1
|
Qi H, Gao Y, Zhang Z, Zhang X, Tian D, Jiang Y, Zhang L, Zeng N, Yang R. HouShiHeiSan attenuates sarcopenia in middle cerebral artery occlusion (MCAO) rats. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118917. [PMID: 39423947 DOI: 10.1016/j.jep.2024.118917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/15/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Physical therapy is the main clinical treatment for limb symptoms after ischemic stroke, and there is a lack of reliable drug intervention programs. HouShiHeiSan (HS)comes from "Synopsis of the Golden Chamber", where it is recorded: "seauelae of wind stroke and heaviness of limbs", indicating this formulae is a promising opion for clinical practice. AIM OF THE STUDY The aim of this study is to explore the therapeutic effect of HS on sarcopenia after ischemic stroke (ISS) by using the middle cerebral artery occlusion (MCAO) rats. MATERIALS AND METHODS After 7 days of adaptive feeding Sprague-Dawley (SD) rats were randomly divided into sham and MCAO surgery groups. After MCAO operation, the agreement of the models was evaluated with a laser speckle instrument, and then, treatment groups were administered HS and related solvent. During the 7 days treatment period, the Zea-Longa score was used to assess the neural function, the treadmill for exercise capacity and traction instrument for grip strength. Besides, the physiological electrical signal system was used to record muscular electrical signals, while the muscle thickness was measured by ultrasound. After data acquisition on the 7th day after MCAO operation, the soleus muscle was dissected, and the indexes of length, weight of whole muscle tissue and cross-sectional area of muscular cells by H&E were recorded. Subsequently, mechanistic indicators were examined. MuRF1 and MAFbx expression was detected by immunohistochemistry (IHC). Furthermore, the expression level of more related indicators of muscular differentiation and cellular proterin balance, including mTOR, p-mTOR, AKT, p-AKT, p70s6k, p-p70s6, FOXO1, p-FOXO1, MyoD1, Myostatin, MuRF1 and MAFbx, were tested via Western blot. RESULTS HS improved motor performance and promoted muscle regeneration in MCAO rats. In terms of motor ability, HS mixed with alcohol significantly improved the neurological function damage, reduce the weight loss, increase the running distance per unit time and increase the grip strength. The postoperative muscle electrical signal intensity increased, and muscle thickness, weight, and length were maintained. The HS with alcohol group significantly maintained the cross-sectional size of muscle cells and reduced the number of MyoD1 and myostatin-positive cells in the muscle tissue. It simultaneously promoted the expression of p-mTOR, p-AKT, p-p70s6k, and MyoD1 to promote the synthesis of muscle proteins and inhibited the expression of p-FOXO1, myostatin, MAFbx, and MuRF1 to reduce muscle protein degradation. CONCLUSION HS can enhance muscle protein synthesis and decrease protein breakdown by activating the AKT/mTOR/FOXO1 pathway, thereby preserving muscle health and enhancing motor performance following stroke in rats.
Collapse
Affiliation(s)
- Hu Qi
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yuanlin Gao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zeyang Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiongwei Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Dan Tian
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yanning Jiang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lihong Zhang
- Department of Otorhinolaryngology, Chengdu Xinjin District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan Province, China.
| | - Nan Zeng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Ruocong Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
2
|
Ahmed FE, Hassen EZ, Mousa FME, Abdelfadeel KF. Ameliorating role of co-administration of granulocyte colony stimulating factor and sodium bicarbonate on the skeletal muscle of a rat model of chronic kidney disease (A histological and immunohistochemical study). Ultrastruct Pathol 2025; 49:67-92. [PMID: 39741386 DOI: 10.1080/01913123.2024.2446242] [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: 10/02/2024] [Revised: 11/22/2024] [Accepted: 12/20/2024] [Indexed: 01/03/2025]
Abstract
Over half million individuals suffer from chronic kidney disease (CKD) worldwide. In addition to raising the possibility of cardiovascular diseases, skeletal myopathy remains a challenging complication that is highly correlated with mortality and a lower quality of life. Granulocyte-colony stimulating factor (G-CSF) is an active cytokine for mobilization of immunological and hematopoietic stem cells that can replace exogenous stem cell infusions. So, it is seen as a less expensive and noninvasive tool for regenerative medicine. Sixty three rats were divided into 4 groups: I control, II CKD induced, IIIa, IIIb treated and IV recovery groups. After induction of CKD in all rats, group II were sacrificed after 4 weeks. Rats of group IIIa received NaHCO3. Group IIIb rats were injected subcutaneously by G-CSF as 100 µg/kg/day for 5 successive days in addition to NaHCO3 as group IIIa. Group IV rats were housed for 4 weeks without treatment. Serum urea, creatinine, tissue MDA& TNF-α were assessed. Renal and gastrocnemius muscle sections were evaluated for histological structure, CD34 and myogenin immune expression, morphometric and statistical analyses. The CKD group revealed a significant increase in MDA and TNF-α. Furthermore, features of renal injury, muscle degenerative changes, increased collagen and decreased CD34 and myogenin expression were observed. Alterations were partially attenuated by NaHCO3, while GCSF remarkably improved most parameters. The current results indicated that co-administration of GCSF and NaHCO3 could ameliorate CKD myopathy via attenuating oxidative stress, immunomodulation, pro-angiogenic ability, myocyte regeneration. In addition to the reduction of mitochondrial stress and maintenance of cellular homeostasis.
Collapse
Affiliation(s)
- Fayza E Ahmed
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ebtahal Z Hassen
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Fatma M E Mousa
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Karima F Abdelfadeel
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
3
|
Chang HH, Wang CH, Lin YL, Kuo CH, Liou HH, Hsu BG. Relationship Between Serum Myostatin and Endothelial Function in Non-Dialysis Patients with Chronic Kidney Disease. Diseases 2024; 12:328. [PMID: 39727658 DOI: 10.3390/diseases12120328] [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: 11/16/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Myostatin, primarily produced by skeletal muscle, inhibits muscle growth and promotes protein degradation. It has been implicated in conditions such as obesity, insulin resistance, and cardiovascular disease. However, its association with endothelial function in chronic kidney disease (CKD) patients remains unclear. This study aimed to investigate the relationship between serum myostatin levels and endothelial function in 136 non-dialysis CKD patients at stages 3-5. METHODS Fasting blood samples were collected to measure serum myostatin levels using enzyme-linked immunosorbent assay kits. Endothelial function was evaluated non-invasively by measuring the vascular reactivity index (VRI) with a digital thermal monitoring test. RESULTS VRI values were classified as poor (<1.0, n = 25, 18.4%), intermediate (1.0 to <2.0, n = 63, 46.3%), or good (≥2.0, n = 48, 35.3%). Factors associated with poor vascular reactivity included older age (p = 0.026), elevated serum blood urea nitrogen (p = 0.020), serum creatinine (p = 0.021), urine protein-to-creatinine ratio (UPCR, p = 0.013), and myostatin levels (p = 0.003), along with reduced estimated glomerular filtration rate (p = 0.015). Multivariate regression analysis identified older age, higher serum creatinine, and log-transformed myostatin levels as significant independent predictors of lower VRI. CONCLUSIONS These findings suggest that myostatin may serve as a potential biomarker for endothelial dysfunction in CKD patients. Future large-scale, longitudinal studies are warranted to confirm and extend our preliminary findings.
Collapse
Affiliation(s)
- Ho-Hsiang Chang
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Chih-Hsien Wang
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Yu-Li Lin
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan
| | - Chiu-Huang Kuo
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Hung-Hsiang Liou
- Division of Nephrology, Department of Internal Medicine, Hsin-Jen Hospital, New Taipei City 24243, Taiwan
| | - Bang-Gee Hsu
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan
| |
Collapse
|
4
|
Shi A, He C, Otten K, Wu G, Forouzanfar T, Wüst RCI, Jaspers RT. Reduced myotube diameter induced by combined inhibition of transforming growth factor-β type I receptors Acvr1b and Tgfbr1 is associated with enhanced β1-syntrophin expression. J Cell Physiol 2024; 239:e31418. [PMID: 39164996 PMCID: PMC11649968 DOI: 10.1002/jcp.31418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Simultaneous inhibition of transforming growth factor-β (TGF-β) type I receptors Acvr1b and Tgfbr1 signalling has been associated with excessive skeletal muscle hypertrophy in vivo. However, it remains unclear whether the increased muscle mass in vivo is a direct result of inhibition of intracellular TGF-β signalling or whether this is an indirect effect of an altered extracellular anabolic environment. Here, we tested whether individual or simultaneous knockdown of TGF-β type I receptors in C2C12 myotubes was sufficient to induce muscle hypertrophy. The expression levels of TGF-β type I receptors Acvr1b and Tgfbr1 in myotubes were knocked down individually or in combination in the absence or presence of TGF-β1 and myostatin. Knocking down either Acvr1b or Tgfbr1 did not significantly change cell phenotype. Unexpectedly, simultaneous knockdown of both receptors reduced C2C12 myotube diameter, mRNA expression levels of Hgf, Ccn2 and Mymx with or without TGF-β1 and myostatin administration. In spite of decreased phosphorylation of Smad2/3, phosphorylation of P70S6K was reduced. In addition, the gene expression level of β1-syntrophin (Sntb1), which encodes a protein associated with the dystrophin-glycoprotein complex, was increased. Parallel experiments where Sntb1 gene expression was reduced showed an increase in myotube diameter and fusion of C2C12 myoblasts. Together, these results indicate that the knockdown of both TGF-β type I receptors reduced myotube diameter. This atrophic effect was attributed to reduced protein synthesis signalling and an increased expression of β1-syntrophin. These results have implications for our fundamental understanding of how TGF-β signalling regulates skeletal muscle size.
