1
|
Tsai CC, Wang PC, Hsiung T, Fan YH, Wu JT, Kan WC, Shiao CC. Sarcopenia in Chronic Kidney Disease: A Narrative Review from Pathophysiology to Therapeutic Approaches. Biomedicines 2025; 13:352. [PMID: 40002765 PMCID: PMC11852367 DOI: 10.3390/biomedicines13020352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Chronic kidney disease (CKD) is a progressive condition linked to sarcopenia, a syndrome characterized by loss of skeletal muscle mass and strength, affecting a quarter of CKD patients globally. Sarcopenia has multiple paths through which it can worsen morbidity and mortality as well as decrease the quality of life in CKD, including systemic inflammation, hormonal imbalances, metabolic changes, and dysbiosis of gut microbiota. There is a regional variation in the criteria set for diagnosis, with two main groups being the European Working Group on Sarcopenia in Older People and the Asian Working Group for Sarcopenia. Management regimes such as nutritional optimization, vitamin D, exercise, correction of metabolic acidosis, and modulation of gut microbiota constitute effective intervention strategies. Emerging therapeutic options include anabolic agents, myostatin inhibitors, and anti-inflammatory treatment options. Future advances such as genomics, proteomics, and personalized medicine will open up new avenues for addressing the complex pathophysiology of sarcopenia. Hence, a comprehensive multidisciplinary approach focused on the specific needs of each patient will be vital in reducing the effects of sarcopenia and improving the situation of people with CKD.
Collapse
Affiliation(s)
- Chung-Ching Tsai
- Division of Orthopaedics, Department of Surgery, Camillian Saint Mary’s Hospital Luodong, No. 160, Zhongzheng S. Rd., Luodong Township, Yilan County 26546, Taiwan;
| | - Ping-Chen Wang
- Department of Medical Research and Education, Camillian Saint Mary’s Hospital Luodong, No. 160, Zhongzheng S. Rd., Luodong Township, Yilan County 26546, Taiwan;
| | - Ted Hsiung
- Division of General Surgery, Department of Surgery, Camillian Saint Mary’s Hospital Luodong, No. 160, Zhongzheng S. Rd., Luodong Township, Yilan County 26546, Taiwan; (T.H.); (Y.-H.F.); (J.-T.W.)
| | - Yang-Hsin Fan
- Division of General Surgery, Department of Surgery, Camillian Saint Mary’s Hospital Luodong, No. 160, Zhongzheng S. Rd., Luodong Township, Yilan County 26546, Taiwan; (T.H.); (Y.-H.F.); (J.-T.W.)
| | - Jui-Teng Wu
- Division of General Surgery, Department of Surgery, Camillian Saint Mary’s Hospital Luodong, No. 160, Zhongzheng S. Rd., Luodong Township, Yilan County 26546, Taiwan; (T.H.); (Y.-H.F.); (J.-T.W.)
| | - Wei-Chih Kan
- Department of Nephrology, Department of Internal Medicine, Chi Mei Medical Center, No. 901, Zhonghua Rd., Yongkang Dist., Tainan City 71004, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, No. 89, Wenhua 1st St., Rende Dist., Tainan City 71703, Taiwan
| | - Chih-Chung Shiao
- Division of Nephrology, Department of Internal Medicine, Camillian Saint Mary’s Hospital Luodong, No. 160, Zhongzheng S. Rd., Luodong Township, Yilan County 26546, Taiwan
| |
Collapse
|
2
|
Fan Z, Wu F, Wang P, Wu L, Zhang J, Li W, Pang Q, Zhang A. Serum Irisin Levels Are Positively Correlated with Physical Activity Capacity in Hemodialysis Patients. Kidney Blood Press Res 2025; 50:105-114. [PMID: 39756383 PMCID: PMC11844682 DOI: 10.1159/000543214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/15/2024] [Indexed: 01/07/2025] Open
Abstract
INTRODUCTION Regular physical activity is beneficial for health but is often reduced in patients receiving maintenance hemodialysis treatment. Irisin is a muscle-secreted hormone that reportedly improves metabolism and slows down the progression of some chronic diseases. In this study, we aimed to investigate the relationship between physical activity capacity and serum irisin levels in hemodialysis patients. METHODS Our study included 252 patients undergoing hemodialysis at Xuanwu Hospital Capital Medical University. Enzyme-linked immunosorbent assay was used to measure blood irisin levels. Body composition was analyzed by bioelectrical impedance analysis. The International Physical Activity Questionnaire (IPAQ) was used to score physical activity ability. RESULTS Bivariate correlation analysis showed a positive correlation between IPAQ scores and ln irisin (the natural logarithm of irisin; r = 0.326, p < 0.001). Independent determinants of IPAQ scores were ln irisin, age, fasting glucose, and carbon dioxide combining power. CONCLUSION Our findings provide the first clinical evidence that serum irisin levels are positively correlated with physical activity capacity in hemodialysis patients.
Collapse
Affiliation(s)
- Zhengjia Fan
- Department of Nephrology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Feng Wu
- Department of Nephrology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Peixin Wang
- Department of Nephrology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Leiyun Wu
- Department of Nephrology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Jialing Zhang
- Department of Nephrology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Wen Li
- Department of Nephrology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Qi Pang
- Department of Nephrology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Aihua Zhang
- Department of Nephrology, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Zheng G, Cao J, Wang XH, He W, Wang B. The gut microbiome, chronic kidney disease, and sarcopenia. Cell Commun Signal 2024; 22:558. [PMID: 39574190 PMCID: PMC11580515 DOI: 10.1186/s12964-024-01922-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 11/01/2024] [Indexed: 11/25/2024] Open
Abstract
Sarcopenia is a prevalent condition in patients with chronic kidney disease (CKD), intricately linked to adverse prognoses, heightened cardiovascular risks, and increased mortality rates. Extensive studies have found a close and complex association between gut microbiota, kidney and muscle. On one front, patients with CKD manifest disturbances in gut microbiota and alterations in serum metabolites. These abnormal microbiota composition and metabolites in turn participate in the development of CKD. On another front, altered gut microbiota and its metabolites may lead to significant changes in metabolic homeostasis and inflammation, ultimately contributing to the onset of sarcopenia. The disturbance of gut microbial homeostasis, coupled with the accumulation of toxic metabolites, exerts deleterious effects on skeletal muscles in CKD patients with sarcopenia. This review meticulously describes the alterations observed in gut microbiota and its serum metabolites in CKD and sarcopenia patients, providing a comprehensive overview of pertinent studies. By delving into the intricate interplay of gut microbiota and serum metabolites in CKD-associated sarcopenia, we aim to unveil novel treatment strategies for ameliorating their symptoms and prognosis.
