscle health in menopausal women, aiming to mitigate frailty and improve TBI outcomes.
Collapse
Affiliation(s)
- Sophie B Sinder
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Sabrina V Sharma
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Isha S Shirvaikar
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Hari Pradhyumnan
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Shahil H Patel
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Indy Cabeda Diaz
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Gina G Perez
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, USA
| |
Collapse
|
43
|
Calvez V, Becherucci G, Covello C, Piccirilli G, Mignini I, Esposto G, Laterza L, Ainora ME, Scaldaferri F, Gasbarrini A, Zocco MA. Navigating the Intersection: Sarcopenia and Sarcopenic Obesity in Inflammatory Bowel Disease. Biomedicines 2024; 12:1218. [PMID: 38927425 PMCID: PMC11200968 DOI: 10.3390/biomedicines12061218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/19/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Inflammatory bowel diseases (IBDs) are intricate systemic conditions that can extend beyond the gastrointestinal tract through both direct and indirect mechanisms. Sarcopenia, characterized by a reduction in muscle mass and strength, often emerges as a consequence of the clinical course of IBDs. Indeed, sarcopenia exhibits a high prevalence in Crohn's disease (52%) and ulcerative colitis (37%). While computed tomography and magnetic resonance imaging remain gold-standard methods for assessing muscle mass, ultrasound is gaining traction as a reliable, cost-effective, and widely available diagnostic method. Muscle strength serves as a key indicator of muscle function, with grip strength test emerging nowadays as the most reliable assessment method. In IBDs, sarcopenia may arise from factors such as inflammation, malnutrition, and gut dysbiosis, leading to the formulation of the 'gut-muscle axis' hypothesis. This condition determines an increased need for surgery with poorer post-surgical outcomes and a reduced response to biological treatments. Sarcopenia and its consequences lead to reduced quality of life (QoL), in addition to the already impaired QoL. Of emerging concern is sarcopenic obesity in IBDs, a challenging condition whose pathogenesis and management are still poorly understood. Resistance exercise and nutritional interventions, particularly those aimed at augmenting protein intake, have demonstrated efficacy in addressing sarcopenia in IBDs. Furthermore, anti-TNF biological therapies showed interesting outcomes in managing this condition. This review seeks to furnish a comprehensive overview of sarcopenia in IBDs, elucidating diagnostic methodologies, pathophysiological mechanisms, and clinical implications and management. Attention will also be paid to sarcopenic obesity, exploring the pathophysiology and possible treatment modalities of this condition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Maria Assunta Zocco
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Catholic University of Rome, 00168 Rome, Italy; (V.C.); (G.B.); (C.C.); (G.P.); (I.M.); (G.E.); (L.L.); (M.E.A.); (F.S.); (A.G.)
| |
Collapse
|
44
|
Du ZY, Zhu HL, Chang W, Zhang YF, Ling Q, Wang KW, Zhang J, Zhang QB, Kan XL, Wang QN, Wang H, Zhou Y. Maternal prednisone exposure during pregnancy elevates susceptibility to osteoporosis in female offspring: The role of mitophagy/FNDC5 alteration in skeletal muscle. JOURNAL OF HAZARDOUS MATERIALS 2024; 469:133997. [PMID: 38508115 DOI: 10.1016/j.jhazmat.2024.133997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024]
Abstract
Maternal exposure to glucocorticoids has been associated with adverse outcomes in offspring. However, the consequences and mechanisms of gestational exposure to prednisone on susceptibility to osteoporosis in the offspring remain unclear. Here, we found that gestational prednisone exposure enhanced susceptibility to osteoporosis in adult mouse offspring. In a further exploration of myogenic mechanisms, results showed that gestational prednisone exposure down-regulated FNDC5/irisin protein expression and activation of OPTN-dependent mitophagy in skeletal muscle of adult offspring. Additional experiments elucidated that activated mitophagy significantly inhibited the expression of FNDC5/irisin in skeletal muscle cells. Likewise, we observed delayed fetal bone development, downregulated FNDC5/irisin expression, and activated mitophagy in fetal skeletal muscle upon gestational prednisone exposure. In addition, an elevated total m6A level was observed in fetal skeletal muscle after gestational prednisone exposure. Finally, gestational supplementation with S-adenosylhomocysteine (SAH), an inhibitor of m6A activity, attenuated mitophagy and restored FNDC5/irisin expression in fetal skeletal muscle, which in turn reversed fetal bone development. Overall, these data indicate that gestational prednisone exposure increases m6A modification, activates mitophagy, and decreases FNDC5/irisin expression in skeletal muscle, thus elevating osteoporosis susceptibility in adult offspring. Our results provide a new perspective on the earlier prevention and treatment of fetal-derived osteoporosis.