Collapse
Affiliation(s)
- Andi Shi
- Laboratory for Myology, Department of Human Movement SciencesFaculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of ProsthodonticsAffiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and ReconstructionGuangzhouChina
| | - Chuqi He
- Laboratory for Myology, Department of Human Movement SciencesFaculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Kirsten Otten
- Laboratory for Myology, Department of Human Movement SciencesFaculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Gang Wu
- Department of Oral Cell BiologyAcademic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU)AmsterdamThe Netherlands
| | - Tymour Forouzanfar
- Department of Oral Cell BiologyAcademic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU)AmsterdamThe Netherlands
- Department of Oral and Maxillofacial SurgeryLeiden University Medical CenterLeidenThe Netherlands
| | - Rob C. I. Wüst
- Laboratory for Myology, Department of Human Movement SciencesFaculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Richard T. Jaspers
- Laboratory for Myology, Department of Human Movement SciencesFaculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Department of ProsthodonticsAffiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and ReconstructionGuangzhouChina
| |
Collapse
|
5
|
Yin L, Wu S, Bai P, Wang X. Combination of transcriptomics and proteomics for analyzing potential biomarker and molecular mechanism underlying skeletal muscle atrophy. J Proteomics 2024; 309:105283. [PMID: 39179024 DOI: 10.1016/j.jprot.2024.105283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/11/2024] [Accepted: 08/15/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND The skeletal muscle atrophy is prevalently occurred in numerous chronic disease complications. Despite its important clinical significance, there are currently no therapeutic drugs, so new biomarkers and molecular mechanisms need to be discovered urgently. METHODS Transcriptome and proteome sequencing data were collected from normal and skeletal muscle atrophic mice. The differentially expressed genes (DEGs) and proteins (DEPs) were analyzed. Applying PPI analysis to obtain overlapping genes and proteins, which were next subjected to GO and KEGG enrichment analysis. Combined analysis of transcriptomics and proteomics was performed to get key genes that were simultaneously found in GO and KEGG enrichment results. Subsequently, RT-qPCR and immunofluorescence were constructed to verify the expression of screened key genes. RESULTS By combination of transcriptomics, proteomics and RT-qPCR results, we identified 14 key genes (Cav1, Col3a1, Dnaja1, Postn, Ptges3, Cd44, Clec3b, Igfbp6, Lamc1, Alb, Itga6, Mmp2, Timp2 and Cd9) that were markedly different in atrophic mice. Single-gene GSEA and immunofluorescence suggested Cd9 was probably the biomarker for skeletal muscle atrophy. CONCLUSIONS Our study hinted that Cd9 was potential biomarker and may interfere with skeletal muscle atrophy through process of aerobic respiration, oxidative phosphorylation, and metabolism of amino acids and fatty acids. SIGNIFICANCE The present study holds the subsequent significance: Frist, we investigated biomarkers for skeletal muscle atrophy using multi-omics approach. A total of 14 genes were markedly different in skeletal muscle atrophic mice. We finally found Cd9 is a potential biomarker for skeletal muscle atrophy. Our work presents novel biomarkers and potential regulatory mechanisms for the early detection and intervention of muscle atrophy.
Collapse
Affiliation(s)
- Lin Yin
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital,Third Hospital of Shanxi Medical University, Taiyuan 030032, China; Beijing Key Laboratory of Drug Target Identification and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Shasha Wu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital,Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Peirong Bai
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital,Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Xuena Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital,Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| |
Collapse
|
6
|
Chen H, Yang C, Yan S, Liu X, Zhou L, Yuan X. Sarcopenia in cirrhosis: From pathophysiology to interventional therapy. Exp Gerontol 2024; 196:112571. [PMID: 39236869 DOI: 10.1016/j.exger.2024.112571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
Sarcopenia, characterized by the loss of skeletal muscle mass and function, is a significant complication in patients with cirrhosis. This condition not only exacerbates the overall morbidity and mortality associated with liver disease but also complicates patient management, increasing the risk of hospitalization, infections, and hepatic encephalopathy. Despite its clinical significance, sarcopenia in cirrhotic patients remains underdiagnosed and undertreated. This review aims to summarize current knowledge on the pathophysiology of sarcopenia in cirrhosis, including mechanisms such as altered metabolism, hormonal imbalances, and inflammation. Additionally, we explore diagnostic challenges and discuss emerging therapeutic strategies, including nutritional support, exercise, and pharmacological interventions. By highlighting the gaps in existing research and proposing directions for future studies, this review seeks to improve the management and outcomes of cirrhotic patients affected by sarcopenia.
Collapse
Affiliation(s)
- Huiling Chen
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai 201399, China; Fudan University, Shanghai, China
| | - Chenyun Yang
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai 201399, China
| | - Shijie Yan
- Department of General Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai 201399, China
| | - Xintao Liu
- Department of General Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai 201399, China
| | - Ligang Zhou
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai 201399, China; Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai, China
| | - Xinlu Yuan
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai 201399, China.
| |
Collapse
|
7
|
Zhang Y, Liu R, Wang W, Wu RJ, Dai WT, Zhang CL, Zhuang Q, Li XH. Association of Myostatin With Complications and Cognition in Lung Cancer Patients With Sarcopenia. J Surg Res 2024; 302:240-249. [PMID: 39111127 DOI: 10.1016/j.jss.2024.07.054] [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: 10/19/2023] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 10/20/2024]
Abstract
INTRODUCTION The risk of surgery and postoperative complications increases greatly in frail older patients with sarcopenia. The purpose of this study is to explore the correlation between myostatin (MSTN) levels and cognitive function and postoperative pulmonary complications (PPCs) in older patients undergoing thoracoscopic lobectomy and to determine whether MSTN could be used to predict the risk of postoperative complications and cognitive impairment. METHODS A prospective observational study was conducted at the First Affiliated Hospital of Bengbu Medical College, China, between January 2023 and June 2023. The risk factors of PPCs and postoperative cognitive impairment were studied using backward stepwise logistic regression analysis. The independent factors were formed into a linear regression equation to construct a risk score model for each patient. The 122 patients who participated in the study were divided into two groups, a low-level group and a high-level group, based on an MSTN level cut-off; the preoperative MSTN cut-off values was 25.55 ng/mL for cognitive dysfunction and 22.29 ng/mL for PPCs. The PPCs and cognitive function of the groups were compared. RESULTS Preoperative MSTN was confirmed as a risk factor for postoperative cognitive dysfunction and PPCs. After surgery, the proportion of patients with cognitive impairment in the high-level group was significantly higher than in the low-level group (P < 0.001). In the high-level group, the incidence of respiratory tract infections was 17.9% higher (P = 0.021), hypoxaemia was 20.5% higher (P = 0.001) and respiratory failure was 14.4% higher (P = 0.012) than in the low-level group. In addition, a high level of MSTN increased the length of hospital stay (P < 0.001) and decreased the Barthel Index score (P < 0.001). CONCLUSIONS The study findings suggest that MSTN could be used as an index to predict complications and cognitive impairment after thoracoscopic lobectomy in older patients with sarcopenia and to provide evidence for reducing postoperative cognitive impairment and PPCs.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Rui Liu
- Department of Anesthesia, Bengbu Medical College, Bengbu, China
| | - Wei Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Ruo-Jie Wu
- Department of Anesthesia, Bengbu Medical College, Bengbu, China
| | - Wen-Tao Dai
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Cong-Li Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Qin Zhuang
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiao-Hong Li
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China.
| |
Collapse
|
8
|
Karava V, Kondou A, Dotis J, Christoforidis A, Taparkou A, Farmaki E, Printza N. Fibroblast growth-factor 23-Klotho axis is associated with systemic inflammation and myokine profile in children with chronic kidney disease. Hormones (Athens) 2024; 23:517-526. [PMID: 39112785 DOI: 10.1007/s42000-024-00586-3] [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: 11/14/2023] [Accepted: 07/16/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Chronic kidney disease is linked to a disturbed fibroblast growth factor-23 (FGF23)-Klotho axis and an imbalance between myostatin and insulin-like growth factor-1 (IGF-1) expression. This cross-sectional study investigates the association of the FGF23-Klotho axis and myokine profile with serum interleukin-6 (IL-6) and their interactions in pediatric patients. METHODS Serum calcium, phosphorus, 25-hydroxyvitamin D, parathormone, c-terminal FGF23, a-Klotho, myostatin, follistatin, IGF-1, and IL-6 were measured in 53 patients with GFR < 60 ml/min/1,73m2. Myostatin to lean mass (LM) and to IGF-1 ratios were calculated. IL-6 level > 3rd quartile was considered as high. RESULTS Myostatin, IGF-1, and follistatin were correlated to LM (rs = 0.513, p < 0.001, rs = 0.652, p < 0.001, rs=-0.483, p < 0.001). Myostatin and follistatin were correlated to IGF-1 (rs = 0.340, p = 0.014, rs=-0.385, p = 0.005). Myostatin/LM but not myostatin or myostatin/IGF-1 ratio was significantly higher in CKD 5D patients (p = 0.001,p = 0.844, p = 0.111). Among mineral bone parameters, lnFGF23 was correlated to lnIL-6 (rs = 0.397, p = 0.004) and associated with high IL-6 (OR 1.905, 95% CI 1.023-3.548). Among myokines, myostatin/IGF-1 ratio was correlated to lnIL-6 (rs = 0.395, p = 0.004) and associated with high IL-6 (OR 1.113, 95% CI 1.028-1.205). All associations were adjusted to CKD stage. Myostatin was correlated to lnFGF23 (rs = 0.331, p = 0.025) and myostatin/IGF-1 ratio to lnKlotho (rs=-0.363, p = 0.013), after adjustment for CKD stage, lnIL-6 and other mineral bone parameters. CONCLUSIONS In pediatric CKD, FGF23 and myostatin/IGF-1 ratio are associated with IL-6, indicating a link between systemic inflammation, mineral bone, and myokine disorders. The correlations between myostatin and FGF23 and between myostatin/IGF-1 and Klotho suggest an interaction between mineral bone and muscle metabolism.
Collapse
Affiliation(s)
- Vasiliki Karava
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, 49 Konstantinoupoleos Street, Thessaloniki, 54642, Greece.
| | - Antonia Kondou
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, 49 Konstantinoupoleos Street, Thessaloniki, 54642, Greece
| | - John Dotis
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, 49 Konstantinoupoleos Street, Thessaloniki, 54642, Greece
| | - Athanasios Christoforidis
- Pediatric Endocrinology Unit, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anna Taparkou
- Pediatric Immunology and Rheumatology Referral Center, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Farmaki
- Pediatric Immunology and Rheumatology Referral Center, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikoleta Printza
- Pediatric Nephrology Unit, 1st Department of Pediatrics, Hippokratio General Hospital, Aristotle University of Thessaloniki, 49 Konstantinoupoleos Street, Thessaloniki, 54642, Greece
| |
Collapse
|
9
|
Sheng L, Sun J, Huang L, Yu M, Meng X, Shan Y, Dai H, Wang F, Shi J, Sheng M. Astragalus membranaceus and its monomers treat peritoneal fibrosis and related muscle atrophy through the AR/TGF-β1 pathway. Front Pharmacol 2024; 15:1418485. [PMID: 39239655 PMCID: PMC11374727 DOI: 10.3389/fphar.2024.1418485] [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: 04/16/2024] [Accepted: 07/16/2024] [Indexed: 09/07/2024] Open
Abstract
Background: To anticipate the potential molecular mechanism of Astragalus membranaceus (AM) and its monomer, Calycosin, against peritoneal fibrosis (PF) and related muscle atrophy using mRNA-seq, network pharmacology, and serum pharmacochemistry. Methods: Animal tissues were examined to evaluate a CKD-PF mice model construction. mRNA sequencing was performed to find differential targets. The core target genes of AM against PF were screened through network pharmacology analysis, and CKD-PF mice models were given high- and low-dose AM to verify common genes. Serum pharmacochemistry was conducted to clarify which components of AM can enter the blood circulation, and the selected monomer was further validated through cell experiments for the effect on PF and mesothelial mesenchymal transition (MMT) of peritoneal mesothelial cells (PMCs). Results: The CKD-PF mice models were successfully constructed. A total of 31,184 genes were detected in the blank and CKD-PF groups, and 228 transcription factors had significant differences between the groups. Combined with network pharmacology analysis, a total of 228 AM-PF-related targets were identified. Androgen receptor (AR) was the remarkable transcription factor involved in regulating transforming growth factor-β1 (TGF-β1). AM may be involved in regulating the AR/TGF-β1 signaling pathway and may alleviate peritoneal dialysis-related fibrosis and muscle atrophy in CKD-PF mice. In 3% peritoneal dialysis solution-stimulated HMrSV5 cells, AR expression levels were dramatically reduced, whereas TGF-β1/p-smads expression levels were considerably increased. Conclusion: AM could ameliorate PF and related muscle atrophy via the co-target AR and modulated AR/TGF-β1 pathway. Calycosin, a monomer of AM, could partially reverse PMC MMT via the AR/TGF-β1/smads pathway. This study explored the traditional Chinese medicine theory of "same treatment for different diseases," and supplied the pharmacological evidence of "AM can treat flaccidity syndrome."