Collapse
Affiliation(s)
- Guohao Zheng
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Jingyuan Cao
- Institute of Nephrology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China
| | - Xiaonan H Wang
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Wei He
- Department of Gastroenterology, Geriatric Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Bin Wang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
4
|
Tazeem MS, Chandrasekaran ND, Srivatsa N. Assessing the Utility of the Metabolic Score for Insulin Resistance (METS-IR) in Evaluating Metabolic Risk Among Individuals Undergoing Master Health Checkups in a Tertiary Care Hospital in South India: A Retrospective Cohort Study. Cureus 2024; 16:e70289. [PMID: 39469359 PMCID: PMC11513226 DOI: 10.7759/cureus.70289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
Background The metabolic score for insulin resistance (METS-IR) is a neoteric score for assessing insulin resistance that has been used as a non-insulin-based, objectively measured method. It is an easily accessible tool that can be used on a large scale to detect insulin resistance in a community. Methods We conducted a retrospective cohort study to explore the utility of this score in identifying metabolic risk in those individuals attending a master health checkup in a tertiary care setting. Data were collected from 254 individuals between October and December 2023. Results According to the univariate regression analysis, METS-IR had a strong correlation in predicting cardiovascular health risks, as evidenced by its positive linear association with an increase in age (β=0.186, p=0.003), weight (β=0.534, p<0.001), waist circumference (β=0.405, p<0.001), and body mass index (BMI; β=0.635, p<0.001). This explained the value of this score in depicting adiposity and insulin resistance. Lab parameters that showed a significant association were fasting blood sugar (β=0.176, p=0.005) and fasting triglycerides (β=0.175, p=0.005). According to the multivariate regression analysis, METS-IR had a significant positive association with fasting blood sugar (B=0.489, p<0.001) and fasting triglycerides (B=0.022, p=0.003), implicating its importance in cardiovascular health. High-density lipoprotein cholesterol (HDL-c; B=-0.168, p=0.005) confirmed its protective role with its negative association in higher quartile groups. An increase in serum albumin levels (B=-0.168, p=0.005) and raised gamma-glutamyl transpeptidase (GGT) (B=0.059, p=0.022) portrays its due diligence in liver health. METS-IR had a weak association with the estimated glomerular filtration rate (eGFR) with Pearson's correlation coefficient of 0.020 (p=0.756) and Spearman's rho of 0.021 (p=0.739). However, raised serum creatinine had a significant association in higher quartile groups, with a p-value of 0.018. Conclusions METS-IR is useful as a screening tool for predicting cardiovascular disease. However, the complex interplay of other confounding factors in identifying renal dysfunction has yet to be explored when considering this score in our study population.
Collapse
Affiliation(s)
- Mohammed Suhail Tazeem
- General Medicine, Sri Ramaswamy Memorial (SRM) Medical College Hospital and Research Centre, SRM Institute of Science and Technology (SRMIST), Chengalpattu, IND
| | - Nirmala Devi Chandrasekaran
- General Medicine, Sri Ramaswamy Memorial (SRM) Medical College Hospital and Research Centre, SRM Institute of Science and Technology (SRMIST), Chengalpattu, IND
| | - Niveda Srivatsa
- Geriatrics, Sri Ramaswamy Memorial (SRM) Medical College Hospital and Research Centre, SRM Institute of Science and Technology (SRMIST), Chengalpattu, IND
| |
Collapse
|
5
|
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
|
6
|
Ahmad SS, Ahmad K, Lim JH, Shaikh S, Lee EJ, Choi I. Therapeutic applications of biological macromolecules and scaffolds for skeletal muscle regeneration: A review. Int J Biol Macromol 2024; 267:131411. [PMID: 38588841 DOI: 10.1016/j.ijbiomac.2024.131411] [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: 01/26/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 04/10/2024]
Abstract
Skeletal muscle (SM) mass and strength maintenance are important requirements for human well-being. SM regeneration to repair minor injuries depends upon the myogenic activities of muscle satellite (stem) cells. However, losses of regenerative properties following volumetric muscle loss or severe trauma or due to congenital muscular abnormalities are not self-restorable, and thus, these conditions have major healthcare implications and pose clinical challenges. In this context, tissue engineering based on different types of biomaterials and scaffolds provides an encouraging means of structural and functional SM reconstruction. In particular, biomimetic (able to transmit biological signals) and several porous scaffolds are rapidly evolving. Several biological macromolecules/biomaterials (collagen, gelatin, alginate, chitosan, and fibrin etc.) are being widely used for SM regeneration. However, available alternatives for SM regeneration must be redesigned to make them more user-friendly and economically feasible with longer shelf lives. This review aimed to explore the biological aspects of SM regeneration and the roles played by several biological macromolecules and scaffolds in SM regeneration in cases of volumetric muscle loss.
Collapse
Affiliation(s)
- Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Jeong Ho Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea.
