1
|
Wang S, Pan Y, Pang Q, Zhang A. Irisin Ameliorates Muscle Atrophy by Inhibiting the Upregulation of the Ubiquitin‒Proteasome System in Chronic Kidney Disease. Calcif Tissue Int 2024; 115:712-724. [PMID: 39283327 DOI: 10.1007/s00223-024-01283-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/29/2024] [Indexed: 11/03/2024]
Abstract
Muscle atrophy is a common complication of chronic kidney disease (CKD). Irisin, a novel muscle cytokine, protects against muscle atrophy, but its specific role in CKD-associated muscle atrophy requires further elucidation. Because the ubiquitin-proteasome system (UPS) plays an important role in CKD muscle atrophy, our study will explore whether irisin affects UPS and alleviate CKD-associated muscle atrophy. In this study, an adenine-fed mouse model of CKD and urotension II (UII)-induced C2C12 myotubes were used as in vivo and in vitro models of muscle atrophy. The results showed that renal function, mouse weight, and the cross-sectional area (CSA) of skeletal muscles were significantly improved in CKD mice treated with irisin. Moreover, irisin effectively mitigated the decreases in phosphorylated Forkhead box O 3a (p-FOXO3A) levels and increases in the levels of E3 ubiquitin ligases, such as muscle RING finger 1 (MuRF1) and muscle atrophy F-box (MAFbx/atrogin1), in both the muscles of CKD mice and UII-induced C2C12 myotubes. In addition, irisin significantly increased the expression levels of myogenic differentiation factor D (MyoD) in the muscles of CKD mice. Our study is the first to demonstrate that irisin ameliorates skeletal muscle atrophy by inhibiting UPS upregulation and improving satellite cell differentiation in CKD.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Fibronectins/metabolism
- Mice, Inbred C57BL
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/drug effects
- Muscular Atrophy/metabolism
- Proteasome Endopeptidase Complex/metabolism
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/drug therapy
- Renal Insufficiency, Chronic/pathology
- Ubiquitin/metabolism
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Shiyuan Wang
- Department of Nephrology, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 10000, China
| | - Yajing Pan
- Department of Nephrology, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 10000, China
| | - Qi Pang
- Department of Nephrology, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 10000, China
| | - Aihua Zhang
- Department of Nephrology, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 10000, China.
| |
Collapse
|
2
|
Du J, Zhao X, Ding X, Han Q, Duan Y, Ren Q, Wang H, Song C, Wang X, Zhang D, Zhu H. The Role of the Gut Microbiota in Complications among Hemodialysis Patients. Microorganisms 2024; 12:1878. [PMID: 39338552 PMCID: PMC11434415 DOI: 10.3390/microorganisms12091878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
The composition of the gut microbiota varies among end-stage renal disease (ESRD) patients on the basis of their mode of renal replacement therapy (RRT), with notably more pronounced dysbiosis occurring in those undergoing hemodialysis (HD). Interventions such as dialysis catheters, unstable hemodynamics, strict dietary restrictions, and pharmacotherapy significantly alter the intestinal microenvironment, thus disrupting the gut microbiota composition in HD patients. The gut microbiota may influence HD-related complications, including cardiovascular disease (CVD), infections, anemia, and malnutrition, through mechanisms such as bacterial translocation, immune regulation, and the production of gut microbial metabolites, thereby affecting both the quality of life and the prognosis of patients. This review focuses on alterations in the gut microbiota and its metabolites in HD patients. Additionally, understanding the impact of the gut microbiota on the complications of HD could provide insights into the development of novel treatment strategies to prevent or alleviate complications in HD patients.
Collapse
Affiliation(s)
- Junxia Du
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Xiaolin Zhao
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xiaonan Ding
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Qiuxia Han
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yingjie Duan
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Qinqin Ren
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Haoran Wang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Chenwen Song
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Xiaochen Wang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
- Medical School of Chinese People's Liberation Army, Beijing 100853, China
| | - Dong Zhang
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Hanyu Zhu
- Department of Nephrology, First Medical Center of Chinese People's Liberation Army General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| |
Collapse
|
3
|
Zou Y, Tang X, Yang S, Chen Z, Liu B, Zhou Z, Peng X, Tang C. New insights into the function of the NLRP3 inflammasome in sarcopenia: mechanism and therapeutic strategies. Metabolism 2024; 158:155972. [PMID: 38972476 DOI: 10.1016/j.metabol.2024.155972] [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: 03/05/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Sarcopenia is one of the most common skeletal muscle disorders and is characterized by infirmity and disability. While extensive research has focused on elucidating the mechanisms underlying the progression of sarcopenia, further comprehensive insights into its pathogenesis are necessary to identify new preventive and therapeutic approaches. The involvement of inflammasomes in sarcopenia is widely recognized, with particular emphasis on the NLRP3 (NLR family pyrin domain containing 3) inflammasome. In this review, we aim to elucidate the underlying mechanisms of the NLRP3 inflammasome and its relevance in sarcopenia of various etiologies. Furthermore, we highlight interventions targeting the NLRP3 inflammasome in the context of sarcopenia and discuss the current limitations of our knowledge in this area.
Collapse
Affiliation(s)
- Yunyi Zou
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Xiangbin Tang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Siyuan Yang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Zhanglin Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Bin Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Zuoqiong Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Xiyang Peng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China.
| | - Changfa Tang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China.
| |
Collapse
|
4
|
Ibrahim Z, Khan NA, Siddiqui R, Qaisar R, Marzook H, Soares NC, Elmoselhi AB. Gut matters in microgravity: potential link of gut microbiota and its metabolites to cardiovascular and musculoskeletal well-being. Nutr Metab (Lond) 2024; 21:66. [PMID: 39123239 PMCID: PMC11316329 DOI: 10.1186/s12986-024-00836-6] [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: 02/08/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
The gut microbiota and its secreted metabolites play a significant role in cardiovascular and musculoskeletal health and diseases. The dysregulation of the intestinal microbiota poses a significant threat to cardiovascular and skeletal muscle well-being. Nonetheless, the precise molecular mechanisms underlying these changes remain unclear. Furthermore, microgravity presents several challenges to cardiovascular and musculoskeletal health compromising muscle strength, endothelial dysfunction, and metabolic changes. The purpose of this review is to critically examine the role of gut microbiota metabolites on cardiovascular and skeletal muscle functions and dysfunctions. It also explores the molecular mechanisms that drive microgravity-induced deconditioning in both cardiovascular and skeletal muscle. Key findings in this review highlight that several alterations in gut microbiota and secreted metabolites in microgravity mirror characteristics seen in cardiovascular and skeletal muscle diseases. Those alterations include increased levels of Firmicutes/Bacteroidetes (F/B) ratio, elevated lipopolysaccharide levels (LPS), increased in para-cresol (p-cresol) and secondary metabolites, along with reduction in bile acids and Akkermansia muciniphila bacteria. Highlighting the potential, modulating gut microbiota in microgravity conditions could play a significant role in mitigating cardiovascular and skeletal muscle diseases not only during space flight but also in prolonged bed rest scenarios here on Earth.
Collapse
Affiliation(s)
- Zeinab Ibrahim
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Naveed A Khan
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Ruqaiyyah Siddiqui
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS,, UK
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Rizwan Qaisar
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hezlin Marzook
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Nelson C Soares
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid university of Medicine and Health Sciences, Dubai, 0000, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Av Padre Cruz, Lisbon, 1649-016, Portugal
| | - Adel B Elmoselhi
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE.
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
5
|
Veloso MP, Coelho VA, Sekercioglu N, Moyses RMA, Elias RM. Phosphate is associated with frailty in older patients with chronic kidney disease not on dialysis. Int Urol Nephrol 2024; 56:2725-2731. [PMID: 38498271 DOI: 10.1007/s11255-024-03985-y] [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: 11/14/2023] [Accepted: 02/12/2024] [Indexed: 03/20/2024]
Abstract
PURPOSE Frailty is common in older patients with chronic kidney disease (CKD) and has been considered an independent risk factor for adverse clinical outcomes in this population. CKD-associated mineral and bone metabolism (CKD-MBD) increases energy expenditure and causes malnutrition and inflammation leading to frailty. We investigated whether CKD-MBD markers and energy metabolism are associated with frailty in patients with advanced CKD on conservative management. METHODS In this cross-sectional study, we investigated factors associated with frailty in a sample of 75 patients ≥ 65 years, with stage 4 or 5 CKD. Collected data included age, sex, body mass index, physical activity status, educational level, Charlson Comorbidity Index, and laboratory markers. Frailty was evaluated according to Fried's classification. RESULTS Frailty was observed in 51.3% and pre-frailty in 47.3%. The frail population was significantly older, with a high proportion of females, more inactive, had lower educational levels, spent a long time sitting throughout the day, and had higher phosphate and fibroblast growth factor 21 (FGF-21). In the multivariate logistic analysis age (odds ratio 1.13, p = 0.026) and phosphate (odds ratio 3.38, p = 0.021) remained independently associated with frailty. CONCLUSION Serum phosphate seems to be a toxin associated with the frailty phenotype in older patients with CKD. Whether strategies to decrease serum phosphate would reduce the risk of frailty in this population deserves further evaluation.