Collapse
Affiliation(s)
- Li Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
- Medical Experimental Research Center, First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinyi Sun
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liyan Huang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Manshu Yu
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaohui Meng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Shan
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Huibo Dai
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Funing Wang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jun Shi
- School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meixiao Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
10
|
Czaja-Stolc S, Chatrenet A, Potrykus M, Ruszkowski J, Torreggiani M, Lichodziejewska-Niemierko M, Dębska-Ślizień A, Piccoli GB, Małgorzewicz S. Adipokines and Myokines as Markers of Malnutrition and Sarcopenia in Patients Receiving Kidney Replacement Therapy: An Observational, Cross-Sectional Study. Nutrients 2024; 16:2480. [PMID: 39125361 PMCID: PMC11314363 DOI: 10.3390/nu16152480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Chronic kidney disease (CKD) is linked to an elevated risk of malnutrition and sarcopenia, contributing to the intricate network of CKD-related metabolic disorders. Adipokines and myokines are markers and effectors of sarcopenia and nutritional status. The aim of this study was to assess whether the adipokine-myokine signature in patients on kidney replacement therapy could help identify malnutrition and sarcopenia. The study involved three groups: 84 hemodialysis (HD) patients, 44 peritoneal dialysis (PD) patients, and 52 kidney transplant recipients (KTR). Mean age was 56.1 ± 16.3 years. Malnutrition was defined using the 7-Point Subjective Global Assessment (SGA) and the Malnutrition-Inflammation Score (MIS). Sarcopenia was diagnosed based on reduced handgrip strength (HGS) and diminished muscle mass. Concentrations of adipokines and myokines were determined using the enzyme-linked immunosorbent assay (ELISA). 32.8% of all study participants were identified as malnourished and 20.6% had sarcopenia. For malnutrition, assessed using the 7-Point SGA, in ROC analysis albumin (area under the curve (AUC) 0.67 was the best single biomarker identified. In dialysis patients, myostatin (AUC 0.79) and IL-6 (AUC 0.67) had a high discrimination value for sarcopenia, and we were able to develop a prediction model for sarcopenia, including age, albumin, adiponectin, and myostatin levels, with an AUC of 0.806 (95% CI: 0.721-0.891). Adipokines and myokines appear to be useful laboratory markers for assessing malnutrition and sarcopenia. The formula we propose could contribute to a better understanding of sarcopenia and potentially lead to more effective interventions and management strategies for dialysis patients.
Collapse
Affiliation(s)
- Sylwia Czaja-Stolc
- Department of Clinical Nutrition and Dietetics, Faculty of Health Sciences, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.C.-S.); (S.M.)
| | - Antoine Chatrenet
- Department of Nephrology, Centre Hospitalier du Mans, 72037 Le Mans, France; (A.C.); (M.T.); (G.B.P.)
- APCoSS—Institute of Physical Education and Sports Sciences (IFEPSA), UCO, 49136 Angers, France
| | - Marta Potrykus
- Department of Oncological, Transplant, and General Surgery, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland
| | - Jakub Ruszkowski
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland; (J.R.); (A.D.-Ś.)
| | - Massimo Torreggiani
- Department of Nephrology, Centre Hospitalier du Mans, 72037 Le Mans, France; (A.C.); (M.T.); (G.B.P.)
| | | | - Alicja Dębska-Ślizień
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland; (J.R.); (A.D.-Ś.)
| | - Giorgina Barbara Piccoli
- Department of Nephrology, Centre Hospitalier du Mans, 72037 Le Mans, France; (A.C.); (M.T.); (G.B.P.)
- Department of Nephrology, University of Angers, 49035 Angers, France
| | - Sylwia Małgorzewicz
- Department of Clinical Nutrition and Dietetics, Faculty of Health Sciences, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.C.-S.); (S.M.)
| |
Collapse
|
11
|
Picciotto D, Macciò L, Verzola D, Baciga F, Momentè C, Russo E, Viazzi F, Battaglia Y, Esposito P. Pathophysiology of Physical Exercise in Kidney Patients: Unveiling New Players - The Role of Myokines. Kidney Blood Press Res 2024; 49:457-471. [PMID: 38815556 DOI: 10.1159/000539489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is a progressive systemic condition characterized by numerous complications. Among these, alterations in skeletal muscle physiology, such as sarcopenia, are particularly significant, as they are associated with poor outcomes and reduced quality of life. SUMMARY Various interventions, including pharmacological approaches and lifestyle modifications have been investigated to slow CKD progression and prevent or treat its complications. Physical exercise, in particular, has emerged as a promising intervention with multiple beneficial effects. These include improvements in physical functioning, increased muscle mass, modulation of metabolic abnormalities, and reduced cardiovascular risk. However, the pathophysiology of physical exercise in patients with kidney disease is complex and remains only partially understood. A crucial advancement in understanding this phenomenon has been the identification of myokines - molecules expressed and released by skeletal muscle in response to physical activity. These myokines can exert both paracrine and systemic effects, influencing not only skeletal muscle physiology but also other processes such as energy metabolism and lipid regulation. KEY MESSAGES The interplay among skeletal muscle, physical activity, and myokines may act as a pivotal regulator in various physiological processes, including aging, as well as in pathological conditions like cachexia and sarcopenia, frequently observed in CKD patients at different stages, including patients on dialysis. Despite the potential importance of this relationship, only a limited number of studies have explored the relationship between exercise and myokine, and the effect of this interaction on experimental models or individuals with kidney disease. In the following sections, we review and discuss this topic.
Collapse
Affiliation(s)
- Daniela Picciotto
- Nephrology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lucia Macciò
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genoa, Italy
| | - Daniela Verzola
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genoa, Italy
| | - Federica Baciga
- Department of Medicine, University of Verona, Verona, Italy
- Nephrology and Dialysis Unit, Pederzoli Hospital, Peschiera del Garda, Italy
| | | | - Elisa Russo
- Nephrology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genoa, Italy
| | - Francesca Viazzi
- Nephrology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genoa, Italy
| | - Yuri Battaglia
- Department of Medicine, University of Verona, Verona, Italy
- Nephrology and Dialysis Unit, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Pasquale Esposito
- Nephrology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genoa, Italy
| |
Collapse
|
12
|
Heitman K, Alexander MS, Faul C. Skeletal Muscle Injury in Chronic Kidney Disease-From Histologic Changes to Molecular Mechanisms and to Novel Therapies. Int J Mol Sci 2024; 25:5117. [PMID: 38791164 PMCID: PMC11121428 DOI: 10.3390/ijms25105117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic kidney disease (CKD) is associated with significant reductions in lean body mass and in the mass of various tissues, including skeletal muscle, which causes fatigue and contributes to high mortality rates. In CKD, the cellular protein turnover is imbalanced, with protein degradation outweighing protein synthesis, leading to a loss of protein and cell mass, which impairs tissue function. As CKD itself, skeletal muscle wasting, or sarcopenia, can have various origins and causes, and both CKD and sarcopenia share common risk factors, such as diabetes, obesity, and age. While these pathologies together with reduced physical performance and malnutrition contribute to muscle loss, they cannot explain all features of CKD-associated sarcopenia. Metabolic acidosis, systemic inflammation, insulin resistance and the accumulation of uremic toxins have been identified as additional factors that occur in CKD and that can contribute to sarcopenia. Here, we discuss the elevation of systemic phosphate levels, also called hyperphosphatemia, and the imbalance in the endocrine regulators of phosphate metabolism as another CKD-associated pathology that can directly and indirectly harm skeletal muscle tissue. To identify causes, affected cell types, and the mechanisms of sarcopenia and thereby novel targets for therapeutic interventions, it is important to first characterize the precise pathologic changes on molecular, cellular, and histologic levels, and to do so in CKD patients as well as in animal models of CKD, which we describe here in detail. We also discuss the currently known pathomechanisms and therapeutic approaches of CKD-associated sarcopenia, as well as the effects of hyperphosphatemia and the novel drug targets it could provide to protect skeletal muscle in CKD.
Collapse
Affiliation(s)
- Kylie Heitman
- Division of Nephrology and Section of Mineral Metabolism, Department of Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Matthew S. Alexander
- Division of Neurology, Department of Pediatrics, The University of Alabama at Birmingham and Children’s of Alabama, Birmingham, AL 35294, USA
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christian Faul
- Division of Nephrology and Section of Mineral Metabolism, Department of Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| |
Collapse
|
13
|
He J, He Z, Wang H, Zhang C, Pei T, Yan S, Yan Y, Wang F, Chen Y, Yuan N, Wang M, Xiao W. Caffeic acid alleviates skeletal muscle atrophy in 5/6 nephrectomy rats through the TLR4/MYD88/NF-kB pathway. Biomed Pharmacother 2024; 174:116556. [PMID: 38636398 DOI: 10.1016/j.biopha.2024.116556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 04/20/2024] Open
Abstract
Skeletal muscle atrophy is a common complication of chronic kidney disease (CKD) that affects the quality of life and prognosis of patients. We aimed to investigate the effects and mechanisms of caffeic acid (CA), a natural phenolic compound, on skeletal muscle atrophy in CKD rats. Male Sprague-Dawley rats underwent 5/6 nephrectomy (NPM) and were treated with CA (20, 40, or 80 mg/kg/day) for 10 weeks. The body and muscle weights, renal function, hemoglobin, and albumin were measured. The histological, molecular, and biochemical changes in skeletal muscles were evaluated using hematoxylin-eosin staining, quantitative real-time PCR, malondialdehyde/catalase/superoxide dismutase/glutathione level detection, and enzyme-linked immunosorbent assay. Western blotting and network pharmacology were applied to identify the potential targets and pathways of CA, CKD, and muscle atrophy. The results showed that CA significantly improved NPM-induced muscle-catabolic effects, reduced the expression of muscle atrophy-related proteins (muscle atrophy F-box and muscle RING finger 1) and proinflammatory cytokines (interleukin [IL]-6, tumor necrosis factor-alpha, and IL-1β), and attenuated muscle oxidative stress. Network pharmacology revealed that CA modulated the response to oxidative stress and nuclear factor kappa B (NF-κB) signaling pathway and that Toll-like receptor 4 (TLR4) was a key target. In vivo experiment confirmed that CA inhibited the TLR4/myeloid differentiation primary response 88 (MYD88)/NF-kB signaling pathway, reduced muscle iron levels, and restored glutathione peroxidase 4 activity, thereby alleviating ferroptosis and inflammation in skeletal muscles. Thus, CA might be a promising therapeutic agent for preventing and treating skeletal muscle atrophy in CKD by modulating the TLR4/MYD88/NF-κB pathway and ferroptosis.