| |
Collapse
|
7
|
Thome T, Vugman NA, Stone LE, Wimberly K, Scali ST, Ryan TE. A tryptophan-derived uremic metabolite/Ahr/Pdk4 axis governs skeletal muscle mitochondrial energetics in chronic kidney disease. JCI Insight 2024; 9:e178372. [PMID: 38652558 PMCID: PMC11141944 DOI: 10.1172/jci.insight.178372] [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: 12/08/2023] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Chronic kidney disease (CKD) causes accumulation of uremic metabolites that negatively affect skeletal muscle. Tryptophan-derived uremic metabolites are agonists of the aryl hydrocarbon receptor (AHR), which has been shown to be activated in CKD. This study investigated the role of the AHR in skeletal muscle pathology of CKD. Compared with controls with normal kidney function, AHR-dependent gene expression (CYP1A1 and CYP1B1) was significantly upregulated in skeletal muscle of patients with CKD, and the magnitude of AHR activation was inversely correlated with mitochondrial respiration. In mice with CKD, muscle mitochondrial oxidative phosphorylation (OXPHOS) was markedly impaired and strongly correlated with the serum level of tryptophan-derived uremic metabolites and AHR activation. Muscle-specific deletion of the AHR substantially improved mitochondrial OXPHOS in male mice with the greatest uremic toxicity (CKD + probenecid) and abolished the relationship between uremic metabolites and OXPHOS. The uremic metabolite/AHR/mitochondrial axis in skeletal muscle was verified using muscle-specific AHR knockdown in C57BL/6J mice harboring a high-affinity AHR allele, as well as ectopic viral expression of constitutively active mutant AHR in mice with normal renal function. Notably, OXPHOS changes in AHRmKO mice were present only when mitochondria were fueled by carbohydrates. Further analyses revealed that AHR activation in mice led to significantly increased pyruvate dehydrogenase kinase 4 (Pdk4) expression and phosphorylation of pyruvate dehydrogenase enzyme. These findings establish a uremic metabolite/AHR/Pdk4 axis in skeletal muscle that governs mitochondrial deficits in carbohydrate oxidation during CKD.
Collapse
Affiliation(s)
- Trace Thome
- Department of Applied Physiology and Kinesiology and
| | | | | | - Keon Wimberly
- Department of Applied Physiology and Kinesiology and
| | - Salvatore T. Scali
- Division of Vascular Surgery and Endovascular Therapy, University of Florida, Gainesville, Florida, USA
- Malcom Randall VA Medical Center, Gainesville, Florida, USA
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology and
- Center for Exercise Science and
- Myology Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
8
|
Ahmad K, Shaikh S, Lim JH, Ahmad SS, Chun HJ, Lee EJ, Choi I. Therapeutic application of natural compounds for skeletal muscle-associated metabolic disorders: A review on diabetes perspective. Biomed Pharmacother 2023; 168:115642. [PMID: 37812896 DOI: 10.1016/j.biopha.2023.115642] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/19/2023] [Accepted: 10/03/2023] [Indexed: 10/11/2023] Open
Abstract
Skeletal muscle (SM) plays a vital role in energy and glucose metabolism by regulating insulin sensitivity, glucose uptake, and blood glucose homeostasis. Impaired SM metabolism is strongly linked to several diseases, particularly type 2 diabetes (T2D). Insulin resistance in SM may result from the impaired activities of insulin receptor tyrosine kinase, insulin receptor substrate 1, phosphoinositide 3-kinase, and AKT pathways. This review briefly discusses SM myogenesis and the critical roles that SM plays in insulin resistance and T2D. The pharmacological targets of T2D which are associated with SM metabolism, such as DPP4, PTB1B, SGLT, PPARγ, and GLP-1R, and their potential modulators/inhibitors, especially natural compounds, are discussed in detail. This review highlights the significance of SM in metabolic disorders and the therapeutic potential of natural compounds in targeting SM-associated T2D targets. It may provide novel insights for the future development of anti-diabetic drug therapies. We believe that scientists working on T2D therapies will benefit from this review by enhancing their knowledge and updating their understanding of the subject.
Collapse
Affiliation(s)
- Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Jeong Ho Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Hee Jin Chun
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea.
| |
Collapse
|
9
|
Yoon J, Heo SJ, Lee JH, Kwon YJ, Lee JE. Comparison of METS-IR and HOMA-IR for predicting new-onset CKD in middle-aged and older adults. Diabetol Metab Syndr 2023; 15:230. [PMID: 37957738 PMCID: PMC10644442 DOI: 10.1186/s13098-023-01214-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 11/05/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) has emerged as a mounting public health issue worldwide; therefore, prompt identification and prevention are imperative in mitigating CKD-associated complications and mortality rate. We aimed to compare the predictive powers of the homeostatic model assessment for insulin resistance (HOMA-IR) and the metabolic score for insulin resistance (METS-IR) for CKD incidence in middle-aged and older adults. METHODS This study used longitudinal prospective cohort data from the Korean Genome and Epidemiology Study. A total of 10,030 participants, aged 40-69 years, residing in the Ansung or Ansan regions of the Republic of Korea, were recruited between 2001 and 2002 through a two-stage cluster sampling method. We compared the predictive powers of METS-IR and HOMA-IR for CKD prevalence and incidence, respectively. CKD prevalence was measured by the area under the receiver operating characteristic (ROC) curve (AUC), and the indices' predictive performance for CKD incidence were assessed using Harrell's concordance index and time-dependent ROC curve analysis. RESULTS A total of 9261 adults aged 40-69 years at baseline and 8243 adults without CKD were included in this study. The AUCs and 95% confidence intervals (CIs) of HOMA-IR and METS-IR for CKD prevalence at baseline were 0.577 (0.537-0.618) and 0.599 (0.560-0.637), respectively, with no significant difference (p = 0.337). The Heagerty's integrated AUC for METS-IR in predicting CKD incidence was 0.772 (95% CI 0.750-0.799), which was significantly higher than that of HOMA-IR (0.767 [95% CI 0.742-0.791], p = 0.015). CONCLUSION METS-IR surpassed HOMA-IR in predicting CKD incidence and was as effective as HOMA-IR in predicting CKD prevalence. This implies that METS-IR could be a valuable indicator for early detection and prevention of CKD among Korean adults.