Collapse
Affiliation(s)
- Mariana P Veloso
- Department of Medicine. Nephrology Service. Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo., São Paulo, Brazil
| | - Venceslau A Coelho
- Department of Medicine. Geriatric Division. Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo, São Paulo, Brazil
| | - Nigar Sekercioglu
- Department of Health Evidence and Impact, McMaster University, Hamilton, Canada
- Department of Medicine, Division of Nephrology, Health Sciences University, Istanbul, Turkey
| | - Rosa M A Moyses
- Department of Medicine. Nephrology Service. Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo., São Paulo, Brazil
| | - Rosilene M Elias
- Department of Medicine. Nephrology Service. Hospital das Clinicas HCFMUSP, Universidade de Sao Paulo., São Paulo, Brazil.
- Universidade Nove de Julho. UNINOVE, São Paulo, Brazil.
| |
Collapse
|
6
|
Wakamatsu T, Yamamoto S, Yoshida S, Narita I. Indoxyl Sulfate-Induced Macrophage Toxicity and Therapeutic Strategies in Uremic Atherosclerosis. Toxins (Basel) 2024; 16:254. [PMID: 38922148 PMCID: PMC11209365 DOI: 10.3390/toxins16060254] [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: 04/14/2024] [Revised: 05/18/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
Cardiovascular disease (CVD) frequently occurs in patients with chronic kidney disease (CKD), particularly those undergoing dialysis. The mechanisms behind this may be related to traditional risk factors and CKD-specific factors that accelerate atherosclerosis and vascular calcification in CKD patients. The accumulation of uremic toxins is a significant factor in CKD-related systemic disorders. Basic research suggests that indoxyl sulfate (IS), a small protein-bound uremic toxin, is associated with macrophage dysfunctions, including increased oxidative stress, exacerbation of chronic inflammation, and abnormalities in lipid metabolism. Strategies to mitigate the toxicity of IS include optimizing gut microbiota, intervening against the abnormality of intracellular signal transduction, and using blood purification therapy with higher efficiency. Further research is needed to examine whether lowering protein-bound uremic toxins through intervention leads to a reduction in CVD in patients with CKD.
Collapse
Affiliation(s)
- Takuya Wakamatsu
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; (T.W.); (S.Y.); (I.N.)
- Ohgo Clinic, Maebashi 371-0232, Japan
| | - Suguru Yamamoto
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; (T.W.); (S.Y.); (I.N.)
| | - Shiori Yoshida
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; (T.W.); (S.Y.); (I.N.)
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; (T.W.); (S.Y.); (I.N.)
| |
Collapse
|
7
|
Lu W, Cheng S, Xu J, Xiao Z, Yu Y, Xie Q, Fang Y, Chen R, Shen B, Xie Y, Ding X. Roles of AhR/CYP1s signaling pathway mediated ROS production in uremic cardiomyopathy. Toxicol Lett 2024; 396:81-93. [PMID: 38670245 DOI: 10.1016/j.toxlet.2024.04.005] [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: 10/16/2023] [Revised: 03/24/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
PURPOSE Uremic cardiomyopathy (UCM) is the leading cause of chronic kidney disease (CKD) related mortality. Uremic toxins including indoxyl sulfate (IS) play important role during the progression of UCM. This study was to explore the underlying mechanism of IS related myocardial injury. METHODS UCM rat model was established through five-sixths nephrectomy to evaluate its effects on blood pressure, cardiac impairment, and histological changes using echocardiography and histological analysis. Additionally, IS was administered to neonatal rat cardiomyocytes (NRCMs) and the human cardiomyocyte cell line AC16. DHE staining and peroxide-sensitive dye 2',7'-dichlorofluorescein diacetate (H2DCFDA) was conducted to assess the reactive oxygen species (ROS) production. Cardiomyocyte hypertrophy was estimated using wheat germ agglutinin (WGA) staining and immunofluorescence. Aryl hydrocarbon receptor (AhR) translocation was observed by immunofluorescence. The activation of AhR was evaluated by immunoblotting of cytochrome P450 1 s (CYP1s) and quantitative real-time PCR (RT-PCR) analysis of AHRR and PTGS2. Additionally, the pro-oxidative and pro-hypertrophic effects were evaluated using the AhR inhibitor CH-223191, the CYP1s inhibitor Alizarin and the ROS scavenger N-Acetylcysteine (NAC). RESULTS UCM rat model was successfully established, and cardiac hypertrophy, accompanied by increased blood pressure, and myocardial fibrosis. Further research confirmed the activation of the AhR pathway in UCM rats including AhR translocation and downstream protein CYP1s expression, accompanied with increasing ROS production detected by DHE staining. In vitro experiment demonstrated a translocation of AhR triggered by IS, leading to significant increase of downstream gene expression. Subsequently study indicated a close relationship between the production of ROS and the activation of AhR/CYP1s, which was effectively blocked by applying AhR inhibitor, CYP1s inhibitor and siRNA against AhR. Moreover, the inhibition of AhR/CYP1s/ROS pathway collectively blocked the pro-hypertrophic effect of IS-mediated cardiomyopathy. CONCLUSION This study provides evidence that the AhR/CYP1s pathway is activated in UCM rats, and this activation is correlated with the uremic toxin IS. In vitro studies indicate that IS can stimulate the AhR translocation in cardiomyocyte, triggering to the production of intracellular ROS via CYP1s. This process leads to prolonged oxidative stress stimulation and thus contributes to the progression of uremic toxin-mediated cardiomyopathy.
Collapse
Affiliation(s)
- Wei Lu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney Disease, China; Kidney and Dialysis Institute of Shanghai, China; Kidney and Blood Purification Key Laboratory of Shanghai, China
| | - Shi Cheng
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney Disease, China; Kidney and Dialysis Institute of Shanghai, China; Kidney and Blood Purification Key Laboratory of Shanghai, China
| | - Jiarui Xu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney Disease, China; Kidney and Dialysis Institute of Shanghai, China; Kidney and Blood Purification Key Laboratory of Shanghai, China
| | - Zilong Xiao
- Division of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yong Yu
- Division of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qiwen Xie
- Department of Nephrology, Xiamen Branch, Zhongshan hospital, Fudan University; Nephrology, China; Clinical Quality Control Center of Xiamen, No.668 Jinhu Road, Xiamen, Fujian 361006, China
| | - Yi Fang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney Disease, China; Kidney and Dialysis Institute of Shanghai, China; Kidney and Blood Purification Key Laboratory of Shanghai, China
| | - Ruizhen Chen
- Division of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Bo Shen
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney Disease, China; Kidney and Dialysis Institute of Shanghai, China; Kidney and Blood Purification Key Laboratory of Shanghai, China.
| | - Yeqing Xie
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney Disease, China; Kidney and Dialysis Institute of Shanghai, China; Kidney and Blood Purification Key Laboratory of Shanghai, China.
| | - Xiaoqiang Ding
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney Disease, China; Kidney and Dialysis Institute of Shanghai, China; Kidney and Blood Purification Key Laboratory of Shanghai, China.
| |
Collapse
|
8
|
Xu X, Zhang CJ, Talifu Z, Liu WB, Li ZH, Wang XX, Du HY, Ke H, Yang DG, Gao F, Du LJ, Yu Y, Jing YL, Li JJ. The Effect of Glycine and N-Acetylcysteine on Oxidative Stress in the Spinal Cord and Skeletal Muscle After Spinal Cord Injury. Inflammation 2024; 47:557-571. [PMID: 37975960 DOI: 10.1007/s10753-023-01929-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/24/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
Oxidative stress is a frequently occurring pathophysiological feature of spinal cord injury (SCI) and can result in secondary injury to the spinal cord and skeletal muscle atrophy. Studies have reported that glycine and N-acetylcysteine (GlyNAC) have anti-aging and anti-oxidative stress properties; however, to date, no study has assessed the effect of GlyNAC in the treatment of SCI. In the present work, we established a rat model of SCI and then administered GlyNAC to the animals by gavage at a dose of 200 mg/kg for four consecutive weeks. The BBB scores of the rats were significantly elevated from the first to the eighth week after GlyNAC intervention, suggesting that GlyNAC promoted the recovery of motor function; it also promoted the significant recovery of body weight of the rats. Meanwhile, the 4-week heat pain results also suggested that GlyNAC intervention could promote the recovery of sensory function in rats to some extent. Additionally, after 4 weeks, the levels of glutathione and superoxide dismutase in spinal cord tissues were significantly elevated, whereas that of malondialdehyde was significantly decreased in GlyNAC-treated animals. The gastrocnemius wet weight ratio and total antioxidant capacity were also significantly increased. After 8 weeks, the malondialdehyde level had decreased significantly in spinal cord tissue, while reactive oxygen species accumulation in skeletal muscle had decreased. These findings suggested that GlyNAC can protect spinal cord tissue, delay skeletal muscle atrophy, and promote functional recovery in rats after SCI.