Collapse
Affiliation(s)
- Jinyue He
- School of traditional Chinese medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhuoen He
- School of traditional Chinese medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hao Wang
- School of traditional Chinese medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Chi Zhang
- School of traditional Chinese medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Tingting Pei
- School of traditional Chinese medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shihua Yan
- School of traditional Chinese medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yangtian Yan
- School of traditional Chinese medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Fujing Wang
- School of traditional Chinese medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yuchi Chen
- School of traditional Chinese medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ningning Yuan
- School of traditional Chinese medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Mingqing Wang
- School of traditional Chinese medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Wei Xiao
- School of traditional Chinese medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
14
|
Sakashita M, Hamasaki Y, Oki R, Komaru Y, Miyamoto Y, Yoshida T, Matsuura R, Doi K, Nangaku M. Serum Myostatin at Dialysis Initiation May Predict 1-Year Mortality and Hospitalization. Nephron Clin Pract 2024; 148:544-552. [PMID: 38522414 PMCID: PMC11332307 DOI: 10.1159/000538533] [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: 10/17/2023] [Accepted: 03/22/2024] [Indexed: 03/26/2024] Open
Abstract
OBJECTIVE Myostatin, which is known as a negative skeleton muscle regulator, is associated with mortality in maintenance hemodialysis patients. However, the significance of serum myostatin concentrations at dialysis initiation has not been established. We investigated the relation between serum myostatin concentrations and mortality or hospitalization within 1 year in incident dialysis patients. METHODS After a patient initiating hemodialysis or peritoneal dialysis during 2016-2018 was enrolled, the patient's serum myostatin at dialysis initiation was measured. Composite outcomes comprising mortality and hospitalization within 1 year after dialysis initiation were compared between two groups divided according to myostatin levels. The Cox proportional hazards model was used to assess significant relations between myostatin and outcomes. RESULTS This study examined 104 incident dialysis patients with a mean age of 65.5 ± 14.0 years (68% male). Kaplan-Meier analyses indicated the 1-year hospitalization-free and survival rate as significantly lower in the lower myostatin group than in the higher myostatin group (p = 0.0020). Cox proportional hazards regression analyses revealed that the value of myostatin logarithm at dialysis initiation was inversely associated with the occurrence of a composite outcome, independently of age (hazard ratio 0.16, 95% confidence interval: 0.05-0.57). Receiver operating characteristic analysis showed the area under the curve of serum myostatin for predicting death or hospitalization within 1 year as higher than those of clinical indices of nutritional disturbance and frailty. CONCLUSION Serum myostatin concentration at dialysis initiation is inversely associated with adverse outcomes in these dialysis-initiated patients.
Collapse
Affiliation(s)
- Midori Sakashita
- Department of Hemodialysis and Apheresis, The University of Tokyo Hospital, Tokyo, Japan,
| | - Yoshifumi Hamasaki
- Department of Hemodialysis and Apheresis, The University of Tokyo Hospital, Tokyo, Japan
| | - Rikako Oki
- Department of Hemodialysis and Apheresis, The University of Tokyo Hospital, Tokyo, Japan
| | - Yohei Komaru
- Department of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo, Japan
| | - Yoshihisa Miyamoto
- Department of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo, Japan
| | - Teruhiko Yoshida
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ryo Matsuura
- Department of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo, Japan
| | - Kent Doi
- Department of Emergency and Critical Care Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Masaomi Nangaku
- Department of Hemodialysis and Apheresis, The University of Tokyo Hospital, Tokyo, Japan
- Department of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
15
|
de Souza Francisco D, Moraes IG, Brito CP, Righetti RF, Yamaguti WP. The phase angle cut-off point capable of discriminating hemodialysis patients with reduced exercise tolerance: a cross-sectional study. BMC Sports Sci Med Rehabil 2024; 16:34. [PMID: 38308310 PMCID: PMC10835815 DOI: 10.1186/s13102-024-00825-5] [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: 04/11/2023] [Accepted: 01/24/2024] [Indexed: 02/04/2024]
Abstract
BACKGROUND Phase angle (PhA) is a prognostic marker of all-cause mortality in chronic kidney disease. However, no study has investigated this marker as a predictor of exercise intolerance in hemodialysis (HD) patients. The aim of this study was to determine a cut-off point for the PhA capable of discriminating HD patients with reduced exercise tolerance. METHODS Thirty-one patients (80.6% men, median age 69 years) were included. The evaluations were performed on three different days, before the HD session. The outcomes evaluated were: biochemical markers, inflammatory and nutritional status, body composition, peripheral muscle strength and exercise tolerance. Performance ≤50% of the predicted value in the six-minute step test (6MST) was defined as reduced exercise tolerance. RESULTS Patients presented an average of 67.6 steps (50.5% of predicted) in the 6MST. Fifteen patients (48.4%) were classified with reduced exercise tolerance. The receiver operating characteristic curve indicated a cut-off point of 3.73° for the PhA (sensitivity = 87%, specificity = 81%, and area under the curve = 0.88 [95% CI: 0.76-1.00]; p < 0.001). Patients with reduced exercise tolerance had worse inflammatory and nutritional status, lower PhA and greater impairment of peripheral muscle strength. CONCLUSION The cut-off point of 3.73° for the PhA is sensitive and specific to discriminate HD patients with reduced exercise tolerance. TRIAL REGISTRATION This study was registered in the Clinical Trials database (no. NCT03779126, date of first registration 19/12/2018).
Collapse
Affiliation(s)
| | | | - Camila Porto Brito
- Hospital Sírio-Libanês, Rehabilitation Service, São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
16
|
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
|
17
|
Ibrahim AH, Kasim SA, Ezzat AA, Ibrahim NE, Hassan DA, Ibrahim AS, Abouelgreed TA, Abdo EM, Aboelsoud NM, Abdelmonem NM, Alnajem MT, Aboomar AA. Relation between myostatin levels and malnutrition and muscle wasting in hemodialysis patients. Arch Ital Urol Androl 2023; 95:11869. [PMID: 38117215 DOI: 10.4081/aiua.2023.11869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND AND AIM Malnutrition is one of the most troublesome comorbidities among hemodialysis patients (HD). Myostatin (MSTN) belongs to the transforming growth factor-β superfamily. In HD patients, MSTN effects are not limited to skeletal muscle growth. The present study aimed to assess MSTN levels in HD patients and its relation to various clinical and biochemical parameters. PATIENTS AND METHODS The present case control study included 60 patients on HD for at least three years. In addition, there were age and sex-matched healthy subjects who constitutes the control group. Nutritional status was evaluated using the malnutrition inflammation score (MIS). Muscle wasting in the present study was evaluated using the lean tissue index (LTI) as assessed by the body composition monitor (BCM). Rectus Femoris Muscle (RFM) thickness was also measured as indicator for nutritional status of patient. RESULTS The present study included 60 HD patients, and ageand sex-matched healthy controls. Patients expressed significantly higher myostatin levels when compared to controls [median (IQR): 221.3 (153.5-688.2) versus 144.8 (97.0-281.7), p < 0.001]. According to MIS, patients were classified into those with no/mild malnutrition (n = 22) and others with moderate/severe malnutrition (n = 38). Comparison between the two subgroups revealed that the former group had significantly lower myostatin levels [167.7 (150.3-236.3) versus 341.7 (160.9-955.9), p = 0.004]. According to LTI, patients were classified into those with muscle wasting (n = 23) and others without muscle wasting (n = 37). Comparative analysis showed that patients in the former group had significantly higher myostatin levels [775.1 (325.1-2133.7) versus 161.8 (142.6-302.3), p < 0.001]. CONCLUSIONS Myostatin seems to be a promising marker for identification of malnutrition and muscle wasting in HD patients.
Collapse
Affiliation(s)
- Amal H Ibrahim
- Department of Internal Medicine, Nephrology Unit, Al-Azhar University, Cairo.
| | - Sammar A Kasim
- Department of Internal Medicine, Nephrology Unit, Al-Azhar University, Cairo.
| | - Alshimaa A Ezzat
- Department of Radiology, Faculty of Medicine, Al-Azhar University, Cairo.
| | - Noha E Ibrahim
- Department of Microbial Biotechnology, Biotechnology Research Institute, National Research Centre (NRC), Giza.
| | - Donia A Hassan
- Department of Clinical Pathology, Al-Azhar University, Cairo.
| | - Amira Sh Ibrahim
- Department of Rheumatology and Rehabilitation, Faculty of Medicine for girls, Al-Azhar University, Cairo.
| | | | - Ehab M Abdo
- Department of Vascular Surgery, Faculty of Medicine, Al-Azhar University, Cairo.
| | - Naglaa M Aboelsoud
- Department of Radiology, Faculty of Medicine, Al-Azhar University, Cairo.
| | | | | | - Ahmed A Aboomar
- Department of internal medicine, Nephrology Unit, Faculty Medicine, Tanta University, Tanta.
| |
Collapse
|
18
|
Bellafronte NT, Barril G, de Oliveira EP. Editorial: Improving body composition and functional capacity in chronic kidney disease patients. Front Nutr 2023; 10:1223975. [PMID: 37575328 PMCID: PMC10420043 DOI: 10.3389/fnut.2023.1223975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/11/2023] [Indexed: 08/15/2023] Open
Affiliation(s)
| | - Guillermina Barril
- Department of Nephrology, Hospital Universitario de la Princesa, Madrid, Spain
| | | |
Collapse
|
19
|
Hung KC, Yao WC, Liu YL, Yang HJ, Liao MT, Chong K, Peng CH, Lu KC. The Potential Influence of Uremic Toxins on the Homeostasis of Bones and Muscles in Chronic Kidney Disease. Biomedicines 2023; 11:2076. [PMID: 37509715 PMCID: PMC10377042 DOI: 10.3390/biomedicines11072076] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Patients with chronic kidney disease (CKD) often experience a high accumulation of protein-bound uremic toxins (PBUTs), specifically indoxyl sulfate (IS) and p-cresyl sulfate (pCS). In the early stages of CKD, the buildup of PBUTs inhibits bone and muscle function. As CKD progresses, elevated PBUT levels further hinder bone turnover and exacerbate muscle wasting. In the late stage of CKD, hyperparathyroidism worsens PBUT-induced muscle damage but can improve low bone turnover. PBUTs play a significant role in reducing both the quantity and quality of bone by affecting osteoblast and osteoclast lineage. IS, in particular, interferes with osteoblastogenesis by activating aryl hydrocarbon receptor (AhR) signaling, which reduces the expression of Runx2 and impedes osteoblast differentiation. High PBUT levels can also reduce calcitriol production, increase the expression of Wnt antagonists (SOST, DKK1), and decrease klotho expression, all of which contribute to low bone turnover disorders. Furthermore, PBUT accumulation leads to continuous muscle protein breakdown through the excessive production of reactive oxygen species (ROS) and inflammatory cytokines. Interactions between muscles and bones, mediated by various factors released from individual tissues, play a crucial role in the mutual modulation of bone and muscle in CKD. Exercise and nutritional therapy have the potential to yield favorable outcomes. Understanding the underlying mechanisms of bone and muscle loss in CKD can aid in developing new therapies for musculoskeletal diseases, particularly those related to bone loss and muscle wasting.