Collapse
Affiliation(s)
- Jihyun Yoon
- Department of Family Medicine, Anam Hospital, Korea University College of Medicine, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02481, Republic of Korea
| | - Seok-Jae Heo
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jun-Hyuk Lee
- Department of Family Medicine, Nowon Eulji Medical Center, Eulji University School of Medicine, Seoul, 01830, Republic of Korea
- Department of Medicine, Hanyang University Graduate School of Medicine, Seoul, 04763, Republic of Korea
| | - Yu-Jin Kwon
- Department of Family Medicine, Yonsei University of College of Medicine, Yongin Severance Hospital, Yongin, 16995, Republic of Korea.
| | - Jung Eun Lee
- Division of Nephrology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Gyeonggi, Republic of Korea.
| |
Collapse
|
10
|
Tang J, Zhang H, Yin L, Zhou Q, Zhang H. The gut microbiota from maintenance hemodialysis patients with sarcopenia influences muscle function in mice. Front Cell Infect Microbiol 2023; 13:1225991. [PMID: 37771694 PMCID: PMC10523162 DOI: 10.3389/fcimb.2023.1225991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/23/2023] [Indexed: 09/30/2023] Open
Abstract
Background Sarcopenia is a common complication in patients undergoing maintenance hemodialysis (MHD). Growing evidence suggests a close relationship between the gut microbiota and skeletal muscle. However, research on gut microbiota in patients with sarcopenia undergoing MHD (MS) remains scarce. To bridge this knowledge gap, we aimed to evaluate the pathogenic influence of gut microbiota in the skeletal muscle of patients with MS, to clarify the causal association between gut microbiota and skeletal muscle symptoms in patients with MS and identify the potential mechanisms underlying this causal association. Methods Fecal samples were collected from 10 patients with MS and 10 patients without MS (MNS). Bacteria were extracted from these samples for transplantation. Mice (n=42) were randomly divided into three groups and, after antibiotic treatment, fecal microbiota transplantation (FMT) was performed once a day for 3 weeks. Skeletal muscle and fecal samples from the mice were collected for 16S rRNA gene sequencing and for histological, real-time PCR, and metabolomic analyses. Results Mice colonized with gut microbiota from MS patients exhibited notable decreases in muscle function and muscle mass, compared with FMT from patients with MNS. Moreover, 16S rRNA sequencing revealed that the colonization of MS gut microbiota reduced the abundance of Akkermansia in the mouse intestines. Metabolome analysis revealed that seven metabolic pathways were notably disrupted in mice transplanted with MS microbiota. Conclusion This study established a connection between skeletal muscle and the gut microbiota of patients with MS, implying that disruption of the gut microbiota may be a driving factor in the development of skeletal muscle disorders in patients undergoing MHD. This finding lays the foundation for understanding the pathogenesis and potential treatment methods for sarcopenia in patients undergoing MHD.
Collapse
Affiliation(s)
- Jie Tang
- Lianyungang Clinical College of Nanjing Medical University, The First People’s Hospital of Lianyungang, Lianyungang, China
| | - Hailin Zhang
- Lianyungang Clinical College of Nanjing Medical University, The First People’s Hospital of Lianyungang, Lianyungang, China
| | - Lixia Yin
- Lianyungang Clinical College of Nanjing Medical University, The First People’s Hospital of Lianyungang, Lianyungang, China
| | - Qifan Zhou
- Lianyungang Clinical College of Nanjing Medical University, The First People’s Hospital of Lianyungang, Lianyungang, China
| | - Huipin Zhang
- Department of Hemopurification Center, The Affliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, China
| |
Collapse
|
11
|
Molsted S, Bennett PN, Wilund K, Bruun K, Pakpour AH, Liljehult JM, Brandi L. Nurses' and medical doctors' attitudes towards exercise for people with chronic kidney disease in Denmark. J Ren Care 2023; 49:206-216. [PMID: 36274056 DOI: 10.1111/jorc.12445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 09/29/2022] [Accepted: 10/02/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Implementation of exercise training in people with kidney failure may be affected by clinicians' attitudes. OBJECTIVES To investigate Danish nephrology nurses' and medical doctors' attitudes towards: exercise for people undergoing dialysis; use of physical activity interventions in chronic kidney disease; and to compare Danish and previously reported Australian nurse attitudes. DESIGN Cross-sectional survey. PARTICIPANTS Nurses and medical doctors from the nephrology field in Denmark. MEASUREMENTS The questionnaire attitudes towards exercise in dialysis, and questions about exercise advice, counselling and interventions. RESULTS Nephrology nurses (n = 167) and 17 medical doctors (women 92%, age 47 ± 11 years) from 19 dialysis units participated. There were no differences between nurses' and medical doctors attitudes about training. Ninety-five % and 88% of nurses and medical doctors, respectively, agreed that most people undergoing dialysis could benefit from exercise. Exercise training was offered to people undergoing haemodialyses in 88% of 17 departments. Danish nurses reported more positive attitudes than Australian towards exercise (p < 0.05). Ninety-five % and 86% of the Danish and Australian nurses, respectively, agreed/strongly agreed that most people undergoing dialysis could benefit from exercise. Six % and 35% of the Danish and Australian nurses, respectively, agreed/strongly agreed that most people with dialysis were too sick to exercise. CONCLUSION Danish nephrology nurses and medical doctors had mostly positive attitudes to exercise training to people undergoing dialysis, and exercise to people with dialysis was offered frequently. Danish and Australian nurses had positive attitudes to exercise to people undergoing dialysis, it was however more positive in Danish nurses.
Collapse
Affiliation(s)
- Stig Molsted
- Department of Clinical Research, Nordsjaellands Hospital, Hillerød, Denmark
- Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| | - Paul N Bennett
- Medical & Clinical Affairs, Satellite Healthcare, San Jose, California, USA
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Ken Wilund
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Karina Bruun
- The Department of Nephrology, Rigshospitalet, Copenhagen, Denmark
| | - Amir H Pakpour
- Department of Nursing, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | | | - Lisbet Brandi
- Department of Endocrinology and Nephrology, Nordsjaellands Hospital, Hillerød, Denmark
| |
Collapse
|
12
|
Giha HA. Hidden chronic metabolic acidosis of diabetes type 2 (CMAD): Clues, causes and consequences. Rev Endocr Metab Disord 2023; 24:735-750. [PMID: 37380824 DOI: 10.1007/s11154-023-09816-2] [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] [Accepted: 06/09/2023] [Indexed: 06/30/2023]
Abstract
Interpretation of existing data revealed that chronic metabolic acidosis is a pathognomic feature for type 2 diabetes (T2D), which is described here as "chronic metabolic acidosis of T2D (CMAD)" for the first time. The biochemical clues for the CMAD are summarised in the following; low blood bicarbonate (high anionic gap), low pH of interstitial fluid and urine, and response to acid neutralization, while the causes of extra protons are worked out to be; mitochondrial dysfunction, systemic inflammation, gut microbiota (GM), and diabetic lung. Although, the intracellular pH is largely preserved by the buffer system and ion transporters, a persistent systemic mild acidosis leaves molecular signature in cellular metabolism in diabetics. Reciprocally, there are evidences that CMAD contributes to the initiation and progression of T2D by; reducing insulin production, triggering insulin resistance directly or via altered GM, and inclined oxidative stress. The details about the above clues, causes and consequences of CMAD are obtained by searching literature spanning between 1955 and 2022. Finally, the molecular bases of CMAD are discussed in details by interpretation of an up-to-date data and aid of well constructed diagrams, with a conclusion unravelling that CMAD is a major player in T2D pathophysiology. To this end, the CMAD disclosure offers several therapeutic potentials for prevention, delay or attenuation of T2D and its complications.