Collapse
Affiliation(s)
- Xin Xu
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, 100068, China
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Chun-Jia Zhang
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, 100068, China
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Zuliyaer Talifu
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Wu-Bo Liu
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, 100068, China
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
- Cheeloo College of Medicine, Shandong University, Jinan, 250100, Shandong Province, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, 250100, Shandong Province, China
| | - Ze-Hui Li
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, 100068, China
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Xiao-Xin Wang
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, 100068, China
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
- Cheeloo College of Medicine, Shandong University, Jinan, 250100, Shandong Province, China
| | - Hua-Yong Du
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, 100068, China
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Han Ke
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, 100068, China
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
- Cheeloo College of Medicine, Shandong University, Jinan, 250100, Shandong Province, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, 250100, Shandong Province, China
| | - De-Gang Yang
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, 100068, China
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Feng Gao
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, 100068, China
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Liang-Jie Du
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, 100068, China
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Yan Yu
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Ying-Li Jing
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Jian-Jun Li
- School of Rehabilitation, Capital Medical University, Beijing, 100068, China.
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, 100068, China.
- Chinese Institute of Rehabilitation Science, Beijing, 100068, China.
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, China.
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China.
- Cheeloo College of Medicine, Shandong University, Jinan, 250100, Shandong Province, China.
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266000, Shandong Province, China.
| |
Collapse
|
9
|
Bataille S, McKay N, Koppe L, Beau A, Benoit B, Bartoli M, Da Silva N, Poitevin S, Aniort J, Chermiti R, Burtey S, Dou L. Indoxyl sulfate inhibits muscle cell differentiation via Myf6/MRF4 and MYH2 downregulation. Nephrol Dial Transplant 2023; 39:103-113. [PMID: 37349959 DOI: 10.1093/ndt/gfad123] [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: 03/14/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is associated with a significant decrease in muscle strength and mass, possibly related to muscle cell damage by uremic toxins. Here, we studied in vitro and in vivo the effect of indoxyl sulfate (IS), an indolic uremic toxin, on myoblast proliferation, differentiation and expression of myogenic regulatory factors (MRF)-myoblast determination protein 1 (MyoD1), myogenin (Myog), Myogenic Factor 5 (Myf5) and myogenic regulatory factor 4 (Myf6/MRF4)-and expression of myosin heavy chain, Myh2. METHODS C2C12 myoblasts were cultured in vitro and differentiated in myotubes for 7 days in the presence of IS at a uremic concentration of 200 µM. Myocytes morphology and differentiation was analyzed after hematoxylin-eosin staining. MRF genes' expression was studied using reverse transcription polymerase chain reaction in myocytes and 5/6th nephrectomized mice muscle. Myf6/MRF4 protein expression was studied using enzyme-linked immunosorbent assay; MYH2 protein expression was studied using western blotting. The role of Aryl Hydrocarbon Receptor (AHR)-the cell receptor of IS-was studied by adding an AHR inhibitor into the cell culture milieu. RESULTS In the presence of IS, the myotubes obtained were narrower and had fewer nuclei than control myotubes. The presence of IS during differentiation did not modify the gene expression of the MRFs Myf5, MyoD1 and Myog, but induced a decrease in expression of Myf6/MRF4 and MYH2 at the mRNA and the protein level. AHR inhibition by CH223191 did not reverse the decrease in Myf6/MRF4 mRNA expression induced by IS, which rules out the implication of the ARH genomic pathway. In 5/6th nephrectomized mice, the Myf6/MRF4 gene was down-regulated in striated muscles. CONCLUSION In conclusion, IS inhibits Myf6/MRF4 and MYH2 expression during differentiation of muscle cells, which could lead to a defect in myotube structure. Through these new mechanisms, IS could participate in muscle atrophy observed in CKD.
Collapse
Affiliation(s)
- Stanislas Bataille
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
- Department of Nephrology, Phocean Nephrology Institute, Clinique Bouchard, ELSAN, Marseille, France
| | - Nathalie McKay
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Laetitia Koppe
- Department of Nephrology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
- University Lyon, CarMeN lab, INSERM U1060, INRAE, Université Claude Bernard Lyon 1, Pierre Bénite, France
| | - Alice Beau
- University Lyon, CarMeN lab, INSERM U1060, INRAE, Université Claude Bernard Lyon 1, Pierre Bénite, France
| | - Bérengère Benoit
- University Lyon, CarMeN lab, INSERM U1060, INRAE, Université Claude Bernard Lyon 1, Pierre Bénite, France
| | - Marc Bartoli
- Aix Marseille University, MMG, INSERM, Marseille, France
| | | | | | - Julien Aniort
- Nephrology, Dialysis and Transplantation Department, Gabriel Montpied University Hospital, University Hospital of Clermont-Ferrand, Clermont-Ferrand, France
| | - Rania Chermiti
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Stéphane Burtey
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
- Aix-Marseille University, Centre de Néphrologie et Transplantation Rénale, AP-HM Hôpital de la Conception, Marseille, France
| | - Laetitia Dou
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
| |
Collapse
|
10
|
Ohashi A, Nakatani M, Hori H, Nakai S, Tsuchida K, Hasegawa M, Tsuboi N. Effects of N-acetyl-L-tryptophan on desorption of the protein-bound uremic toxin indoxyl sulfate and effects on uremic sarcopenia. Ther Apher Dial 2023; 27:1023-1027. [PMID: 37596835 DOI: 10.1111/1744-9987.14047] [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: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 08/20/2023]
Abstract
INTRODUCTION Indoxyl sulfate (IS) is a protein-bound uremic toxin that causes uremic sarcopenia. IS has poor dialysis clearance; however, the addition of a binding competitor improves its removal efficiency. METHODS Dialysis experiments were performed using N-acetyl-l-tryptophan (L-NAT) instead of l-tryptophan (Trp) using pooled sera obtained from dialysis patients. The molecular structures of L-NAT and Trp were similar to that of IS. Therefore, we examined whether Trp and L-NAT were involved in muscle atrophy in the same manner as IS by performing culture experiments using a human myotube cell line. RESULTS The removal efficiency of L-NAT was the same as that of Trp. However, L-NAT concentrations in the pooled sera increased at the end of the experiment. Trp (1 mM) decreased the area of human myocytes, similar to IS, whereas L-NAT did not. CONCLUSION L-NAT is a binding competitor with the ability to remove protein-bound IS while preventing sarcopenia.
Collapse
Affiliation(s)
- Atsushi Ohashi
- Faculty of Clinical Science, School of Medical Sciences, Fujita Health University, Toyoake, Japan
| | - Masashi Nakatani
- Faculty of Rehabilitation and Care, Seijoh University, Tokai, Japan
| | - Hideo Hori
- Faculty of Clinical Science, School of Medical Sciences, Fujita Health University, Toyoake, Japan
| | - Shigeru Nakai
- Faculty of Clinical Science, School of Medical Sciences, Fujita Health University, Toyoake, Japan
| | - Kunihiro Tsuchida
- Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Midori Hasegawa
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Naotake Tsuboi
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Japan
| |
Collapse
|
11
|
Mo X, Shen L, Cheng R, Wang P, Wen L, Sun Y, Wang Q, Chen J, Lin S, Liao Y, Yang W, Yan H, Liu L. Faecal microbiota transplantation from young rats attenuates age-related sarcopenia revealed by multiomics analysis. J Cachexia Sarcopenia Muscle 2023; 14:2168-2183. [PMID: 37439281 PMCID: PMC10570072 DOI: 10.1002/jcsm.13294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 04/12/2023] [Accepted: 05/22/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Gut microbiota plays a key role in the development of sarcopenia via the 'gut-muscle' axis, and probiotics-based therapy might be a strategy for sarcopenia. Fecal microbiota transplantation from young donors (yFMT) has attracted much attention because of its probiotic function. However, whether or not yFMT is effective for sarcopenia in old recipients is largely unknown. Thus, we aimed to investigate the effect and mechanism of yFMT on age-related sarcopenia. METHODS The fecal microbiota of either young (12 weeks) or old (88 weeks) donor rats was transplanted into aged recipient rats for 8 weeks. Then, muscle mass, muscle strength, muscle function, muscle atrophy, and muscle regeneration capacity were measured. Analysis of fecal 16 s rRNA, serum non-targeted metabolomic, gut barrier integrity, and muscle transcriptome was conducted to elucidate the interaction between gut microbiota and skeletal muscles. RESULTS As evaluated by magnetic resonance imaging examination, grip strength test (P < 0.01), rotarod test (P < 0.05), and exhaustive running test (P < 0.05), we found that yFMT mitigated muscle mass loss, muscle strength weakness, and muscle function impairment in aged rats. yFMT also countered age-related atrophy and poor regeneration capacity in fast- and slow-switch muscles, which were manifested by the decrease in slow-switch myofibres (both P < 0.01) and muscle interstitial fibrosis (both P < 0.05) and the increase in the cross-section area of myofibres (both P < 0.001), fast-switch myofibres (both P < 0.01), and muscle satellite cells (both P < 0.001). In addition, yFMT ameliorated age-related dysbiosis of gut microbiota and metabolites by promoting the production of beneficial bacteria and metabolites-Akkermansia, Lactococcus, Lactobacillus, γ-glutamyltyrosine, 3R-hydroxy-butanoic acid, and methoxyacetic acid and inhibiting the production of deleterious bacteria and metabolites-Family_XIII_AD3011_group, Collinsella, indoxyl sulfate, indole-3-carboxilic acid-O-sulphate, and trimethylamine N-oxide. Also, yFMT prevented age-related destruction of gut barrier integrity by increasing the density of goblet cells (P < 0.0001) and the expression levels of mucin-2 (P < 0.0001) and tight junctional proteins (all P < 0.05). Meanwhile, yFMT attenuated age-related impairment of mitochondrial biogenesis and function in fast- and slow-switch muscles. Correlation analysis revealed that yFMT-induced alterations of gut microbiota and metabolites might be closely related to mitochondria-related genes and sarcopenia-related phenotypes. CONCLUSIONS yFMT could reshape the dysbiosis of gut microbiota and metabolites, maintain gut barrier integrity, and improve muscle mitochondrial dysfunction, eventually alleviating sarcopenia in aged rats. yFMT might be a new therapeutic strategy for age-related sarcopenia.