Collapse
Affiliation(s)
- Kuo-Chin Hung
- Division of Nephrology, Department of Medicine, Min-Sheng General Hospital, Taoyuan City 330, Taiwan
- Department of Pharmacy, Tajen University, Pingtung 907, Taiwan
| | - Wei-Cheng Yao
- Department of Anesthesiology, Min-Sheng General Hospital, Taoyuan City 330, Taiwan
- Department of Medical Education and Clinical Research, Min-Sheng General Hospital, Taoyuan City 330, Taiwan
| | - Yi-Lien Liu
- Department of Family Medicine, Min-Sheng General Hospital, Taoyuan City 330, Taiwan
| | - Hung-Jen Yang
- Department of General Medicine, Min-Sheng General Hospital, Taoyuan City 330, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital Hsinchu Branch, Hsinchu City 300, Taiwan
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan 325, Taiwan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Keong Chong
- Division of Endocrinology and Metabolism, Department of Medicine, Min-Sheng General Hospital, Taoyuan City 330, Taiwan
| | - Ching-Hsiu Peng
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and School of Medicine, Tzu Chi University, Hualien 970, Taiwan
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, School of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
20
|
Salemi S, Schori LJ, Gerwinn T, Horst M, Eberli D. Myostatin Overexpression and Smad Pathway in Detrusor Derived from Pediatric Patients with End-Stage Lower Urinary Tract Dysfunction. Int J Mol Sci 2023; 24:ijms24054462. [PMID: 36901894 PMCID: PMC10003571 DOI: 10.3390/ijms24054462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
Cell therapies and tissue engineering approaches using smooth muscle cells (SMCs) may provide treatment alternatives for end-stage lower urinary tract dysfunction (ESLUTD). Myostatin, a negative regulator of muscle mass, is a promising target to improve muscle function through tissue engineering. The ultimate goal of our project was to investigate the expression of myostatin and its potential impact in SMCs derived from healthy pediatric bladders and pediatric ESLUTD patients. Human bladder tissue samples were evaluated histologically, and SMCs were isolated and characterized. The proliferation of SMCs was assessed by WST-1 assay. The expression pattern of myostatin, its pathway and the contractile phenotype of the cells were investigated at gene and protein levels by real-time PCR, flow cytometry, immunofluorescence, WES and gel contraction assay. Our results show that myostatin is expressed in human bladder smooth muscle tissue and in isolated SMCs at gene and protein levels. A higher expression of myostatin was detected in ESLUTD-derived compared to control SMCs. Histological assessment of bladder tissue confirmed structural changes and decreased muscle-to-collagen ratios in ESLUTD bladders. A decrease in cell proliferation and in the expression of key contractile genes and proteins, α-SMA, calponin, smoothelin and MyH11, as well as a lower degree of in vitro contractility was observed in ESLUTD-derived compared to control SMCs. A reduction in the myostatin-related proteins Smad 2 and follistatin, and an upregulation in the proteins p-Smad 2 and Smad 7 were observed in ESLUTD SMC samples. This is the first demonstration of myostatin expression in bladder tissue and cells. The increased expression of myostatin and the changes in the Smad pathways were observed in ESLUTD patients. Therefore, myostatin inhibitors could be considered for the enhancement of SMCs for tissue engineering applications and as a therapeutic option for patients with ESLUTD and other smooth muscle disorders.
Collapse
Affiliation(s)
- Souzan Salemi
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, 8952 Schlieren, Switzerland
- Correspondence: ; Tel.: +41-795-788-654
| | - Larissa J. Schori
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, 8952 Schlieren, Switzerland
| | - Tim Gerwinn
- Division of Pediatric Urology, University Children’s Hospital Zürich, 8032 Zürich, Switzerland
| | - Maya Horst
- Division of Pediatric Urology, University Children’s Hospital Zürich, 8032 Zürich, Switzerland
| | - Daniel Eberli
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, 8952 Schlieren, Switzerland
| |
Collapse
|
21
|
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
|
22
|
Silva MZC, Vogt BP, Reis NSC, Minicucci FC, Dorna MS, Minicucci MF, Caramori JCT. Serum myostatin levels are associated with physical function and hospitalization in peritoneal dialysis patients. Eur J Clin Nutr 2023; 77:292-294. [PMID: 36329200 DOI: 10.1038/s41430-022-01227-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Myostatin functions as a negative regulator of skeletal muscle growth. The association of myostatin with muscle parameters in dialysis patients is inconsistent, and there are no studies associating myostatin with physical function and outcomes in peritoneal dialysis (PD) patients. Therefore, we assessed the association of serum myostatin with lean mass, physical function, and hospitalization in a prospective cohort of PD patients. METHODS Lean mass, physical function, and serum myostatin were assessed at baseline. Patients were followed up for at least 24 months and hospitalization was recorded. RESULTS Serum myostatin levels were positively correlated with handgrip strength and Appendicular Lean Mass Index among male patients. Binary logistic regression models were performed including myostatin levels and physical function parameters as independent variables. Serum myostatin, handgrip strength, gait speed, and Short Physical Performance Battery were associated with hospitalization. CONCLUSION Lower serum myostatin and physical function were associated with hospitalization in PD patients.
Collapse
Affiliation(s)
| | - Barbara Perez Vogt
- Federal University of Uberlândia (UFU), Graduate Program in Health Sciences, Medicine Faculty, Uberlândia, Brazil
| | - Nayrana Soares Carmo Reis
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Fernanda Chiuso Minicucci
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Mariana Souza Dorna
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | - Marcos Ferreira Minicucci
- Internal Medicine Department, Botucatu Medical School, São Paulo State University, UNESP, Botucatu, Brazil
| | | |
Collapse
|
23
|
Wong L, McMahon LP. Crosstalk between bone and muscle in chronic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1146868. [PMID: 37033253 PMCID: PMC10076741 DOI: 10.3389/fendo.2023.1146868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
With increasing life expectancy, the related disorders of bone loss, metabolic dysregulation and sarcopenia have become major health threats to the elderly. Each of these conditions is prevalent in patients with chronic kidney disease (CKD), particularly in more advanced stages. Our current understanding of the bone-muscle interaction is beyond mechanical coupling, where bone and muscle have been identified as interrelated secretory organs, and regulation of both bone and muscle metabolism occurs through osteokines and myokines via autocrine, paracrine and endocrine systems. This review appraises the current knowledge regarding biochemical crosstalk between bone and muscle, and considers recent progress related to the role of osteokines and myokines in CKD, including modulatory effects of physical exercise and potential therapeutic targets to improve musculoskeletal health in CKD patients.
Collapse
Affiliation(s)
- Limy Wong
- Department of Renal Medicine, Monash University Eastern Health Clinical School, Box Hill, VIC, Australia
- Department of Renal Medicine, Eastern Health, Box Hill, VIC, Australia
- *Correspondence: Limy Wong,
| | - Lawrence P. McMahon
- Department of Renal Medicine, Monash University Eastern Health Clinical School, Box Hill, VIC, Australia
- Department of Renal Medicine, Eastern Health, Box Hill, VIC, Australia
| |
Collapse
|
24
|
Troutman AD, Arroyo E, Lim K, Moorthi RN, Avin KG. Skeletal Muscle Complications in Chronic Kidney Disease. Curr Osteoporos Rep 2022; 20:410-421. [PMID: 36149594 PMCID: PMC10064704 DOI: 10.1007/s11914-022-00751-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 01/30/2023]
Abstract
PURPOSE OF REVIEW To provide an overview of the recent literature investigating the pathophysiology of skeletal muscle changes, interventions for skeletal muscle, and effects of exercise in chronic kidney disease (CKD). RECENT FINDINGS There are multiple CKD-related changes that negatively impact muscle size and function. However, the variability in the assessment of muscle size, in particular, hinders the ability to truly understand the impact it may have in CKD. Exercise interventions to improve muscle size and function demonstrate inconsistent responses that warrant further investigation to optimize exercise prescription. Despite progress in the field, there are many gaps in the knowledge of the pathophysiology of sarcopenia of CKD. Identifying these gaps will help in the design of interventions that can be tested to target muscle loss and its consequences such as impaired mobility, falls, and poor quality of life in patients with CKD.
Collapse
Affiliation(s)
- Ashley D Troutman
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, CF-326, 1140 W. Michigan St., Indianapolis, IN, 46202, USA
| | - Eliott Arroyo
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kenneth Lim
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ranjani N Moorthi
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Keith G Avin
- Department of Physical Therapy, School of Health and Human Sciences, Indiana University Purdue University, CF-326, 1140 W. Michigan St., Indianapolis, IN, 46202, USA.
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
25
|
Gaillard F, Ould Rabah M, Garcelon N, Touam M, Neuraz A, Legendre C, Anglicheau D, Prié D, Bienaimé F. Allograft function and muscle mass evolution after kidney transplantation. J Cachexia Sarcopenia Muscle 2022; 13:2875-2887. [PMID: 36106518 PMCID: PMC9745471 DOI: 10.1002/jcsm.13066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Advanced chronic kidney disease is associated with muscle wasting, but how glomerular filtration rate (GFR) recovery after kidney transplantation is associated with muscle mass is unknown. METHODS We took advantage of the simultaneous measurement of GFR (using iohexol plasma clearance; ioGFR) and creatinine excretion rate (a surrogate marker of muscle mass; CER) performed 3 months after transplantation and at a later time point at our institution to investigate the interplay between allograft function, muscle mass, and outcome in kidney transplant recipients. RESULTS Between June 2005 and October 2019, 1319 successive kidney transplant recipients (mean age 50.4 ± 14.6; 38.7% female) underwent GFR measurement at our institution 3 months after kidney transplantation. CER (CER3 ) and ioGFR (ioGFR3 ) were 7.7 ± 2.6 μmol/min and 53 ± 17.1 mL/min/1.73 m2 , respectively. Multivariable analysis identified female gender, older donor and recipient age, reduced body mass index, coronary disease, dialysis history, proteinuria, and reduced ioGFR3 as independent predictors of low CER3 (ioGFR3 : β coefficient 0.19 [95% confidence interval 0.14 to 0.24]). A total of 1165 patients had a subsequent CER measurement after a median follow-up of 9.5 months. Of them, 373 (32%) experienced an increase in CER > 10%, while 222 (19%) showed a CER decrease of more than 10%. Multivariable analysis adjusted for CER3 and other confounders identified ioGFR3 as an independent predictor of CER at follow-up (β coefficient 0.11 [95% confidence interval 0.07 to 0.16]). In multivariable Cox analysis, reduced CER at 3 months or at follow-up were consistently associated with mortality (hazard ratio [95% confidence interval] at 3 months: 0.82 [0.74 to 0.91]; at follow-up: 0.79 [0.69 to 0.99]) but not with graft loss. CONCLUSIONS Glomerular filtration rate recovery is a determinant of muscle mass variation after kidney transplantation. Early interventions targeting muscle mass gain may be beneficial for kidney transplant recipients.