Collapse
Affiliation(s)
- Zun-Yu Du
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Hua-Long Zhu
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Wei Chang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Yu-Feng Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Qing Ling
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Kai-Wen Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Jin Zhang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Quan-Bing Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiu-Li Kan
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qu-Nan Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Hua Wang
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China; Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China.
| | - Yun Zhou
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
45
|
Raleigh SM, Orchard KJA. Sarcopenia as a Risk Factor for Alzheimer's Disease: Genetic and Epigenetic Perspectives. Genes (Basel) 2024; 15:561. [PMID: 38790190 PMCID: PMC11121242 DOI: 10.3390/genes15050561] [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: 03/28/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Sarcopenia, defined as the age-associated loss of muscle mass and increased fragility with age, is increasing worldwide. The condition often precedes the development of Alzheimer's disease, thereby decreasing the levels of mobility and physical activity in those affected. Indeed, the loss of muscle mass has, in some studies, been associated with an increased risk of Alzheimer's disease and other dementias. However, a detailed understanding of the interplay between both conditions is not available and needs to be thoroughly addressed. In the following review, we focus on several genes, specifically APOE, BDNF, ACE, FTO, and FNDC5, that have been associated with both conditions. We also discuss the epigenetic regulation of each of these genes along with non-coding RNAs (ncRNAs) that may have a role in the development of both the sarcopenic and Alzheimer's disease phenotypes. Finally, we assert that the application of systems biology will unravel the relationship between sarcopenia and Alzheimer's disease and believe that the prevention of muscle loss in older age will reduce the incidence of debilitating cognitive decline.
Collapse
Affiliation(s)
- Stuart M. Raleigh
- Centre for Health and Life Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Kayleigh J. A. Orchard
- School of Life, Health and Chemical Sciences, Open University, Milton Keynes MK7 6AA, UK;
| |
Collapse
|
46
|
Falsetti I, Palmini G, Donati S, Aurilia C, Iantomasi T, Brandi ML. Irisin and Its Role in Postmenopausal Osteoporosis and Sarcopenia. Biomedicines 2024; 12:928. [PMID: 38672282 PMCID: PMC11048342 DOI: 10.3390/biomedicines12040928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Menopause, an extremely delicate phase in a woman's life, is characterized by a drop in estrogen levels. This decrease has been associated with the onset of several diseases, including postmenopausal osteoporosis and sarcopenia, which often coexist in the same person, leading to an increased risk of fractures, morbidity, and mortality. To date, there are no approved pharmacological treatments for sarcopenia, while not all of those approved for postmenopausal osteoporosis are beneficial to muscles. In recent years, research has focused on the field of myokines, cytokines, or peptides secreted by skeletal muscle fibers following exercise. Among these, irisin has attracted great interest as it possesses myogenic properties but at the same time exerts anabolic effects on bone and could therefore represent the link between muscle and bone. Therefore, irisin could represent a new therapeutic strategy for the treatment of osteoporosis and also serve as a new biomarker of sarcopenia, thus facilitating diagnosis and pharmacological intervention. The purpose of this review is to provide an updated summary of what we know about the role of irisin in postmenopausal osteoporosis and sarcopenia.
Collapse
Affiliation(s)
- Irene Falsetti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (I.F.); (S.D.); (C.A.); (T.I.)
| | - Gaia Palmini
- Fondazione Italiana Ricerca Sulle Malattie dell’Osso (F.I.R.M.O Onlus), 50129 Florence, Italy;
| | - Simone Donati
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (I.F.); (S.D.); (C.A.); (T.I.)
| | - Cinzia Aurilia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (I.F.); (S.D.); (C.A.); (T.I.)