Collapse
Affiliation(s)
- Xiaoxing Mo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lihui Shen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ruijie Cheng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Pei Wang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lin Wen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yunhong Sun
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Qiang Wang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Juan Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Shan Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wei Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hong Yan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
12
|
Zhang H, Qi G, Wang K, Yang J, Shen Y, Yang X, Chen X, Yao X, Gu X, Qi L, Zhou C, Sun H. Oxidative stress: roles in skeletal muscle atrophy. Biochem Pharmacol 2023:115664. [PMID: 37331636 DOI: 10.1016/j.bcp.2023.115664] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
Oxidative stress, inflammation, mitochondrial dysfunction, reduced protein synthesis, and increased proteolysis are all critical factors in the process of muscle atrophy. In particular, oxidative stress is the key factor that triggers skeletal muscle atrophy. It is activated in the early stages of muscle atrophy and can be regulated by various factors. The mechanisms of oxidative stress in the development of muscle atrophy have not been completely elucidated. This review provides an overview of the sources of oxidative stress in skeletal muscle and the correlation of oxidative stress with inflammation, mitochondrial dysfunction, autophagy, protein synthesis, proteolysis, and muscle regeneration in muscle atrophy. Additionally, the role of oxidative stress in skeletal muscle atrophy caused by several pathological conditions, including denervation, unloading, chronic inflammatory diseases (diabetes mellitus, chronic kidney disease, chronic heart failure, and chronic obstructive pulmonary disease), sarcopenia, hereditary neuromuscular diseases (spinal muscular atrophy, amyotrophic lateral sclerosis, and Duchenne muscular dystrophy), and cancer cachexia, have been discussed. Finally, this review proposes the alleviation oxidative stress using antioxidants, Chinese herbal extracts, stem cell and extracellular vesicles as a promising therapeutic strategy for muscle atrophy. This review will aid in the development of novel therapeutic strategies and drugs for muscle atrophy.
Collapse
Affiliation(s)
- Han Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Guangdong Qi
- Department of Endocrinology, Binhai County People's Hospital, Yancheng, Jiangsu Province, 224500, PR China
| | - Kexin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Jiawen Yang
- Department of Clinical Medicine, Medical College, Nantong University, Nantong 226001, China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Xiaoming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Xin Chen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Xinlei Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, PR China.
| | - Chun Zhou
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, PR China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, Nantong, Jiangsu Province, 226001, PR China; Research and Development Center for E-Learning, Ministry of Education, Beijing 100816, PR China.
| |
Collapse
|
13
|
Pan Y, Zhou T, Dong X, Wu L, Wang P, Wang S, Zhang A. Urotensin II can Induce Skeletal Muscle Atrophy Associated with Upregulating Ubiquitin-Proteasome System and Inhibiting the Differentiation of Satellite Cells in CRF Mice. Calcif Tissue Int 2023; 112:603-612. [PMID: 36892588 DOI: 10.1007/s00223-023-01073-4] [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: 11/01/2022] [Accepted: 02/21/2023] [Indexed: 03/10/2023]
Abstract
Skeletal muscle wasting and atrophy is highly prevalent in chronic renal failure (CRF) and increases the risk of mortality. According to our previous study, we speculate that urotensin II (UII) can induce skeletal muscle atrophy by upregulating ubiquitin-proteasome system(UPS) in CRF. C2C12 mouse myoblast cells were differentiated into myotubes, and myotubes were exposed to different concentrations of UII. Myotube diameters, myosin heavy chain(MHC), p-Fxo03A, skeletal muscle-specific E3 ubiquitin ligases such as muscle RING finger 1 (MuRF1) and muscle atrophy F-box (MAFbx/atrogin1) were detected. Three animal models (the sham operation mice as normal control (NC) group, wild-type C57BL/6 mice with 5/6 nephrectomy (WT CRF) group, UII receptor gene knock out (UT KO) mice with 5/6 nephrectomy (UT KO CRF) group) were designed. Cross-sectional area (CSA) of skeletal muscle tissues in three animal models were measured, and western blot detected protein of UII, p-Fxo03A, MAFbx and MuRF1, and immunofluorescence assays explored the satellite cell marker of Myod1 and Pax7, and PCR arrays detected the muscle protein degradation genes, protein synthesis genes and the genes which were involved in muscle components. UII could decrease mouse myotube diameters, and upregulate dephosphorylated Fxo03A protein. MAFbx and MuRF1 were higher in WT CRF group than that in NC group, but after UII receptor gene was knocked out (UT KO CRF), their expressions were downregulated. UII could inhibit the expression of Myod1 but not Pax7 in animal study. We first demonstrate that skeletal muscle atrophy induced by UII associated with upregulating ubiquitin-proteasome system and inhibiting the differentiation of satellite cells in CRF mice.
Collapse
Affiliation(s)
- Yajing Pan
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China
| | - Ting Zhou
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China
| | - Xingtong Dong
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China
| | - Leiyun Wu
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China
| | - Peiwen Wang
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China
| | - Shiyuan Wang
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China
| | - Aihua Zhang
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China.
| |
Collapse
|
14
|
Wang K, Liu Q, Tang M, Qi G, Qiu C, Huang Y, Yu W, Wang W, Sun H, Ni X, Shen Y, Fang X. Chronic kidney disease-induced muscle atrophy: Molecular mechanisms and promising therapies. Biochem Pharmacol 2023; 208:115407. [PMID: 36596414 DOI: 10.1016/j.bcp.2022.115407] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/02/2023]
Abstract
Chronic kidney disease (CKD) is a high-risk chronic catabolic disease due to its high morbidity and mortality. CKD is accompanied by many complications, leading to a poor quality of life, and serious complications may even threaten the life of CKD patients. Muscle atrophy is a common complication of CKD. Muscle atrophy and sarcopenia in CKD patients have complex pathways that are related to multiple mechanisms and related factors. This review not only discusses the mechanisms by which inflammation, oxidative stress, mitochondrial dysfunction promote CKD-induced muscle atrophy but also explores other CKD-related complications, such as metabolic acidosis, vitamin D deficiency, anorexia, and excess angiotensin II, as well as other related factors that play a role in CKD muscle atrophy, such as insulin resistance, hormones, hemodialysis, uremic toxins, intestinal flora imbalance, and miRNA. We highlight potential treatments and drugs that can effectively treat CKD-induced muscle atrophy in terms of complication treatment, nutritional supplementation, physical exercise, and drug intervention, thereby helping to improve the prognosis and quality of life of CKD patients.
Collapse
Affiliation(s)
- Kexin Wang
- Department of Nephrology, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, PR China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Qingyuan Liu
- Department of Endocrinology, Binhai County People's Hospital, Yancheng, Jiangsu Province 224500, PR China
| | - Mingyu Tang
- Xinglin College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Guangdong Qi
- Department of Endocrinology, Binhai County People's Hospital, Yancheng, Jiangsu Province 224500, PR China
| | - Chong Qiu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Yan Huang
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Weiran Yu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Wei Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, PR China; Department of Pathology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, PR China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Xuejun Ni
- Department of Ultrasound Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, PR China.