Collapse
Affiliation(s)
- Hayder A Giha
- Medical Biochemistry and Molecular Biology, Khartoum, Sudan.
| |
Collapse
|
13
|
Xiang T, Fu P, Zhou L. Sarcopenia and osteosarcopenia among patients undergoing hemodialysis. Front Endocrinol (Lausanne) 2023; 14:1181139. [PMID: 37265691 PMCID: PMC10230055 DOI: 10.3389/fendo.2023.1181139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/04/2023] [Indexed: 06/03/2023] Open
Abstract
Background Sarcopenia and osteoporosis are closely interconnected and associated with adverse health outcomes. Osteosarcopenia is the concurrent presence of the two conditions and has rarely been reported in hemodialysis patients. Whether hemodialysis patients with osteosarcopenia are at greater risk of mortality than those with either condition alone remains unknown. The aim of this study was to explore the prevalence of sarcopenia and its association with osteoporosis and to determine its impact on survival risk in hemodialysis patients. Methods A total of 209 adults undergoing hemodialysis were enrolled from the dialysis center in the West China Hospital of Sichuan University, and our study was registered at the Chinese Clinical Trial Register (number: ChiCTR2100043932). Muscle mass, handgrip strength, bone mineral density (BMD), and biochemical parameters were assessed. All deaths were recorded during a follow-up of 35.15 ± 15.37 months. Results Seventy-eight patients were diagnosed with sarcopenia, with a prevalence of 37.3%. After adjustment for potential confounders, age (OR=1.094, P <0.001), female sex (OR= 3.44, P =0.005), diabetes (OR=3.756, P =0.008), CRP (OR=1.09, P =0.015), serum magnesium (OR=0.755, p=0.042) and BMI (OR=0.701, P <0.001) were independently associated with sarcopenia. Among the 209 patients, 103 patients completed the BMD assessment. The prevalence of osteosarcopenia was 22.3%, while 20.4% of participants had sarcopenia alone and 12.6% had osteoporosis alone. The proportions of patients who died were 13.0% for nonsarcopenia&nonosteoporosis, 15.4% for osteoporosis alone, 47.6% for sarcopenia alone, and 52.2% for osteosarcopenia. Cox regression analysis showed that osteosarcopenia was independently associated with all-cause mortality (HR=3.74, 95% CI: 1.172-11.938), while osteoporosis alone and sarcopenia alone were not. Conclusion Patients undergoing hemodialysis had a high incidence of sarcopenia and osteosarcopenia, muscle mass and strength showed a significant association with BMD, and osteosarcopenia might have a powerful impact on mortality in those patients. Clinical trial registration http://www.chictr.org.cn/, identifier ChiCTR2100043932.
Collapse
Affiliation(s)
| | - Ping Fu
- *Correspondence: Ping Fu, ; Li Zhou,
| | - Li Zhou
- *Correspondence: Ping Fu, ; Li Zhou,
| |
Collapse
|
14
|
Benoit B, Beau A, Bres É, Chanon S, Pinteur C, Vieille-Marchiset A, Jalabert A, Zhang H, Garg P, Strigini M, Vico L, Ruzzin J, Vidal H, Koppe L. Treatment with fibroblast growth factor 19 increases skeletal muscle fiber size, ameliorates metabolic perturbations and hepatic inflammation in 5/6 nephrectomized mice. Sci Rep 2023; 13:5520. [PMID: 37015932 PMCID: PMC10073190 DOI: 10.1038/s41598-023-31874-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 03/20/2023] [Indexed: 04/06/2023] Open
Abstract
Chronic kidney disease (CKD) is associated with osteosarcopenia, and because a physical decline in patients correlates with an increased risk of morbidity, an improvement of the musculoskeletal system is expected to improve morbi-mortality. We recently uncovered that the intestinal hormone Fibroblast Growth Factor 19 (FGF19) is able to promote skeletal muscle mass and strength in rodent models, in addition to its capacity to improve glucose homeostasis. Here, we tested the effects of a treatment with recombinant human FGF19 in a CKD mouse model, which associates sarcopenia and metabolic disorders. In 5/6 nephrectomized (5/6Nx) mice, subcutaneous FGF19 injection (0.1 mg/kg) during 18 days increased skeletal muscle fiber size independently of food intake and weight gain, associated with decreased gene expression of myostatin. Furthermore, FGF19 treatment attenuated glucose intolerance and reduced hepatic expression of gluconeogenic genes in uremic mice. Importantly, the treatment also decreased gene expression of liver inflammatory markers in CKD mice. Therefore, our results suggest that FGF19 may represent a novel interesting therapeutic strategy for a global improvement of sarcopenia and metabolic complications in CKD.