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (I.F.); (S.D.); (C.A.); (T.I.)
| | - Maria Luisa Brandi
- Fondazione Italiana Ricerca Sulle Malattie dell’Osso (F.I.R.M.O Onlus), 50129 Florence, Italy;
| |
Collapse
|
47
|
Guo Q, Luo Q, Song G. Control of muscle satellite cell function by specific exercise-induced cytokines and their applications in muscle maintenance. J Cachexia Sarcopenia Muscle 2024; 15:466-476. [PMID: 38375571 PMCID: PMC10995279 DOI: 10.1002/jcsm.13440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/05/2024] [Accepted: 01/14/2024] [Indexed: 02/21/2024] Open
Abstract
Exercise is recognized to play an observable role in improving human health, especially in promoting muscle hypertrophy and intervening in muscle mass loss-related diseases, including sarcopenia. Recent rapid advances have demonstrated that exercise induces the release of abundant cytokines from several tissues (e.g., liver, muscle, and adipose tissue), and multiple cytokines improve the functions or expand the numbers of adult stem cells, providing candidate cytokines for alleviating a wide range of diseases. Muscle satellite cells (SCs) are a population of muscle stem cells that are mitotically quiescent but exit from the dormancy state to become activated in response to physical stimuli, after which SCs undergo asymmetric divisions to generate new SCs (stem cell pool maintenance) and commit to later differentiation into myocytes (skeletal muscle replenishment). SCs are essential for the postnatal growth, maintenance, and regeneration of skeletal muscle. Emerging evidence reveals that exercise regulates muscle function largely via the exercise-induced cytokines that govern SC potential, but this phenomenon is complicated and confusing. This review provides a comprehensive integrative overview of the identified exercise-induced cytokines and the roles of these cytokines in SC function, providing a more complete picture regarding the mechanism of SC homeostasis and rejuvenation therapies for skeletal muscle.
Collapse
Affiliation(s)
- Qian Guo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| |
Collapse
|
48
|
Wu Q, Liu Z, Li B, Liu YE, Wang P. Immunoregulation in cancer-associated cachexia. J Adv Res 2024; 58:45-62. [PMID: 37150253 PMCID: PMC10982873 DOI: 10.1016/j.jare.2023.04.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/31/2023] [Accepted: 04/26/2023] [Indexed: 05/09/2023] Open
Abstract
BACKGROUND Cancer-associated cachexia is a multi-organ disorder associated with progressive weight loss due to a variable combination of anorexia, systemic inflammation and excessive energy wasting. Considering the importance of immunoregulation in cachexia, it still lacks a complete understanding of the immunological mechanisms in cachectic progression. AIM OF REVIEW Our aim here is to describe the complex immunoregulatory system in cachexia. We summarize the effects and translational potential of the immune system on the development of cancer-associated cachexia and we attempt to conclude with thoughts on precise and integrated therapeutic strategies under the complex immunological context of cachexia. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three main key concepts. First, we highlight the inflammatory factors and additional mediators that have been identified to modulate this syndrome. Second, we decipher the potential role of immune checkpoints in tissue wasting. Third, we discuss the multilayered insights in cachexia through the immunometabolic axis, immune-gut axis and immune-nerve axis.
Collapse
Affiliation(s)
- Qi Wu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University.
| | - Zhou Liu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Bei Li
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Yu-E Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University.
| |
Collapse
|
49
|
Delfan M, Saeidi A, Supriya R, Escobar KA, Laher I, Heinrich KM, Weiss K, Knechtle B, Zouhal H. Enhancing cardiometabolic health: unveiling the synergistic effects of high-intensity interval training with spirulina supplementation on selected adipokines, insulin resistance, and anthropometric indices in obese males. Nutr Metab (Lond) 2024; 21:11. [PMID: 38454429 PMCID: PMC10921712 DOI: 10.1186/s12986-024-00785-0] [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: 12/05/2023] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
This study investigated the combined effects of 12 weeks of high-intensity interval training (HIIT) and spirulina supplementation on adipokine levels, insulin resistance, anthropometric indices, and cardiorespiratory fitness in 44 obese males (aged 25-40 years). The participants were randomly assigned to one of four groups: control (CG), supplement (SG), training (TG), or training plus supplement (TSG). The intervention involved daily administration of either spirulina or a placebo and HIIT three times a week for the training groups. Anthropometric indices, HOMA-IR, VO2peak, and circulating adipokines (asprosin and lipocalin2, omentin-1, irisin, and spexin) were measured before and after the 12-week intervention. Post-intervention analysis indicated differences between the CG and the three interventional groups for body weight, fat-free mass (FFM), percent body fat (%BF), HOMA-IR, and adipokine levels (p < 0.05). TG and SG participants had increased VO2peak (p < 0.05). Spirulina supplementation with HIIT increased VO2peak, omentin-1, irisin, and spexin, while causing decreases in lipocalin-2 and asprosin levels and improvements in body composition (weight, %fat), BMI, and HOMA-IR. Notably, the combination of spirulina and HIIT produced more significant changes in circulating adipokines and cardiometabolic health in obese males compared to either supplementation or HIIT alone (p < 0.05). These findings highlight the synergistic benefits of combining spirulina supplementation with HIIT, showcasing their potential in improving various health parameters and addressing obesity-related concerns in a comprehensive manner.