Collapse
Affiliation(s)
- François Gaillard
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot et faculté de médecine, Université Lyon 1, Lyon, France
| | - Mélissa Ould Rabah
- Service de Physiologie, Hôpital Necker-Enfants Malades, Assistance-Publique-Hôpitaux de Paris, Paris, France.,Faculté de médecine, Université de Paris-Cité, Paris, France
| | - Nicolas Garcelon
- Université de Paris-Cité, Imagine Institute, Data Science Platform, INSERM UMR 1163, Paris, France
| | - Malik Touam
- Service de Néphrologie et Transplantation, Hôpital Necker-Enfants Malades, Assistance-Publique-Hôpitaux de Paris, Paris, France
| | - Antoine Neuraz
- Service d'Informatique Médical, Hôpital Necker-Enfants Malades, Assistance-Publique-Hôpitaux de Paris, Paris, France
| | - Christophe Legendre
- Faculté de médecine, Université de Paris-Cité, Paris, France.,Service de Néphrologie et Transplantation, Hôpital Necker-Enfants Malades, Assistance-Publique-Hôpitaux de Paris, Paris, France.,Institut Necker-Enfants Malades, INSERM U1151, Paris, France
| | - Dany Anglicheau
- Faculté de médecine, Université de Paris-Cité, Paris, France.,Service de Néphrologie et Transplantation, Hôpital Necker-Enfants Malades, Assistance-Publique-Hôpitaux de Paris, Paris, France.,Institut Necker-Enfants Malades, INSERM U1151, Paris, France
| | - Dominique Prié
- Service de Physiologie, Hôpital Necker-Enfants Malades, Assistance-Publique-Hôpitaux de Paris, Paris, France.,Faculté de médecine, Université de Paris-Cité, Paris, France.,Institut Necker-Enfants Malades, INSERM U1151, Paris, France
| | - Frank Bienaimé
- Service de Physiologie, Hôpital Necker-Enfants Malades, Assistance-Publique-Hôpitaux de Paris, Paris, France.,Faculté de médecine, Université de Paris-Cité, Paris, France.,Institut Necker-Enfants Malades, INSERM U1151, Paris, France
| |
Collapse
|
26
|
Sarcopenia and cardiovascular disease in patients with and without kidney disease: what do we know? Int Urol Nephrol 2022; 55:1161-1171. [PMID: 36327007 DOI: 10.1007/s11255-022-03393-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 10/15/2022] [Indexed: 11/06/2022]
Abstract
Cardiovascular disease (CVD) incidence is high in patients with chronic kidney disease (CKD) and is the most frequent cause of mortality in this population. Advanced age, hypertension, uremic toxins, endothelial dysfunction, atherosclerosis, hyperhomocysteinemia, oxidative stress, and inflammation are among the leading causes of increased CVD in advanced stages of CKD. Although defined as a decrease in muscle strength associated with aging, sarcopenia is also prevalent in CKD patients. Sarcopenia causes physical disability, low quality of life, and mortality. Regular exercise and nutritional supplementation may slow the progression of sarcopenia. Recent studies have shown that sarcopenia increases the risk of CVD and mortality in people with or without kidney disease. This review discusses the relationship between sarcopenia and CVD in light of the current literature.
Collapse
|
27
|
Chen Y, Banie L, Breyer BN, Tan Y, Wang Z, Zhou F, Wang G, Lin G, Liu J, Qi LS, Lue TF. Enhanced Myogenesis by Silencing Myostatin with Nonviral Delivery of dCas9 Ribonucleoprotein Complex. CRISPR J 2022; 5:598-608. [PMID: 35758824 DOI: 10.1089/crispr.2022.0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Stress urinary incontinence (SUI) and pelvic floor disorder (PFD) are common conditions with limited treatment options in women worldwide. Regenerative therapy to restore urethral striated and pelvic floor muscles represents a valuable therapeutic approach. We aim to determine the CRISPR interference-mediated gene silencing effect of the nonviral delivery of nuclease-deactivated dCas9 ribonucleoprotein (RNP) complex on muscle regeneration at the cellular and molecular level. We designed four myostatin (MSTN)-targeting sgRNAs and transfected them into rat myoblast L6 cells together with the dCas9 protein. Myogenesis assay and immunofluorescence staining were performed to evaluate muscle differentiation, while CCK8 assay, cell cycle assay, and 5-ethynyl-2'-deoxyuridine staining were used to measure muscle proliferation. Reverse transcription-polymerase chain reaction and Western blotting were also performed to examine cellular signaling. Myogenic factors (including myosin heavy chain, MSTN, myocardin, and serum response factor) increased significantly after day 5 during myogenesis. MSTN was efficiently silenced after transfecting the dCas9 RNP complex, which significantly promoted more myotube formation and a higher fusion index for L6 cells. In cellular signaling, MSTN repression enhanced the expression of MyoG and MyoD, phosphorylation of Smad2, and the activity of Wnt1/GSK-3β/β-catenin pathway. Moreover, MSTN repression accelerated L6 cell growth with a higher cell proliferation index as well as a higher expression of cyclin D1 and cyclin E. Nonviral delivery of the dCas9 RNP complex significantly promoted myoblast differentiation and proliferation, providing a promising approach to improve muscle regeneration for SUI and PFD. Further characterization and validation of this approach in vivo are needed.
Collapse
Affiliation(s)
- Yinwei Chen
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, San Francisco, California, USA.,Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Benjamin N Breyer
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Yan Tan
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Zhao Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Feng Zhou
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Guifang Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jihong Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei S Qi
- Department of Bioengineering, Stanford University, Stanford, California, USA.,ChEM-H, Stanford University, Stanford, California, USA
| | - Tom F Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
28
|
Bellafronte NT, Govêia TR, Chiarello PG. Sarcopenia in chronic kidney disease: prevalence by different definitions and relationship with adiposity. Appl Physiol Nutr Metab 2022; 47:915-925. [PMID: 35658617 DOI: 10.1139/apnm-2021-0521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This was a cross-sectional study with Chronic Kidney Disease (CKD) patients under non-dialysis-dependent (NDD), hemodialysis (HD) and kidney transplant (KTx) treatment aimed to evaluate the prevalence of sarcopenia using the European Working Group on Sarcopenia in Older People (EWGSOP2) and the Foundation of the National Institutes of Health (FNIH) guidelines; also analyze the relationship between sarcopenia and its components with body adiposity. Body composition was assessed by dual energy X-ray absorptiometry (DXA) and anthropometry. Bioelectrical impedance provided data of phase angle and body water. The prevalence of sarcopenia in total sample (n=243; 53% men, 48±10 y) was 7% by FNIH and 5% by EWGSOP2 criteria; and was low in each CKD group independently of the criteria applied (max 11% prevalence). Low muscle mass was present in 39% (FNIH) and 36% (EWGSOP2) and dynapenia in 10% of patients. Sarcopenic patients by EWGSOP2 criteria presented low body adiposity. Conversely, sarcopenic patients by FNIH presented high adiposity. This study suggests that in CKD (i) sarcopenia and low muscle mass prevalence varies according to the diagnostic criteria, (ii) are common conditions, (iii) association with body adiposity depends on the criteria used to define low muscle mass, (iv) FNIH criteria detected high adiposity in individuals with sarcopenia. Novelty bullets: Prevalence of sarcopenia and low muscle mass in CKD varied according to the diagnostic criteria; association of excess adiposity with sarcopenia and low muscle mass depends on muscle mass index applied; FNIH criteria detected higher adiposity in individuals with sarcopenia. NOVELTY BULLETS Prevalence of sarcopenia and low muscle mass in CKD varied according to the diagnostic criteria; Association of excess adiposity with sarcopenia and low muscle mass depends on muscle mass index applied; FNIH criteria detected higher adiposity in individuals with sarcopenia and low muscle mass.
Collapse
Affiliation(s)
| | - Thaísa Ribeiro Govêia
- University of São Paulo, Nutrition and Metabolism Undergraduate Course, Ribeirão Preto City, São Paulo State, Brazil;
| | - Paula Garcia Chiarello
- University of São Paulo, Department of Health Sciences, Ribeirão Preto City, São Paulo State, Brazil;
| |
Collapse
|
29
|
Shin JY. Low serum creatinine to cystatin C ratio is independently associated with sarcopenia and high carotid plaque score in patients with type 2 diabetes. Nutr Metab Cardiovasc Dis 2022; 32:1454-1462. [PMID: 35256230 DOI: 10.1016/j.numecd.2022.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/24/2022] [Accepted: 02/07/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND AIMS Low serum creatinine (Cr) to cystatin C (cysC) ratio has been suggested to be associated with low muscle mass and strength and poor prognosis in various chronic disease. We investigated the associations of CCR with sarcopenia and carotid plaque score (PS) in patients with type 2 diabetes mellitus. METHODS AND RESULTS A total of 1577 patients with type 2 diabetes were enrolled. High PS was defined as PS ≥ 3. Sarcopenia was assessed by the measurement of appendicular skeletal muscle mass (ASM) and grip strength (GS). Compared to the highest CCR group, the lowest tertile group was older; had higher C-reactive protein levels, CIMT, and PS, but lower cysC-based estimated glomerular filtration rate (cysC-eGFR), ASM/BMI, and GS. Positive correlations between CCR and ASM/BMI (r = 0.239 in men and 0.303 in women, p < 0.001) and GS (r = 0.282 in men and 0.270 in women, p < 0.001) were observed in both genders. Odds ratios and 95% confidence intervals for high PS after adjusting for age and sex were 1.22 (0.92-1.61, p = 0.18) in the middle and 1.74 (1.31-2.30, p < 0.001) in the lowest tertiles, respectively, with those of the lowest tertile remaining significant after further adjusting for multiple confounders. CONCLUSIONS Low CCR was independently associated with sarcopenia and high PS in patients with type 2 diabetes mellitus, especially after adjusting for ASM/BMI and GS.
Collapse
Affiliation(s)
- Jang Yel Shin
- Department of Internal Medicine, Yonsei University, Wonju College of Medicine, Wonju, Republic of Korea.
| |
Collapse
|
30
|
Muscle Wasting in Chronic Kidney Disease: Mechanism and Clinical Implications—A Narrative Review. Int J Mol Sci 2022; 23:ijms23116047. [PMID: 35682722 PMCID: PMC9181340 DOI: 10.3390/ijms23116047] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/21/2022] [Accepted: 05/26/2022] [Indexed: 12/15/2022] Open
Abstract
Muscle wasting, known to develop in patients with chronic kidney disease (CKD), is a deleterious consequence of numerous complications associated with deteriorated renal function. Muscle wasting in CKD mainly involves dysregulated muscle protein metabolism and impaired muscle cell regeneration. In this narrative review, we discuss the cardinal role of the insulin-like growth factor 1 and myostatin signaling pathways, which have been extensively investigated using animal and human studies, as well as the emerging concepts in microRNA- and gut microbiota-mediated regulation of muscle mass and myogenesis. To ameliorate muscle loss, therapeutic strategies, including nutritional support, exercise programs, pharmacological interventions, and physical modalities, are being increasingly developed based on advances in understanding its underlying pathophysiology.