Collapse
Affiliation(s)
- Berengère Benoit
- CarMeN Laboratory, INSERM, INRAE, Claude Bernard Lyon 1 University, Pierre Bénite, France
| | - Alice Beau
- CarMeN Laboratory, INSERM, INRAE, Claude Bernard Lyon 1 University, Pierre Bénite, France
| | - Émilie Bres
- CarMeN Laboratory, INSERM, INRAE, Claude Bernard Lyon 1 University, Pierre Bénite, France
- Department of Nephrology and Nutrition, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Chemin du Grand Revoyet, 69495, Pierre Bénite, France
| | - Stéphanie Chanon
- CarMeN Laboratory, INSERM, INRAE, Claude Bernard Lyon 1 University, Pierre Bénite, France
| | - Claudie Pinteur
- CarMeN Laboratory, INSERM, INRAE, Claude Bernard Lyon 1 University, Pierre Bénite, France
| | | | - Audrey Jalabert
- CarMeN Laboratory, INSERM, INRAE, Claude Bernard Lyon 1 University, Pierre Bénite, France
| | - Hao Zhang
- INSERM U1059, Sainbiose, Jean Monnet University, Saint-Etienne, France
| | - Priyanka Garg
- INSERM U1059, Sainbiose, Jean Monnet University, Saint-Etienne, France
| | - Maura Strigini
- INSERM U1059, Sainbiose, Jean Monnet University, Saint-Etienne, France
| | - Laurence Vico
- INSERM U1059, Sainbiose, Jean Monnet University, Saint-Etienne, France
| | - Jérôme Ruzzin
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Hubert Vidal
- CarMeN Laboratory, INSERM, INRAE, Claude Bernard Lyon 1 University, Pierre Bénite, France
| | - Laetitia Koppe
- CarMeN Laboratory, INSERM, INRAE, Claude Bernard Lyon 1 University, Pierre Bénite, France.
- Department of Nephrology and Nutrition, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Chemin du Grand Revoyet, 69495, Pierre Bénite, France.
| |
Collapse
|
15
|
Serrano E, Shenoy P, Martinez Cantarin MP. Adipose tissue metabolic changes in chronic kidney disease. IMMUNOMETABOLISM (COBHAM (SURREY, ENGLAND)) 2023; 5:e00023. [PMID: 37128293 PMCID: PMC10144329 DOI: 10.1097/in9.0000000000000023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/04/2023] [Indexed: 05/03/2023]
Abstract
Adipose tissue is a complex organ whose functions go beyond being an energy reservoir to sustain proper body energy homeostasis. Functioning as an endocrine organ, the adipose tissue has an active role in the body's metabolic balance regulation through several secreted factors generally termed as adipokines. Thus, adipose tissue dysregulation in chronic kidney disease (CKD) can have a deep impact in the pathophysiology of diseases associated with metabolic dysregulation including metabolic syndrome, insulin resistance (IR), atherosclerosis, and even cachexia. CKD is a progressive disorder linked to increased morbidity and mortality. Despite being characterized by renal function loss, CKD is accompanied by metabolic disturbances such as dyslipidemia, protein energy wasting, chronic low-grade inflammation, IR, and lipid redistribution. Thus far, the mechanisms by which these changes occur and the role of adipose tissue in CKD development and progression are unclear. Further understanding of how these factors develop could have implications for the management of CKD by helping identify pharmacological targets to improve CKD outcomes.
Collapse
Affiliation(s)
- Eurico Serrano
- Division of Nephrology, Department of Medicine, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Prashamsa Shenoy
- Division of Nephrology, Department of Medicine, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Maria Paula Martinez Cantarin
- Division of Nephrology, Department of Medicine, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
- *Correspondence: Maria Paula Martinez Cantarin, E-mail:
| |
Collapse
|
16
|
Ahmadi A, Huda MN, Bennett BJ, Gamboa J, Zelnick LR, Smith LR, Chondronikola M, Raftery D, de Boer IH, Roshanravan B. Chronic Kidney Disease is Associated With Attenuated Plasma Metabolome Response to Oral Glucose Tolerance Testing. J Ren Nutr 2023; 33:316-325. [PMID: 36270479 PMCID: PMC11196097 DOI: 10.1053/j.jrn.2022.09.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/01/2022] [Accepted: 09/25/2022] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Chronic kidney disease (CKD) is associated with decreased anabolic response to insulin contributing to protein-energy wasting. Targeted metabolic profiling of oral glucose tolerance testing (OGTT) may help identify metabolic pathways contributing to disruptions to insulin response in CKD. METHODS Using targeted metabolic profiling, we studied the plasma metabolome response in 41 moderate-to-severe nondiabetic CKD patients and 20 healthy controls at fasting and 2 hours after an oral glucose load. We used linear mixed modeling with random intercepts, adjusting for age, gender, race/ethnicity, body weight, and batch to assess heterogeneity in response to OGTT by CKD status. RESULTS Mean estimated glomerular filtration rate among CKD participants was 38.9 ± 12.7 mL/min per 1.73 m2 compared to 87.2 ± 17.7 mL/min per 1.73 m2 among controls. Glucose ingestion induced an anabolic response resulting in increased glycolysis products and a reduction in a wide range of metabolites including amino acids, tricarboxylic acid cycle intermediates, and purine nucleotides compared to fasting. Participants with CKD demonstrated a blunted anabolic response to OGTT evidenced by significant changes in 13 metabolites compared to controls. The attenuated metabolome response predominant involved mitochondrial energy metabolism, vitamin B family, and purine nucleotides. Compared to controls, CKD participants had elevated lactate:pyruvate (L:P) ratio and decreased guanosine diphosphate:guanosine triphosphate ratio during OGTT. CONCLUSION Metabolic profiling of OGTT response suggests a broad disruption of mitochondrial energy metabolism in CKD patients. These findings motivate further investigation into the impact of insulin sensitizers and mitochondrial targeted therapeutics on energy metabolism in patients with nondiabetic CKD.
Collapse
Affiliation(s)
- Armin Ahmadi
- Department of Medicine, Division of Nephrology, University of California Davis, Davis, California
| | - M Nazmul Huda
- Obesity and Metabolism Research Unit, Western Human Nutrition Research Center, USDA, ARS, Davis, California; Department of Nutrition, University of California Davis, Davis, California
| | - Brian J Bennett
- Obesity and Metabolism Research Unit, Western Human Nutrition Research Center, USDA, ARS, Davis, California; Department of Nutrition, University of California Davis, Davis, California
| | - Jorge Gamboa
- Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Leila R Zelnick
- Division of Nephrology and Kidney Research Institute, University of Washington, Seattle, Washington
| | - Lucas R Smith
- Department of Physical Medicine and Rehabilitation, School of Medicine, UCD, Davis, California
| | | | - Daniel Raftery
- Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | - Ian H de Boer
- Division of Nephrology and Kidney Research Institute, University of Washington, Seattle, Washington; Puget Sound Health Care System, Seattle, Washington
| | - Baback Roshanravan
- Department of Medicine, Division of Nephrology, University of California Davis, Davis, California.