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu Province 226001, PR China.
| | - Xingxing Fang
- Department of Nephrology, the Second Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, PR China.
| |
Collapse
|
15
|
Zhang T, Cheng JK, Hu YM. Gut microbiota as a promising therapeutic target for age-related sarcopenia. Ageing Res Rev 2022; 81:101739. [PMID: 36182084 DOI: 10.1016/j.arr.2022.101739] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/30/2022] [Accepted: 09/25/2022] [Indexed: 01/31/2023]
Abstract
Sarcopenia is characterized by a progressive loss of skeletal muscle mass and function with aging. Recently, sarcopenia has been shown to be closely related with gut microbiota. Strategies such as probiotics and fecal microbiota transplantation have shown potential to ameliorate the muscle loss. This review will focus on the age-related sarcopenia, in particular on the relationship between gut microbiota and age-related sarcopenia, how gut microbiota is engaged in sarcopenia, and the potential role of gut microbiota in the treatment of age-related sarcopenia.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Jin-Ke Cheng
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital Affiliated, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yao-Min Hu
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China.
| |
Collapse
|
16
|
Research progress on the relationship between IS and kidney disease and its complications. Int Urol Nephrol 2022; 54:2881-2890. [PMID: 35488145 DOI: 10.1007/s11255-022-03209-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 04/09/2022] [Indexed: 10/18/2022]
Abstract
Indoxyl sulphate (IS) a representative uraemic toxin in the blood of patients with chronic kidney disease (CKD). Its accumulation may be closely related to CKD and the increasing morbidity and mortality of the disease's related complications. Timely and effective detection of the IS level and efficient clearance of IS may effectively prevent the progression of CKD and its related complications. Therefore, this article summarizes the research progress of IS related, including IS in CKD and its associated complications including chronic kidney disease, chronic kidney disease with cardiovascular disease, renal anemia, bone mineral metabolic disease and neuropsychiatric disorders, looking for IS accurate rapid detection methods, and explore the efficient treatment to reduce blood levels of indole phenol sulphate.
Collapse
|
17
|
Serum concentrations of free indoxyl and p-cresyl sulfate are associated with mineral metabolism variables and cardiovascular risk in hemodialysis patients. J Nephrol 2022; 35:1457-1465. [PMID: 35175580 DOI: 10.1007/s40620-022-01271-7] [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: 11/08/2021] [Accepted: 02/01/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Indoxyl sulfate (IS) and p-cresyl sulfate (PCS) are uremic toxins associated with cardiovascular outcome in CKD patients. The present work is an analysis of the association of serum free, total IS and PCS with cardiovascular events and calcium-phosphate metabolism variables in hemodialysis patients. METHODS Serum levels of total and free IS and PCS were measured in 139 hemodialysis patients. Their relationship with calcium-phosphate metabolism variables were tested in an observational cohort study. In addition, their association with cardiovascular events was investigated during a 4-year follow-up. RESULTS Patients in the highest tertile (T3) of serum free IS showed lower serum 1,25(OH)2D compared to patients in the middle (T2) and lowest tertile (T1); in addition to this, T3 patients showed lower serum irisin than T1 patients and lower serum PTH than all the other subjects (T1 + T2) combined. Serum PTH was also measured during the two years after the baseline measurement and was higher in patients in the T1 than in those in the T3 of serum free IS. Cox regression analysis showed that cardiovascular risk was lower in T1 patients than in those in the T3 of serum free PCS, both using a univariate (OR 2.55, 95% CI 1.2-5.43; p = 0.015) or multivariate model (OR 2.48, 95% CI 1.12-5.51; p = 0.003). CONCLUSIONS Serum free IS may be associated with PTH and 1,25(OH)2D secretion, whereas free PCS may predict cardiovascular risk in hemodialysis patients.
Collapse
|
18
|
Yang J, Li H, Zhang C, Zhou Y. Indoxyl sulfate reduces Ito,f by activating ROS/MAPK and NF-κB signaling pathways. JCI Insight 2022; 7:145475. [PMID: 35132967 PMCID: PMC8855797 DOI: 10.1172/jci.insight.145475] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/08/2021] [Indexed: 12/17/2022] Open
Abstract
There is a high prevalence of ventricular arrhythmias related to sudden cardiac death in patients with chronic kidney disease (CKD). To explored the possible mechanism of CKD-related ventricular arrhythmias, a CKD rat model was created, and indoxyl sulfate (IS) was further used in vivo and in vitro. This project used the following methods: patch clamp, electrocardiogram, and some molecular biology experimental techniques. IS was found to be significantly elevated in the serum of CKD rats. Interestingly, the expression levels of the fast transient outward potassium current-related (Ito,f-related) proteins (Kv4.2, Kv4.3, and KChIP2) in the heart of CKD rats and rats treated with IS decreased. IS dose-dependently reduced Ito,f density, accompanied by the decreases in Kv4.2, Kv4.3, and KChIP2 proteins in vitro. IS also prolonged the action potential duration and QT interval, and paroxysmal ventricular tachycardia could be induced by IS. In-depth studies have shown that ROS/p38MAPK, ROS-p44/42 MAPK, and NF-κB signaling pathways play key roles in the reduction of Ito,f density and Ito,f-related proteins caused by IS. These data suggest that IS reduces Ito,f-related proteins and Ito,f density by activating ROS/MAPK and NF-κB signaling pathways, and the action potential duration and QT interval are subsequently prolonged, which contributes to increasing the susceptibility to arrhythmia in CKD.
Collapse
Affiliation(s)
- Jing Yang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hongxia Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chi Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yafeng Zhou
- Department of Cardiology, Dushu Public Hospital Affiliated to Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
19
|
The Atrophic Effect of 1,25(OH) 2 Vitamin D 3 (Calcitriol) on C2C12 Myotubes Depends on Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10121980. [PMID: 34943083 PMCID: PMC8750283 DOI: 10.3390/antiox10121980] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 11/17/2022] Open
Abstract
Dysfunctional mitochondrial metabolism has been linked to skeletal muscle loss in several physio-pathological states. Although it has been reported that vitamin D (VD) supports cellular redox homeostasis by maintaining normal mitochondrial functions, and VD deficiency often occurs in conditions associated with skeletal muscle loss, the efficacy of VD supplementation to overcome muscle wasting is debated. Investigations on the direct effects of VD metabolites on skeletal muscle using C2C12 myotubes have revealed an unexpected pro-atrophic activity of calcitriol (1,25VD), while its upstream metabolites cholecalciferol (VD3) and calcidiol (25VD) have anti-atrophic effects. Here, we investigated if the atrophic effects of 1,25VD on myotubes depend on its activity on mitochondrial metabolism. The impact of 1,25VD and its upstream metabolites VD3 and 25VD on mitochondria dynamics and the activity of C2C12 myotubes was evaluated by measuring mitochondrial content, architecture, metabolism, and reactive oxygen species (ROS) production. We found that 1,25VD induces atrophy through protein kinase C (PKC)-mediated ROS production, mainly of extramitochondrial origin. Consistent with this, cotreatment with the antioxidant N-acetylcysteine (NAC), but not with the mitochondria-specific antioxidant mitoTEMPO, was sufficient to blunt the atrophic activity of 1,25VD. In contrast, VD3 and 25VD have antioxidant properties, suggesting that the efficacy of VD supplementation might result from the balance between atrophic pro-oxidant (1,25VD) and protective antioxidant (VD3 and 25VD) metabolites.
Collapse
|
20
|
Liu C, Cheung W, Li J, Chow SK, Yu J, Wong SH, Ip M, Sung JJY, Wong RMY. Understanding the gut microbiota and sarcopenia: a systematic review. J Cachexia Sarcopenia Muscle 2021; 12:1393-1407. [PMID: 34523250 PMCID: PMC8718038 DOI: 10.1002/jcsm.12784] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 04/03/2021] [Accepted: 08/02/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Gut microbiota dysbiosis and sarcopenia commonly occur in the elderly. Although the concept of the gut-muscle axis has been raised, the casual relationship is still unclear. This systematic review analyses the current evidence of gut microbiota effects on muscle/sarcopenia. METHODS A systematic review was performed in PubMed, Embase, Web of Science, and The Cochrane Library databases using the keywords (microbiota* OR microbiome*) AND (sarcopen* OR muscle). Studies reporting the alterations of gut microbiota and muscle/physical performance were analysed. RESULTS A total of 26 pre-clinical and 10 clinical studies were included. For animal studies, three revealed age-related changes and relationships between gut microbiota and muscle. Three studies focused on muscle characteristics of germ-free mice. Seventy-five per cent of eight faecal microbiota transplantation studies showed that the recipient mice successfully replicated the muscle phenotype of donors. There were positive effects on muscle from seven probiotics, two prebiotics, and short-chain fatty acids (SCFAs). Ten studies investigated on other dietary supplements, antibiotics, exercise, and food withdrawal that affected both muscle and gut microbiota. Twelve studies explored the potential mechanisms of the gut-muscle axis. For clinical studies, 6 studies recruited 676 elderly people (72.8 ± 5.6 years, 57.8% female), while 4 studies focused on 244 young adults (29.7 ± 7.8 years, 55.4% female). The associations of gut microbiota and muscle had been shown in four observational studies. Probiotics, prebiotics, synbiotics, fermented milk, caloric restriction, and exercise in six studies displayed inconsistent effects on muscle mass, function, and gut microbiota. CONCLUSIONS Altering the gut microbiota through bacteria depletion, faecal transplantation, and various supplements was shown to directly affect muscle phenotypes. Probiotics, prebiotics, SCFAs, and bacterial products are potential novel therapies to enhance muscle mass and physical performance. Lactobacillus and Bifidobacterium strains restored age-related muscle loss. Potential mechanisms of microbiome modulating muscle mainly include protein, energy, lipid, and glucose metabolism, inflammation level, neuromuscular junction, and mitochondrial function. The role of the gut microbiota in the development of muscle loss during aging is a crucial area that requires further studies for translation to patients.