Collapse
|
31
|
Li Q, Wu J, Huang J, Hu R, You H, Liu L, Wang D, Wei L. Paeoniflorin Ameliorates Skeletal Muscle Atrophy in Chronic Kidney Disease via AMPK/SIRT1/PGC-1α-Mediated Oxidative Stress and Mitochondrial Dysfunction. Front Pharmacol 2022; 13:859723. [PMID: 35370668 PMCID: PMC8964350 DOI: 10.3389/fphar.2022.859723] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
Skeletal muscle atrophy is a common and serious complication of chronic kidney disease (CKD). Oxidative stress and mitochondrial dysfunction are involved in the pathogenesis of muscle atrophy. The aim of this study was to explore the effects and mechanisms of paeoniflorin on CKD skeletal muscle atrophy. We demonstrated that paeoniflorin significantly improved renal function, calcium/phosphorus disorders, nutrition index and skeletal muscle atrophy in the 5/6 nephrectomized model rats. Paeoniflorin ameliorated the expression of proteins associated with muscle atrophy and muscle differentiation, including muscle atrophy F-box (MAFbx/atrogin-1), muscle RING finger 1 (MuRF1), MyoD and myogenin (MyoG). In addition, paeoniflorin modulated redox homeostasis by increasing antioxidant activity and suppressing excessive accumulation of reactive oxygen species (ROS). Paeoniflorin alleviated mitochondrial dysfunction by increasing the activities of electron transport chain complexes and mitochondrial membrane potential. Furthermore, paeoniflorin also regulates mitochondrial dynamics. Importantly, paeoniflorin upregulated the expression of silent information regulator 1 (SIRT1), peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), and phosphorylation of AMP-activated protein kinase (AMPK). Similar results were observed in C2C12 myoblasts treated with TNF-α and paeoniflorin. Notably, these beneficial effects of paeoniflorin on muscle atrophy were abolished by inhibiting AMPK and SIRT1 and knocking down PGC-1α. Taken together, this study showed for the first time that paeoniflorin has great therapeutic potential for CKD skeletal muscle atrophy through AMPK/SIRT1/PGC-1α-mediated oxidative stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Qiang Li
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, China.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jing Wu
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiawen Huang
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, China.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Rong Hu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Haiyan You
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lingyu Liu
- First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Dongtao Wang
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Lianbo Wei
- Department of Traditional Chinese Medicine, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| |
Collapse
|
32
|
Alizadeh Pahlavani H. Exercise Therapy for People With Sarcopenic Obesity: Myokines and Adipokines as Effective Actors. Front Endocrinol (Lausanne) 2022; 13:811751. [PMID: 35250869 PMCID: PMC8892203 DOI: 10.3389/fendo.2022.811751] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
Sarcopenic obesity is defined as a multifactorial disease in aging with decreased body muscle, decreased muscle strength, decreased independence, increased fat mass, due to decreased physical activity, changes in adipokines and myokines, and decreased satellite cells. People with sarcopenic obesity cause harmful changes in myokines and adipokines. These changes are due to a decrease interleukin-10 (IL-10), interleukin-15 (IL-15), insulin-like growth factor hormone (IGF-1), irisin, leukemia inhibitory factor (LIF), fibroblast growth factor-21 (FGF-21), adiponectin, and apelin. While factors such as myostatin, leptin, interleukin-6 (IL-6), interleukin-8 (IL-8), and resistin increase. The consequences of these changes are an increase in inflammatory factors, increased degradation of muscle proteins, increased fat mass, and decreased muscle tissue, which exacerbates sarcopenia obesity. In contrast, exercise, especially strength training, reverses this process, which includes increasing muscle protein synthesis, increasing myogenesis, increasing mitochondrial biogenesis, increasing brown fat, reducing white fat, reducing inflammatory factors, and reducing muscle atrophy. Since some people with chronic diseases are not able to do high-intensity strength training, exercises with blood flow restriction (BFR) are newly recommended. Numerous studies have shown that low-intensity BFR training produces the same increase in hypertrophy and muscle strength such as high-intensity strength training. Therefore, it seems that exercise interventions with BFR can be an effective way to prevent the exacerbation of sarcopenia obesity. However, due to limited studies on adipokines and exercises with BFR in people with sarcopenic obesity, more research is needed.
Collapse
|
33
|
The Correlation of Serum Myostatin Levels with Gait Speed in Kidney Transplantation Recipients. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19010465. [PMID: 35010726 PMCID: PMC8744722 DOI: 10.3390/ijerph19010465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 12/10/2022]
Abstract
The primary role of myostatin is to negatively regulate skeletal muscle growth. The gait speed is a noninvasive, reliable parameter that predicts cardiovascular risk and mortality. This study evaluated the relationship between serum myostatin concentrations and gait speeds in patients who had undergone kidney transplantation (KT). A total of 84 KT recipients were evaluated. A speed of less than 1.0 m/s was categorized into the low gait speed group. We measured serum myostatin concentrations with a commercial enzyme-linked immunosorbent assay. KT recipients in the low gait speed group had significantly older age, as well as higher body weight, body mass index (BMI), skeletal muscle index, serum triglyceride levels, glucose levels, and blood urea nitrogen levels, lower estimated glomerular filtration rates and serum myostatin levels, a higher percentage of steroid use, and a lower proportion of mycophenolate mofetil use. Multivariable logistic regression analysis revealed that lower myostatin levels and lower frequency of mycophenolate mofetil use were independently associated with low gait speed. In multivariable stepwise linear regression analysis, myostatin levels were positively correlated with gait speeds, and age and BMI were negatively correlated with gait speeds. In the study, serum myostatin levels were significantly lower in the low gait speed group. Subjects in the low gait speed group also had greater BMI and older age.
Collapse
|
34
|
Gungor O, Ulu S, Hasbal NB, Anker SD, Kalantar‐Zadeh K. Effects of hormonal changes on sarcopenia in chronic kidney disease: where are we now and what can we do? J Cachexia Sarcopenia Muscle 2021; 12:1380-1392. [PMID: 34676694 PMCID: PMC8718043 DOI: 10.1002/jcsm.12839] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/02/2021] [Accepted: 09/19/2021] [Indexed: 01/06/2023] Open
Abstract
Sarcopenia or muscle wasting is a progressive and generalized skeletal muscle disorder involving the accelerated loss of muscle mass and function, often associated with muscle weakness (dynapenia) and frailty. Whereas primary sarcopenia is related to ageing, secondary sarcopenia happens independent of age in the context of chronic disease states such as chronic kidney disease (CKD). Sarcopenia has become a major focus of research and public policy debate due to its impact on patient's health-related quality of life, health-care expenditure, morbidity, and mortality. The development of sarcopenia in patients with CKD is multifactorial and it may occur independently of weight loss or cachexia including under obese sarcopenia. Hormonal imbalances can facilitate the development of sarcopenia in the general population and is a common finding in CKD. Hormones that may influence the development of sarcopenia are testosterone, growth hormone, insulin, thyroid hormones, and vitamin D. Although the relationship between free testosterone level that is low in uraemic patients and sarcopenia in CKD is not well-defined, functional improvement may be seen. Unlike testosterone, it is known that vitamin D is associated with muscle strength, muscle size, and physical performance in patients with CKD. Outcomes after vitamin D replacement therapy are still controversial. The half-life of growth hormone (GH) is prolonged in patients with CKD. Besides, IGF-1 levels are normal in patients with Stage 4 CKD-a minimal reduction is seen in the end-stage renal disease. Unresponsiveness or resistance of IGF-1 and changes in the GH/IGF-1 axis are the main causes of sarcopenia in CKD. Low serum T3 level is frequent in CKD, but the net effect on sarcopenia is not well-studied. CKD patients develop insulin resistance (IR) from the earliest period even before GFR decline begins. IR reduces glucose utilization as an energy source by hepatic gluconeogenesis, decreasing muscle glucose uptake, impairing intracellular glucose metabolism. This cascade results in muscle protein breakdown. IR and sarcopenia might also be a new pathway for targeting. Ghrelin, oestrogen, cortisol, and dehydroepiandrosterone may be other players in the setting of sarcopenia. In this review, we mainly examine the effects of hormonal changes on the occurrence of sarcopenia in patients with CKD via the available data.
Collapse
Affiliation(s)
- Ozkan Gungor
- Division of Nephrology, Department of Internal Medicine, Faculty of MedicineKahramanmaras Sutcu Imam UniversityKahramanmarasTurkey
| | - Sena Ulu
- Department of Internal Medicine and Nephrology, Faculty of MedicineBahcesehir UniversityIstanbulTurkey
| | - Nuri Baris Hasbal
- Clinic of NephrologyBasaksehir Cam and Sakura City HospitalIstanbulTurkey
| | - Stefan D. Anker
- Department of Cardiology (CVK), Berlin Institute of Health Center for Regenerative Therapies (BCRT)German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin BerlinBerlinGermany
| | - Kamyar Kalantar‐Zadeh
- Division of Nephrology, Hypertension and Kidney TransplantationUniversity of California Irvine School of MedicineOrangeCAUSA
| |
Collapse
|
35
|
Molecular and Metabolic Mechanism of Low-Intensity Pulsed Ultrasound Improving Muscle Atrophy in Hindlimb Unloading Rats. Int J Mol Sci 2021; 22:ijms222212112. [PMID: 34829990 PMCID: PMC8625684 DOI: 10.3390/ijms222212112] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/31/2021] [Accepted: 11/02/2021] [Indexed: 12/16/2022] Open
Abstract
Low-intensity pulsed ultrasound (LIPUS) has been proved to promote the proliferation of myoblast C2C12. However, whether LIPUS can effectively prevent muscle atrophy has not been clarified, and if so, what is the possible mechanism. The aim of this study is to evaluate the effects of LIPUS on muscle atrophy in hindlimb unloading rats, and explore the mechanisms. The rats were randomly divided into four groups: normal control group (NC), hindlimb unloading group (UL), hindlimb unloading plus 30 mW/cm2 LIPUS irradiation group (UL + 30 mW/cm2), hindlimb unloading plus 80 mW/cm2 LIPUS irradiation group (UL + 80 mW/cm2). The tails of rats in hindlimb unloading group were suspended for 28 days. The rats in the LIPUS treated group were simultaneously irradiated with LIPUS on gastrocnemius muscle in both lower legs at the sound intensity of 30 mW/cm2 or 80 mW/cm2 for 20 min/d for 28 days. C2C12 cells were exposed to LIPUS at 30 or 80 mW/cm2 for 5 days. The results showed that LIPUS significantly promoted the proliferation and differentiation of myoblast C2C12, and prevented the decrease of cross-sectional area of muscle fiber and gastrocnemius mass in hindlimb unloading rats. LIPUS also significantly down regulated the expression of MSTN and its receptors ActRIIB, and up-regulated the expression of Akt and mTOR in gastrocnemius muscle of hindlimb unloading rats. In addition, three metabolic pathways (phenylalanine, tyrosine and tryptophan biosynthesis; alanine, aspartate and glutamate metabolism; glycine, serine and threonine metabolism) were selected as important metabolic pathways for hindlimb unloading effect. However, LIPUS promoted the stability of alanine, aspartate and glutamate metabolism pathway. These results suggest that the key mechanism of LIPUS in preventing muscle atrophy induced by hindlimb unloading may be related to promoting protein synthesis through MSTN/Akt/mTOR signaling pathway and stabilizing alanine, aspartate and glutamate metabolism.