| |
Collapse
|
17
|
Ke B, Shen W, Song J, Fang X. MG53: A potential therapeutic target for kidney disease. Pharmacol Res Perspect 2023; 11:e01049. [PMID: 36583464 PMCID: PMC9801490 DOI: 10.1002/prp2.1049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022] Open
Abstract
Ensuring cell survival and tissue regeneration by maintaining cellular integrity is important to the pathophysiology of many human diseases, including kidney disease. Mitsugumin 53 (MG53) is a member of the tripartite motif-containing (TRIM) protein family that plays an essential role in repairing cell membrane injury and improving tissue regeneration. In recent years, an increasing number of studies have demonstrated that MG53 plays a renoprotective role in kidney diseases. Moreover, with the beneficial effects of the recombinant human MG53 (rhMG53) protein in the treatment of kidney diseases in different animal models, rhMG53 shows significant therapeutic potential in kidney disease. In this review, we elucidate the role of MG53 and its molecular mechanism in kidney disease to provide new approaches to the treatment of kidney disease.
Collapse
Affiliation(s)
- Ben Ke
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wen Shen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital to Nanchang University, Nanchang, China
| | - Jianling Song
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiangdong Fang
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
18
|
Sadri H, Ghaffari MH, Sauerwein H. Invited review: Muscle protein breakdown and its assessment in periparturient dairy cows. J Dairy Sci 2023; 106:822-842. [PMID: 36460512 DOI: 10.3168/jds.2022-22068] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022]
Abstract
Mobilization of body reserves including fat, protein, and glycogen is necessary to overcome phases of negative nutrient balance typical for high-yielding dairy cows during the periparturient period. Skeletal muscle, the largest internal organ in mammals, plays a crucial role in maintaining metabolic homeostasis. However, unlike in liver and adipose tissue, the metabolic and regulatory role of skeletal muscle in the adaptation of dairy cows to the physiological needs of pregnancy and lactation has not been studied extensively. The functional integrity and quality of skeletal muscle are maintained through a constant turnover of protein, resulting from both protein breakdown and protein synthesis. Thus, muscle protein breakdown (MPB) and synthesis are intimately connected and tightly controlled to ensure proper protein homeostasis. Understanding the regulation of MPB, the catabolic component of muscle turnover, and its assessment are therefore important considerations to provide information about the timing and extent of tissue mobilization in periparturient dairy cows. Based on animal models and human studies, it is now evident that MPB occurs via the integration of 3 main systems: autophagy-lysosomal, calpain Ca2+-dependent cysteine proteases, and the ubiquitin-proteasome system. These 3 main systems are interconnected and do not work separately, and the regulation is complex. The ubiquitin-proteasomal system is the most well-known cellular proteolytic system and plays a fundamental role in muscle physiology. Complete degradation of a protein often requires a combination of the systems, depending on the physiological situation. Determination of MPB in dairy cows is technically challenging, resulting in a relative dearth of information. The methods for assessing MPB can be divided into either direct or indirect measurements, both having their strengths and limitations. Available information on the direct measures of MPB primarily comes from stable isotopic tracer methods and those of indirect measurements from assessing expression and activity measures of the components of the 3 MPB systems in muscle biopsy samples. Other indirect approaches (i.e., potential indicators of MPB), including ultrasound imaging and measuring metabolites from muscle degradation (i.e., 3-methylhistidine and creatinine), seem to be applicable methods and can provide useful information about the extent and timing of MPB. This review presents our current understanding, including methodological considerations, of the process of MPB in periparturient dairy cows.
Collapse
Affiliation(s)
- H Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, 5166616471 Tabriz, Iran; Institute of Animal Science, Physiology Unit, University of Bonn, 53111 Bonn, Germany.
| | - M H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53111 Bonn, Germany
| | - H Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53111 Bonn, Germany
| |
Collapse
|
19
|
Impaired muscle stem cell function and abnormal myogenesis in acquired myopathies. Biosci Rep 2023; 43:232343. [PMID: 36538023 PMCID: PMC9829652 DOI: 10.1042/bsr20220284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle possesses a high plasticity and a remarkable regenerative capacity that relies mainly on muscle stem cells (MuSCs). Molecular and cellular components of the MuSC niche, such as immune cells, play key roles to coordinate MuSC function and to orchestrate muscle regeneration. An abnormal infiltration of immune cells and/or imbalance of pro- and anti-inflammatory cytokines could lead to MuSC dysfunctions that could have long lasting effects on muscle function. Different genetic variants were shown to cause muscular dystrophies that intrinsically compromise MuSC function and/or disturb their microenvironment leading to impaired muscle regeneration that contributes to disease progression. Alternatively, many acquired myopathies caused by comorbidities (e.g., cardiopulmonary or kidney diseases), chronic inflammation/infection, or side effects of different drugs can also perturb MuSC function and their microenvironment. The goal of this review is to comprehensively summarize the current knowledge on acquired myopathies and their impact on MuSC function. We further describe potential therapeutic strategies to restore MuSC regenerative capacity.
Collapse
|
20
|
Genders AJ, Kuang J, Saner NJ, Botella J, Bishop DJ. Ammonium chloride administration prevents training-induced improvements in mitochondrial respiratory function in the soleus muscle of male rats. Am J Physiol Cell Physiol 2023; 324:C67-C75. [PMID: 36542512 DOI: 10.1152/ajpcell.00165.2022] [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: 04/22/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022]
Abstract
Exercise training can increase both mitochondrial content and mitochondrial respiration. Despite its popularity, high-intensity exercise can be accompanied by mild acidosis (also present in certain pathological states), which may limit exercise-induced adaptations to skeletal muscle mitochondria. The aim of this study was to determine if administration of ammonium chloride (0.05 g/kg) to Wistar rats before each individual exercise session (5 high-intensity exercise sessions/wk for 8 wk) reduced training-induced increases in mitochondrial content (measured by citrate synthase activity and protein content of electron transport system complexes) and respiration (measured in permeabilized muscle fibers). In the soleus muscle, the exercise-training-induced increase in mitochondrial respiration was reduced in rats administered ammonium chloride compared to control animals, but mitochondrial content was not altered. These effects were not present in the white gastrocnemius muscle. In conclusion, ammonium chloride administration before each exercise session over 8 wk reduced improvements in mitochondrial respiration in the soleus muscle but did not alter mitochondrial content. This suggests that mild acidosis may affect training-induced improvements in the respiration of mitochondria in some muscles.