Collapse
Affiliation(s)
- Chaoran Liu
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| | - Wing‐Hoi Cheung
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| | - Jie Li
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| | - Simon Kwoon‐Ho Chow
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| | - Jun Yu
- Department of Medicine and TherapeuticsThe Chinese University of Hong KongHong Kong SARChina
| | - Sunny Hei Wong
- Department of Medicine and TherapeuticsThe Chinese University of Hong KongHong Kong SARChina
| | - Margaret Ip
- Department of MicrobiologyThe Chinese University of Hong KongHong Kong SARChina
| | - Joseph Jao Yiu Sung
- Department of Medicine and TherapeuticsThe Chinese University of Hong KongHong Kong SARChina
| | - Ronald Man Yeung Wong
- Department of Orthopaedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
| |
Collapse
|
21
|
Margiotta E, Caldiroli L, Callegari ML, Miragoli F, Zanoni F, Armelloni S, Rizzo V, Messa P, Vettoretti S. Association of Sarcopenia and Gut Microbiota Composition in Older Patients with Advanced Chronic Kidney Disease, Investigation of the Interactions with Uremic Toxins, Inflammation and Oxidative Stress. Toxins (Basel) 2021; 13:toxins13070472. [PMID: 34357944 PMCID: PMC8309956 DOI: 10.3390/toxins13070472] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/16/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Sarcopenia is a prevalent condition in chronic kidney disease (CKD). We determined gut microbiota (gMB) composition in CKD patients with or without sarcopenia. Furthermore, we investigated whether in these patients, there was any association between gMB, uremic toxins, inflammation and oxidative stress. We analyzed gMB composition, uremic toxins (indoxyl sulphate and p-cresyl sulphate), inflammatory cytokines (interleukin 10, tumor necrosis factor α, interleukin 6, interleukin 17, interleukin 12 p70, monocyte chemoattractant protein-1 and fetuin-A) and oxidative stress (malondialdehyde) of 64 elderly CKD patients (10 < eGFR < 45 mL/min/1.73 m2, not on dialysis) categorized as sarcopenic and not-sarcopenic. Sarcopenia was defined according to European Working Group on Sarcopenia in Older People 2 criteria. Sarcopenic patients had a greater abundance of the Micrococcaceae and Verrucomicrobiaceae families and of Megasphaera, Rothia, Veillonella, Akkermansia and Coprobacillus genera. They had a lower abundance of the Gemellaceae and Veillonellaceae families and of Acidaminococcus and Gemella genera. GMB was associated with uremic toxins, inflammatory cytokines and MDA. However, uremic toxins, inflammatory cytokines and MDA were not different in sarcopenic compared with not-sarcopenic individuals, except for interleukin 10, which was higher in not-sarcopenic patients. In older CKD patients, gMB was different in sarcopenic than in not-sarcopenic ones. Several bacterial families and genera were associated with uremic toxins and inflammatory cytokines, although none of these latter substantially different in sarcopenic versus not-sarcopenic patients.
Collapse
Affiliation(s)
- Elisabetta Margiotta
- Division of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (E.M.); (L.C.); (F.Z.); (S.A.); (P.M.)
| | - Lara Caldiroli
- Division of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (E.M.); (L.C.); (F.Z.); (S.A.); (P.M.)
| | - Maria Luisa Callegari
- Centro di Ricerche Biotecnologiche, Università Cattolica del Sacro Cuore, 26100 Cremona, Italy; (M.L.C.); (F.M.)
| | - Francesco Miragoli
- Centro di Ricerche Biotecnologiche, Università Cattolica del Sacro Cuore, 26100 Cremona, Italy; (M.L.C.); (F.M.)
| | - Francesca Zanoni
- Division of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (E.M.); (L.C.); (F.Z.); (S.A.); (P.M.)
| | - Silvia Armelloni
- Division of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (E.M.); (L.C.); (F.Z.); (S.A.); (P.M.)
| | - Vittoria Rizzo
- Laboratory Medicine and Clinical Biochemical Analysis, Ospedale San Matteo di Pavia, 27100 Pavia, Italy;
| | - Piergiorgio Messa
- Division of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (E.M.); (L.C.); (F.Z.); (S.A.); (P.M.)
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Simone Vettoretti
- Division of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (E.M.); (L.C.); (F.Z.); (S.A.); (P.M.)
- Correspondence: ; Tel.: +39-0255-034552
| |
Collapse
|
22
|
Chao CT, Lin SH. Uremic Toxins and Frailty in Patients with Chronic Kidney Disease: A Molecular Insight. Int J Mol Sci 2021; 22:ijms22126270. [PMID: 34200937 PMCID: PMC8230495 DOI: 10.3390/ijms22126270] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022] Open
Abstract
The accumulation of uremic toxins (UTs) is a prototypical manifestation of uremic milieu that follows renal function decline (chronic kidney disease, CKD). Frailty as a potential outcome-relevant indicator is also prevalent in CKD. The intertwined relationship between uremic toxins, including small/large solutes (phosphate, asymmetric dimethylarginine) and protein-bound ones like indoxyl sulfate (IS) and p-cresyl sulfate (pCS), and frailty pathogenesis has been documented recently. Uremic toxins were shown in vitro and in vivo to induce noxious effects on many organ systems and likely influenced frailty development through their effects on multiple preceding events and companions of frailty, such as sarcopenia/muscle wasting, cognitive impairment/cognitive frailty, osteoporosis/osteodystrophy, vascular calcification, and cardiopulmonary deconditioning. These organ-specific effects may be mediated through different molecular mechanisms or signal pathways such as peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α), mitogen-activated protein kinase (MAPK) signaling, aryl hydrocarbon receptor (AhR)/nuclear factor-κB (NF-κB), nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), Runt-related transcription factor 2 (RUNX2), bone morphogenic protein 2 (BMP2), osterix, Notch signaling, autophagy effectors, microRNAs, and reactive oxygen species induction. Anecdotal clinical studies also suggest that frailty may further accelerate renal function decline, thereby augmenting the accumulation of UTs in affected individuals. Judging from these threads of evidence, management strategies aiming for uremic toxin reduction may be a promising approach for frailty amelioration in patients with CKD. Uremic toxin lowering strategies may bear the potential of improving patients’ outcomes and restoring their quality of life, through frailty attenuation. Pathogenic molecule-targeted therapeutics potentially disconnect the association between uremic toxins and frailty, additionally serving as an outcome-modifying approach in the future.
Collapse
Affiliation(s)
- Chia-Ter Chao
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei 10845, Taiwan;
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100255, Taiwan
- Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Shih-Hua Lin
- Nephrology Division, Department of Internal Medicine, National Defense Medical Center, Taipei 11490, Taiwan
- Correspondence: or
| |
Collapse
|
23
|
β2-adrenergic receptor agonist counteracts skeletal muscle atrophy and oxidative stress in uremic mice. Sci Rep 2021; 11:9130. [PMID: 33911115 PMCID: PMC8080640 DOI: 10.1038/s41598-021-88438-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
In patients with chronic kidney disease, skeletal muscle dysfunction is associated with mortality. Uremic sarcopenia is caused by ageing, malnutrition, and chronic inflammation, but the molecular mechanism and potential therapeutics have not been fully elucidated yet. We hypothesize that accumulated uremic toxins might exert a direct deteriorative effect on skeletal muscle and explore the pharmacological treatment in experimental animal and culture cell models. The mice intraperitoneally injected with indoxyl sulfate (IS) after unilateral nephrectomy displayed an elevation of IS concentration in skeletal muscle and a reduction of instantaneous muscle strength, along with the predominant loss of fast-twitch myofibers and intramuscular reactive oxygen species (ROS) generation. The addition of IS in the culture media decreased the size of fully differentiated mouse C2C12 myotubes as well. ROS accumulation and mitochondrial dysfunction were also noted. Next, the effect of the β2-adrenergic receptor (β2-AR) agonist, clenbuterol, was evaluated as a potential treatment for uremic sarcopenia. In mice injected with IS, clenbuterol treatment increased the muscle mass and restored the tissue ROS level but failed to improve muscle weakness. In C2C12 myotubes stimulated with IS, although β2-AR activation also attenuated myotube size reduction and ROS accumulation as did other anti-oxidant reagents, it failed to augment the mitochondrial membrane potential. In conclusion, IS provokes muscular strength loss (uremic dynapenia), ROS generation, and mitochondrial impairment. Although the β2-AR agonist can increase the muscular mass with ROS reduction, development of therapeutic interventions for restoring skeletal muscle function is still awaited.