Collapse
Affiliation(s)
- Maryam Delfan
- Department of Exercise Physiology, Faculty of Sport Sciences, Alzahra University, Tehran, Iran.
| | - Ayoub Saeidi
- Department of Physical Education and Sport Sciences, Faculty of Humanities and Social Sciences, University of Kurdistan, Sanandaj, Kurdistan, Iran
| | - Rashmi Supriya
- Centre for Health and Exercise Science Research, SPEH, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Kurt A Escobar
- Department of Kinesiology, California State University, Long Beach, CA, 90840, USA
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology, and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Katie M Heinrich
- Department of Kinesiology, Kansas State University, Manhattan, KS, 66502, USA
| | - Katja Weiss
- Institute of Primary Care, University of Zurich, Zurich, Switzerland
| | - Beat Knechtle
- Institute of Primary Care, University of Zurich, Zurich, Switzerland.
- Medbase St. Gallen Am Vadianplatz, Vadianstrasse 26, St. Gallen, 9001, Switzerland.
| | - Hassane Zouhal
- Univ Rennes, M2S (Laboratoire Mouvement, Sport, Rennes, Santé, EA 1274, F-35000, France.
- Institut International des Sciences du Sport (2I2S), Irodouer, 35850, France.
| |
Collapse
|
50
|
Lei M, Feng T, Zhang M, Chang F, Liu J, Sun B, Chen M, Li Y, Zhang L, Tang P, Yin P. CHRONIC CRITICAL ILLNESS-INDUCED MUSCLE ATROPHY: INSIGHTS FROM A TRAUMA MOUSE MODEL AND POTENTIAL MECHANISM MEDIATED VIA SERUM AMYLOID A. Shock 2024; 61:465-476. [PMID: 38517246 DOI: 10.1097/shk.0000000000002322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
ABSTRACT Background: Chronic critical illness (CCI), which was characterized by persistent inflammation, immunosuppression, and catabolism syndrome (PICS), often leads to muscle atrophy. Serum amyloid A (SAA), a protein upregulated in critical illness myopathy, may play a crucial role in these processes. However, the effects of SAA on muscle atrophy in PICS require further investigation. This study aims to develop a mouse model of PICS combined with bone trauma to investigate the mechanisms underlying muscle weakness, with a focus on SAA. Methods: Mice were used to examine the effects of PICS after bone trauma on immune response, muscle atrophy, and bone healing. The mice were divided into two groups: a bone trauma group and a bone trauma with cecal ligation and puncture group. Tibia fracture surgery was performed on all mice, and PICS was induced through cecal ligation and puncture surgery in the PICS group. Various assessments were conducted, including weight change analysis, cytokine analysis, hematological analysis, grip strength analysis, histochemical staining, and immunofluorescence staining for SAA. In vitro experiments using C2C12 cells (myoblasts) were also conducted to investigate the role of SAA in muscle atrophy. The effects of inhibiting receptor for advanced glycation endproducts (RAGE) or JAK2 on SAA-induced muscle atrophy were examined. Bioinformatic analysis was conducted using a dataset from the GEO database to identify differentially expressed genes and construct a coexpression network. Results: Bioinformatic analysis confirmed that SAA was significantly upregulated in muscle tissue of patients with intensive care unit-induced muscle atrophy. The PICS animal models exhibited significant weight loss, spleen enlargement, elevated levels of proinflammatory cytokines, and altered hematological profiles. Evaluation of muscle atrophy in the animal models demonstrated decreased muscle mass, grip strength loss, decreased diameter of muscle fibers, and significantly increased expression of SAA. In vitro experiment demonstrated that SAA decreased myotube formation, reduced myotube diameter, and increased the expression of muscle atrophy-related genes. Furthermore, SAA expression was associated with activation of the FOXO signaling pathway, and inhibition of RAGE or JAK2/STAT3-FOXO signaling partially reversed SAA-induced muscle atrophy. Conclusions: This study successfully develops a mouse model that mimics PICS in CCI patients with bone trauma. Serum amyloid A plays a crucial role in muscle atrophy through the JAK2/STAT3-FOXO signaling pathway, and targeting RAGE or JAK2 may hold therapeutic potential in mitigating SAA-induced muscle atrophy.
Collapse
|