Collapse
|
36
|
Bellafronte NT, Vega-Piris L, Cuadrado GB, Chiarello PG. Performance of Bioelectrical Impedance and Anthropometric Predictive Equations for Estimation of Muscle Mass in Chronic Kidney Disease Patients. Front Nutr 2021; 8:683393. [PMID: 34095195 PMCID: PMC8177428 DOI: 10.3389/fnut.2021.683393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 04/26/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Patients with chronic kidney disease (CKD) are vulnerable to loss of muscle mass due to several metabolic alterations derived from the uremic syndrome. Reference methods for body composition evaluation are usually unfeasible in clinical settings. Aims: To evaluate the accuracy of predictive equations based on bioelectrical impedance analyses (BIA) and anthropometry parameters for estimating fat free mass (FFM) and appendicular FFM (AFFM), compared to dual energy X-ray absorptiometry (DXA), in CKD patients. Methods: We performed a longitudinal study with patients in non-dialysis-dependent, hemodialysis, peritoneal dialysis and kidney transplant treatment. FFM and AFFM were evaluated by DXA, BIA (Sergi, Kyle, Janssen and MacDonald equations) and anthropometry (Hume, Lee, Tian, and Noori equations). Low muscle mass was diagnosed by DXA analysis. Intra-class correlation coefficient (ICC), Bland-Altman graphic and multiple regression analysis were used to evaluate equation accuracy, linear regression analysis to evaluate bias, and ROC curve analysis and kappa for reproducibility. Results: In total sample and in each CKD group, the predictive equation with the best accuracy was AFFMSergi (men, n = 137: ICC = 0.91, 95% CI = 0.79–0.96, bias = 1.11 kg; women, n = 129: ICC = 0.94, 95% CI = 0.92–0.96, bias = −0.28 kg). AFFMSergi also presented the best performance for low muscle mass diagnosis (men, kappa = 0.68, AUC = 0.83; women, kappa = 0.65, AUC = 0.85). Bias between AFFMSergi and AFFMDXA was mainly affected by total body water and fat mass. None of the predictive equations was able to accurately predict changes in AFFM and FFM, with all ICC lower than 0.5. Conclusion: The predictive equation with the best performance to asses muscle mass in CKD patients was AFFMSergi, including evaluation of low muscle mass diagnosis. However, assessment of changes in body composition was biased, mainly due to variations in fluid status together with adiposity, limiting its applicability for longitudinal evaluations.
Collapse
Affiliation(s)
- Natália Tomborelli Bellafronte
- Post-graduate Program in Health Sciences, Ribeirão Preto Faculty of Medicine, University of São Paulo, Ribeirão Preto, Brazil
| | - Lorena Vega-Piris
- Methodology Unit, Instituto de Investigación Sanitaria del Hospital Universitario de la Princesa, Madrid, Spain
| | | | - Paula Garcia Chiarello
- Department of Health Sciences, Ribeirão Preto Faculty of Medicine, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
37
|
Maintenance of Skeletal Muscle to Counteract Sarcopenia in Patients with Advanced Chronic Kidney Disease and Especially Those Undergoing Hemodialysis. Nutrients 2021; 13:nu13051538. [PMID: 34063269 PMCID: PMC8147474 DOI: 10.3390/nu13051538] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 12/18/2022] Open
Abstract
Life extension in modern society has introduced new concepts regarding such disorders as frailty and sarcopenia, which has been recognized in various studies. At the same time, cutting-edge technology methods, e.g., renal replacement therapy for conditions such as hemodialysis (HD), have made it possible to protect patients from advanced lethal chronic kidney disease (CKD). Loss of muscle and fat mass, termed protein energy wasting (PEW), has been recognized as prognostic factor and, along with the increasing rate of HD introduction in elderly individuals in Japan, appropriate countermeasures are necessary. Although their origins differ, frailty, sarcopenia, and PEW share common components, among which skeletal muscle plays a central role in their etiologies. The nearest concept may be sarcopenia, for which diagnosis techniques have recently been reported. The focus of this review is on maintenance of skeletal muscle against aging and CKD/HD, based on muscle physiology and pathology. Clinically relevant and topical factors related to muscle wasting including sarcopenia, such as vitamin D, myostatin, insulin (related to diabetes), insulin-like growth factor I, mitochondria, and physical inactivity, are discussed. Findings presented thus far indicate that in addition to modulation of the aforementioned factors, exercise combined with nutritional supplementation may be a useful approach to overcome muscle wasting and sarcopenia in elderly patients undergoing HD treatments.
Collapse
|
38
|
Leal DV, Ferreira A, Watson EL, Wilund KR, Viana JL. Muscle-Bone Crosstalk in Chronic Kidney Disease: The Potential Modulatory Effects of Exercise. Calcif Tissue Int 2021; 108:461-475. [PMID: 33388899 DOI: 10.1007/s00223-020-00782-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) is a prevalent worldwide public burden that increasingly compromises overall health as the disease progresses. Two of the most negatively affected tissues are bone and skeletal muscle, with CKD negatively impacting their structure, function and activity, impairing the quality of life of these patients and contributing to morbidity and mortality. Whereas skeletal health in this population has conventionally been associated with bone and mineral disorders, sarcopenia has been observed to impact skeletal muscle health in CKD. Indeed, bone and muscle tissues are linked anatomically and physiologically, and together regulate functional and metabolic mechanisms. With the initial crosstalk between the skeleton and muscle proposed to explain bone formation through muscle contraction, it is now understood that this communication occurs through the interaction of myokines and osteokines, with the skeletal muscle secretome playing a pivotal role in the regulation of bone activity. Regular exercise has been reported to be beneficial to overall health. Also, the positive regulatory effect that exercise has been proposed to have on bone and muscle anatomical, functional, and metabolic activity has led to the proposal of regular physical exercise as a therapeutic strategy for muscle and bone-related disorders. The detection of bone- and muscle-derived cytokine secretion following physical exercise has strengthened the idea of a cross communication between these organs. Hence, this review presents an overview of the impact of CKD in bone and skeletal muscle, and narrates how these tissues intrinsically communicate with each other, with focus on the potential effect of exercise in the modulation of this intercommunication.
Collapse
Affiliation(s)
- Diogo V Leal
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University Institute of Maia, ISMAI, Maia, Portugal
| | - Aníbal Ferreira
- Department of Nephrology, Curry Cabral Hospital, Hospital Centre of Central Lisbon, Lisbon, Portugal
- Nova Medical School, Lisbon, Portugal
| | - Emma L Watson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Kenneth R Wilund
- Department of Kinesiology and Community Health, University of Illinois At Urbana-Champaign, Champaign, IL, USA
| | - João L Viana
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University Institute of Maia, ISMAI, Maia, Portugal.
| |
Collapse
|
39
|
Filgueira TO, Castoldi A, Santos LER, de Amorim GJ, de Sousa Fernandes MS, Anastácio WDLDN, Campos EZ, Santos TM, Souto FO. The Relevance of a Physical Active Lifestyle and Physical Fitness on Immune Defense: Mitigating Disease Burden, With Focus on COVID-19 Consequences. Front Immunol 2021; 12:587146. [PMID: 33613573 PMCID: PMC7892446 DOI: 10.3389/fimmu.2021.587146] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/13/2021] [Indexed: 12/15/2022] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a fast spreading virus leading to the development of Coronavirus Disease-2019 (COVID-19). Severe and critical cases are characterized by damage to the respiratory system, endothelial inflammation, and multiple organ failure triggered by an excessive production of proinflammatory cytokines, culminating in the high number of deaths all over the world. Sedentarism induces worse, continuous, and progressive consequences to health. On the other hand, physical activity provides benefits to health and improves low-grade systemic inflammation. The aim of this review is to elucidate the effects of physical activity in physical fitness, immune defense, and its contribution to mitigate the severe inflammatory response mediated by SARS-CoV-2. Physical exercise is an effective therapeutic strategy to mitigate the consequences of SARS-CoV-2 infection. In this sense, studies have shown that acute physical exercise induces the production of myokines that are secreted in tissues and into the bloodstream, supporting its systemic modulatory effect. Therefore, maintaining physical activity influence balance the immune system and increases immune vigilance, and also might promote potent effects against the consequences of infectious diseases and chronic diseases associated with the development of severe forms of COVID-19. Protocols to maintain exercise practice are suggested and have been strongly established, such as home-based exercise (HBE) and outdoor-based exercise (OBE). In this regard, HBE might help to reduce levels of physical inactivity, bed rest, and sitting time, impacting on adherence to physical activity, promoting all the benefits related to exercise, and attracting patients in different stages of treatment for COVID-19. In parallel, OBE must improve health, but also prevent and mitigate COVID-19 severe outcomes in all populations. In conclusion, HBE or OBE models can be a potent strategy to mitigate the progress of infection, and a coadjutant therapy for COVID-19 at all ages and different chronic conditions.
Collapse
Affiliation(s)
| | - Angela Castoldi
- Keizo Asami Immunopathology Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
| | - Lucas Eduardo R. Santos
- Pós Graduação em Educação Física, Universidade Federal de Pernambuco, Recife, Brazil
- Pós Graduação em Neuropsiquiatria e Ciências do Comportamento, Universidade Federal de Pernambuco, Recife, Brazil
| | - Geraldo José de Amorim
- Keizo Asami Immunopathology Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
- Serviço de Nefrologia do Hospital das Clínicas, Universidade Federal de Pernambuco, Recife, Brazil
| | - Matheus Santos de Sousa Fernandes
- Pós Graduação em Educação Física, Universidade Federal de Pernambuco, Recife, Brazil
- Pós Graduação em Neuropsiquiatria e Ciências do Comportamento, Universidade Federal de Pernambuco, Recife, Brazil
| | | | | | - Tony Meireles Santos
- Pós Graduação em Educação Física, Universidade Federal de Pernambuco, Recife, Brazil
| | - Fabrício Oliveira Souto
- Keizo Asami Immunopathology Laboratory, Universidade Federal de Pernambuco, Recife, Brazil
- Núcleo de Ciências da Vida, Centro Acadêmico do Agreste, Universidade Federal de Pernambuco, Caruaru, Brazil
| |
Collapse
|