Collapse
|
24
|
Kumar S, Singh SK, Rana B, Rana A. The regulatory function of mixed lineage kinase 3 in tumor and host immunity. Pharmacol Ther 2021; 219:107704. [PMID: 33045253 PMCID: PMC7887016 DOI: 10.1016/j.pharmthera.2020.107704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/02/2020] [Indexed: 12/26/2022]
Abstract
Protein kinases are the second most sought-after G-protein coupled receptors as drug targets because of their overexpression, mutations, and dysregulated catalytic activities in various pathological conditions. Till 2019, 48 protein kinase inhibitors have received FDA approval for the treatment of multiple illnesses, of which the majority of them are indicated for different malignancies. One of the attractive sub-group of protein kinases that has attracted attention for drug development is the family members of MAPKs that are recognized to play significant roles in different cancers. Several inhibitors have been developed against various MAPK members; however, none of them as monotherapy has shown sustainable efficacy. One of the MAPK members, called Mixed Lineage Kinase 3 (MLK3), has attracted considerable attention due to its role in inflammation and neurodegenerative diseases; however, its role in cancer is an emerging area that needs more investigation. Recent advances have shown that MLK3 plays a role in cancer cell survival, migration, drug resistance, cell death, and tumor immunity. This review describes how MLK3 regulates different MAPK pathways, cancer cell growth and survival, apoptosis, and host's immunity. We also discuss how MLK3 inhibitors can potentially be used along with immunotherapy for different malignancies.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA.
| | - Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
25
|
Alcalde-Estévez E, Sosa P, Asenjo-Bueno A, Plaza P, Olmos G, Naves-Díaz M, Rodríguez-Puyol D, López-Ongil S, Ruiz-Torres MP. Uraemic toxins impair skeletal muscle regeneration by inhibiting myoblast proliferation, reducing myogenic differentiation, and promoting muscular fibrosis. Sci Rep 2021; 11:512. [PMID: 33436654 PMCID: PMC7804102 DOI: 10.1038/s41598-020-79186-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 12/04/2020] [Indexed: 01/29/2023] Open
Abstract
Uraemic toxins increase in serum parallel to a decline in the glomerular filtration rate and the development of sarcopenia in patients with chronic kidney disease (CKD). This study analyses the role of uraemic toxins in sarcopenia at different stages of CKD, evaluating changes in the muscular regeneration process. Cultured C2C12 cells were incubated with a combination of indoxyl sulphate and p-cresol at high doses (100 µg/mL) or low doses (25 µg/mL and 10 µg/mL) resembling late or early CKD stages, respectively. Cell proliferation (analysed by scratch assays and flow cytometry) was inhibited only by high doses of uraemic toxins, which inactivated the cdc2-cyclin B complex, inhibiting mitosis and inducing apoptosis (analysed by annexin V staining). By contrast, low doses of uraemic toxins did not affect proliferation, but reduced myogenic differentiation, primed with 2% horse serum, by inhibiting myogenin expression and promoting fibro-adipogenic differentiation. Finally, to assess the in vivo relevance of these results, studies were performed in gastrocnemii from uraemic rats, which showed higher collagen expression and lower myosin heavy chain expression than those from healthy rats. In conclusion, uraemic toxins impair the skeletal muscular regeneration process, even at low concentrations, suggesting that sarcopenia can progress from the early stages of CKD.
Collapse
Affiliation(s)
- Elena Alcalde-Estévez
- grid.7159.a0000 0004 1937 0239Departamento de Biología de Sistemas, Facultad de Medicina Y Ciencias de La Salud, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain
| | - Patricia Sosa
- grid.7159.a0000 0004 1937 0239Departamento de Biología de Sistemas, Facultad de Medicina Y Ciencias de La Salud, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain
| | - Ana Asenjo-Bueno
- grid.411336.20000 0004 1765 5855Unidad de Investigación de La Fundación Para La Investigación Biomédica del Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Patricia Plaza
- grid.411336.20000 0004 1765 5855Unidad de Investigación de La Fundación Para La Investigación Biomédica del Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Gemma Olmos
- grid.7159.a0000 0004 1937 0239Departamento de Biología de Sistemas, Facultad de Medicina Y Ciencias de La Salud, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain ,Instituto Reina Sofía de Investigación Nefrológica, IRSIN, Madrid, Spain ,grid.420232.50000 0004 7643 3507Area 3-Fisiología y Fisiopatología Renal Y Vascular del IRYCIS, Madrid, Spain
| | - Manuel Naves-Díaz
- Unidad de Gestión Clínica de Metabolismo Óseo. Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Diego Rodríguez-Puyol
- Instituto Reina Sofía de Investigación Nefrológica, IRSIN, Madrid, Spain ,grid.420232.50000 0004 7643 3507Area 3-Fisiología y Fisiopatología Renal Y Vascular del IRYCIS, Madrid, Spain ,grid.411336.20000 0004 1765 5855Departamento de Medicina Y Especialidades Médicas, Universidad de Alcalá Y Servicio de Nefrología del Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Susana López-Ongil
- grid.411336.20000 0004 1765 5855Unidad de Investigación de La Fundación Para La Investigación Biomédica del Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain ,Instituto Reina Sofía de Investigación Nefrológica, IRSIN, Madrid, Spain ,grid.420232.50000 0004 7643 3507Area 3-Fisiología y Fisiopatología Renal Y Vascular del IRYCIS, Madrid, Spain
| | - María P. Ruiz-Torres
- grid.7159.a0000 0004 1937 0239Departamento de Biología de Sistemas, Facultad de Medicina Y Ciencias de La Salud, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain ,Instituto Reina Sofía de Investigación Nefrológica, IRSIN, Madrid, Spain ,grid.420232.50000 0004 7643 3507Area 3-Fisiología y Fisiopatología Renal Y Vascular del IRYCIS, Madrid, Spain
| |
Collapse
|
26
|
He X, Wang Z, Wei L, Cheng X, Chen L, Gao F, Jiang H. Indoxyl sulfate promotes osteogenic differentiation of vascular smooth muscle cells by miR-155-5p-dependent downregulation of matrix Gla protein via ROS/NF-κB signaling. Exp Cell Res 2020; 397:112301. [PMID: 32979364 DOI: 10.1016/j.yexcr.2020.112301] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 11/25/2022]
Abstract
Vascular calcification (VC) is a major risk factor for increasing cardiovascular morbidity and mortality in patients with chronic kidney disease (CKD). Indoxyl sulfate (IS), a representative uremic toxin, is closely associated with VC in CKD patients. Matrix Gla protein (MGP) plays pivotal role in VC as a calcification inhibitor. The aim of this work was to explore whether MGP was involved in IS-induced VC. Here, we demonstrated the role of MGP in the IS-induced osteogenic differentiation of human aortic smooth muscle cells (HASMCs). The methods included Von Kossa staining, immunohistochemistry, Alizarin Red staining, quantitative real-time PCR and western blotting. MGP was decreased in calcified arteries both in CKD patients and rats. In vitro, IS suppressed MGP expression in HASMCs by activating ROS/NF-κB signaling in parallel with osteogenic differentiation, which was mitigated by inhibiting ROS and NF-κB with diphenyleneiodonium and Bay11-7082. Further investigation showed that IS induced NF-κB-responsive microRNA (miR)-155-5p mediating MGP downregulation. Overexpression of miR-155-5p with mimics aggravated IS-induced MGP reduction and osteogenic differentiation. In contrast, these conditions were diminished by silencing miR-155-5p. We demonstrate that IS promotes the HASMCs phenotype switch by suppressing MGP expression via ROS/NF-κB/miR-155-5p signaling and provide a new insight for the pathogenesis of IS-induced VC.
Collapse
Affiliation(s)
- Xin He
- Dialysis Department of Nephrology Hospital, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Zhigang Wang
- Department of Nephrology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Limin Wei
- Dialysis Department of Nephrology Hospital, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xin Cheng
- Dialysis Department of Nephrology Hospital, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Lei Chen
- Dialysis Department of Nephrology Hospital, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Fanfan Gao
- Dialysis Department of Nephrology Hospital, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Hongli Jiang
- Dialysis Department of Nephrology Hospital, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
27
|
Uremic Sarcopenia: Clinical Evidence and Basic Experimental Approach. Nutrients 2020; 12:nu12061814. [PMID: 32570738 PMCID: PMC7353433 DOI: 10.3390/nu12061814] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/27/2020] [Accepted: 06/16/2020] [Indexed: 12/17/2022] Open
Abstract
Sustained physical activity extends healthy life years while a lower activity due to sarcopenia can reduce them. Sarcopenia is defined as a decrease in skeletal muscle mass and strength due not only to aging, but also from a variety of debilitating chronic illnesses such as cancer and heart failure. Patients with chronic kidney disease (CKD), who tend to be cachexic and in frail health, may develop uremic sarcopenia or uremic myopathy due to an imbalance between muscle protein synthesis and catabolism. Here, we review clinical evidence indicating reduced physical activity as renal function deteriorates and explore evidence-supported therapeutic options focusing on nutrition and physical training. In addition, although sarcopenia is a clinical concept and difficult to recapitulate in basic research, several in vivo approaches have been attempted, such as rodent subtotal nephrectomy representing both renal dysfunction and muscle weakness. This review highlights molecular mechanisms and promising interventions for uremic sarcopenia that were revealed through basic research. Extensive study is still needed to cast light on the many aspects of locomotive organ impairments in CKD and explore the ways that diet and exercise therapies can improve both outcomes and quality of life at every level.
Collapse
|
28
|
Changchien CY, Sung MH, Chang HH, Tsai WC, Peng YS, Chen Y. Uremic toxin indoxyl sulfate suppresses myocardial Cx43 assembly and expression via JNK activation. Chem Biol Interact 2020; 319:108979. [PMID: 32045570 DOI: 10.1016/j.cbi.2020.108979] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/30/2020] [Accepted: 02/06/2020] [Indexed: 12/14/2022]
Abstract
Heart rhythm disturbances have been widely recognized as major triggers of cardiovascular (CV) mortality in chronic kidney disease (CKD) patients. Connexin 43 (Cx43)-composed gap junctions are essential in cardiomyocyte synchronization and may be involved in the pathological response to uremic toxins. Indoxyl sulfate (IS) is one of the most dominant uremic toxins that contribute to CKD-related cardiovascular diseases. In primary cultures of rat neonatal cardiomyocytes, we demonstrated that IS treatment decreased spontaneous contraction without impairing viability. In addition, there was disruption of gap junction intercellular communication (GJIC) between cardiomyocytes after 30 min of IS stimulation. IS caused time- and dose-dependent Cx43 redistribution, and the patterns of Cx43 immunostaining returned to baseline while IS stimulation was removed. Furthermore, IS exposure downregulated Cx43 protein and mRNA levels. Elevated JNK1 and JNK2 phosphorylation was further identified after IS exposure in both rat cardiomyocytes and H9c2 cells. The above changes as well as GJIC and Cx43 suppression were reversed by pretreatment with a JNK inhibitor (SP600125). Inhibition of p-JNK attenuated IS-mediated downward trends in Cx43 transcription and translation. In cardiac muscle from nephrectomy-induced CKD mice, an alteration in Cx43 level was identified at intercalated discs. Our findings disclosed that JNK activation might participate in the remodeling of gap junction and Cx43 expression by uremic toxin-IS both in vitro and in vivo.
Collapse
Affiliation(s)
- Chih-Ying Changchien
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan; Department of General Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Meng-Ho Sung
- Department of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan
| | - Hsin-Han Chang
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Sen Peng
- Division of Nephrology, Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan; College of Electrical and Communication Engineering, Yuan Ze University, Taoyuan City, Taiwan.
| | - Ying Chen
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
29
|
Watanabe H, Enoki Y, Maruyama T. Sarcopenia in Chronic Kidney Disease: Factors, Mechanisms, and Therapeutic Interventions. Biol Pharm Bull 2020; 42:1437-1445. [PMID: 31474705 DOI: 10.1248/bpb.b19-00513] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic kidney disease (CKD), a chronic catabolic condition, is characterized by muscle wasting and decreased muscle endurance. Many insights into the molecular mechanisms of muscle wasting in CKD have been obtained. A persistent imbalance between protein degradation and synthesis in muscle causes muscle wasting. During muscle wasting, high levels of reactive oxygen species (ROS) and inflammatory cytokines are detected in muscle. These increased ROS and inflammatory cytokine levels induce the expression of myostatin. The myostatin binding to its receptor activin A receptor type IIB stimulates the expression of atrogenes such as atrogin-1 and muscle ring factor 1, members of the muscle-specific ubiquitin ligase family. Impaired mitochondrial function also contributes to reducing muscle endurance. The increased protein-bound uremic toxin, parathyroid hormone, glucocorticoid, and angiotensin II levels that are observed in CKD all have a negative effect on muscle mass and endurance. Among the protein-bound uremic toxins, indoxyl sulfate, an indole-containing compound has the potential to induce muscle atrophy by stimulating ROS-mediated myostatin and atrogenes expression. Indoxyl sulfate also impairs mitochondrial function. Some potential therapeutic approaches based on the muscle wasting mechanisms in CKD are currently in the testing stages.
Collapse
Affiliation(s)
- Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Yuki Enoki
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
30
|
The Kidney-Gut-Muscle Axis in End-Stage Renal Disease is Similarly Represented in Older Adults. Nutrients 2019; 12:nu12010106. [PMID: 31905970 PMCID: PMC7019845 DOI: 10.3390/nu12010106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/26/2019] [Accepted: 12/27/2019] [Indexed: 12/15/2022] Open
Abstract
Decreased renal function, elevated circulating levels of urea, intestinal levels of urea-degrading bacteria, and gut-derived uremic metabolites are present in end-stage renal disease (ESRD), a cohort that has reduced muscle mass and physical function, and poor muscle composition. This phenotype, defined as the kidney–gut–muscle axis, is similarly represented in older adults that do not have ESRD. The purpose of this short communication is to illuminate these findings, and to propose a strategy that can positively impact the kidney–gut–muscle axis. For example, dietary fiber is fermented by intestinal bacteria, thereby producing the short-chain fatty acids (SCFAs) acetate, propionate, and butyrate, which affect each component of the kidney–gut–muscle axis. Accordingly, a high-fiber diet may be an important approach for improving the kidney–gut–muscle axis in ESRD and in older adults that do not have ESRD.
Collapse
|
31
|
Ryu Y, Lee D, Jung SH, Lee KJ, Jin H, Kim SJ, Lee HM, Kim B, Won KJ. Sabinene Prevents Skeletal Muscle Atrophy by Inhibiting the MAPK-MuRF-1 Pathway in Rats. Int J Mol Sci 2019; 20:ijms20194955. [PMID: 31597276 PMCID: PMC6801606 DOI: 10.3390/ijms20194955] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 09/28/2019] [Accepted: 10/03/2019] [Indexed: 12/21/2022] Open
Abstract
Chrysanthemum boreale Makino essential oil (CBMEO) has diverse biological activities including a skin regenerating effect. However, its role in muscle atrophy remains unknown. This study explored the effects of CBMEO and its active ingredients on skeletal muscle atrophy using in vitro and in vivo models of muscle atrophy. CBMEO reversed the size decrease of L6 myoblasts under starvation. Among the eight monoterpene compounds of CBMEO without cytotoxicity for L6 cells, sabinene induced predominant recovery of reductions of myotube diameters under starvation. Sabinene diminished the elevated E3 ubiquitin ligase muscle ring-finger protein-1 (MuRF-1) expression and p38 mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinase1/2 (ERK1/2) phosphorylations in starved myotubes. Moreover, sabinene decreased the increased level of reactive oxygen species (ROS) in myotubes under starvation. The ROS inhibitor antagonized expression of MuRF-1 and phosphorylation of MAPKs, which were elevated in starved myotubes. In addition, levels of muscle fiber atrophy and MuRF-1 expression in gastrocnemius from fasted rats were reduced after administration of sabinene. These findings demonstrate that sabinene, a bioactive component from CBMEO, may attenuate skeletal muscle atrophy by regulating the activation mechanism of ROS-mediated MAPK/MuRF-1 pathways in starved myotubes, probably leading to the reverse of reduced muscle fiber size in fasted rats.
Collapse
Affiliation(s)
- Yunkyoung Ryu
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Donghyen Lee
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Seung Hyo Jung
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Kyung-Jin Lee
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Hengzhe Jin
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Su Jung Kim
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Hwan Myung Lee
- Department of Cosmetic Science, College of Life and Health Sciences, Hoseo University, 20 Hoseo-ro79beon-gil, Hoseo-ro, Baebang-eup, Asan 31499, Korea.
| | - Bokyung Kim
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Kyung-Jong Won
- Department of Physiology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| |
Collapse
|
32
|
Lipotoxicity in Kidney, Heart, and Skeletal Muscle Dysfunction. Nutrients 2019; 11:nu11071664. [PMID: 31330812 PMCID: PMC6682887 DOI: 10.3390/nu11071664] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/01/2019] [Accepted: 07/17/2019] [Indexed: 12/19/2022] Open
Abstract
Dyslipidemia is a common nutritional and metabolic disorder in patients with chronic kidney disease. Accumulating evidence supports the hypothesis that prolonged metabolic imbalance of lipids leads to ectopic fat distribution in the peripheral organs (lipotoxicity), including the kidney, heart, and skeletal muscle, which accelerates peripheral inflammation and afflictions. Thus, lipotoxicity may partly explain progression of renal dysfunction and even extrarenal complications, including renal anemia, heart failure, and sarcopenia. Additionally, endoplasmic reticulum stress activated by the unfolded protein response pathway plays a pivotal role in lipotoxicity by modulating the expression of key enzymes in lipid synthesis and oxidation. Here, we review the molecular mechanisms underlying lipid deposition and resultant tissue damage in the kidney, heart, and skeletal muscle, with the goal of illuminating the nutritional aspects of these pathologies.
Collapse
|