Collapse
Affiliation(s)
- Amanda J Genders
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Jujiao Kuang
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Australian Institute for Musculoskeletal Sciences, Melbourne, Australia
| | - Nicholas J Saner
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Human Integrative Physiology, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Javier Botella
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Metabolic Research Unit, Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Australia
| | - David J Bishop
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| |
Collapse
|
21
|
Naser IA, Abutair AS, Zourob RJ, Qeshta RI, Tawil RL, Lafi AH, Bardwil RW, Tabasi FM. Nutritional Assessment of Adult Patients Undergoing Maintenance Hemodialysis in the Gaza Strip. SAUDI JOURNAL OF KIDNEY DISEASES AND TRANSPLANTATION 2023; 34:1-12. [PMID: 38092711 DOI: 10.4103/1319-2442.390997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Malnutrition is a common condition in patients undergoing hemodialysis (HD), and it is associated with increased morbidity and mortality. The main objective of the study was to evaluate the nutritional status of patients on maintenance HD. After applying eligibility criteria, 141 HD patients attending major governmental dialysis centers were randomly recruited in this cross-sectional study and assessed for nutritional status using the Patient-Generated Subjective Global Assessment (PG-SGA) tool. The PG-SGA categorizes patients as well-nourished, moderately malnourished, and severely malnourished. Different anthropometric measurements, laboratory investigations, blood pressure measurements, and 24-h dietary recall were collected from each patient. According to PG- SGA results, 78% of patients were moderately malnourished and 22% of patients were severely malnourished. The mean body mass index was 27.8 kg/m2, and 5.7% of patients were underweight. There were significant differences in the mid-upper arm muscle circumference (P = 0.020) between the PG-SGA groups. The total energy and protein intake were significantly (P <0.001) less than the recommended dietary intake by 1268.9 kcal and 41.4 g, respectively. The albumin level in 37.6% of patients was less than the normal level, and the results indicated that there were significant differences in serum iron (P = 0.022) between the moderately and severely malnourished patients. The results of this study indicated that all HD patients were suffering from different degrees of malnutrition and, unfortunately, most of their energy and nutrient intake was far less than the requirements, which might be the reason why they face nutritional and health risks.
Collapse
Affiliation(s)
- Ihab A Naser
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, Al-Azhar University, Gaza, Palestine
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Association between the triglyceride–glucose index and chronic kidney disease in adults. Int Urol Nephrol 2022; 55:1279-1289. [PMID: 36472799 DOI: 10.1007/s11255-022-03433-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Chronic kidney disease (CKD) is characterized as a progressive dysfunction of the kidney, and it might have a close relationship with insulin resistance. We utilized the triglyceride-glucose (TyG) index, a reliable marker of insulin resistance, to evaluate the association between the TyG index and CKD in adults from the general population. METHODS This was a cross-sectional study obtaining data from the 2015-2018 National Health and Nutrition Examination Survey. The estimated glomerular filtration rate (eGFR) and urinary albumin-to-creatinine ratio (UACR) served as kidney function indicators. We defined CKD as the existence of either low eGFR (eGFR < 60 mL/min/1.73 m2 BSA) or albuminuria (UACR > 30 mg/g). Multivariate regressions, correlated subgroup analyses, and interaction terms were performed in this study. RESULTS For 4361 recruited participants, the mean TyG index was 8.60 ± 0.68, and the prevalence of CKD was 13.35%. Participants with a higher TyG index showed a higher UACR level (β = 25.10, 95% CI: 6.76, 43.44, P = 0.0074) and higher levels of CKD (OR = 1.34, 95% CI: 1.13, 1.59, P = 0.0006). The positive relationship between the TyG index and CKD became stronger and remained significant in the overweight (OR = 1.61, 95% CI: 1.18, 2.20, P = 0.0027) and obese (OR = 2.48, 95% CI: 1.95, 3.15, P < 0.0001) groups and in people with diabetes (OR = 1.94, 95% CI: 1.46, 2.56, P < 0.0001). CONCLUSIONS Higher TyG index was strongly associated with a higher UACR level and higher values of albuminuria and CKD, which might be useful in kidney function screening especially among people in disadvantageous socioeconomic conditions with no availability for direct measurement of kidney function. However, more well-designed studies are still needed to validate this relationship.
Collapse
|
23
|
Transcription factor NRF2 as potential therapeutic target for preventing muscle wasting in aging chronic kidney disease patients. J Nephrol 2022; 35:2215-2225. [PMID: 36322291 PMCID: PMC9700608 DOI: 10.1007/s40620-022-01484-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/01/2022] [Indexed: 11/27/2022]
Abstract
Increased muscle protein catabolism leading to muscle wasting is a prominent feature of the syndrome of protein-energy wasting (PEW) in patients with chronic kidney disease (CKD). PEW and muscle wasting are induced by factors such as inflammation, oxidative stress and metabolic acidosis that activate the ubiquitin-proteasome system, the main regulatory mechanism of skeletal muscle degradation. Whether deficiency of nuclear factor erythroid 2-related factor 2 (NRF2), which regulates expression of antioxidant proteins protecting against oxidative damage triggered by inflammation, may exacerbate PEW has yet to be examined in aging patients with CKD. This review focuses on the hypothesis that NRF2 is involved in the maintenance of muscle mass and explores whether sustained activation of NRF2 by non-pharmacological interventions using nutraceutical activators to improve redox homeostasis could be a plausible strategy to prevent skeletal muscle disorders, including muscle wasting, sarcopenia and frailty associated with PEW in aging CKD patients.
Collapse
|
24
|
Tariq H, Dobre M. Metabolic acidosis post kidney transplantation. Front Physiol 2022; 13:989816. [PMID: 36082221 PMCID: PMC9445136 DOI: 10.3389/fphys.2022.989816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022] Open
Abstract
Metabolic acidosis, a common complication in patients with chronic kidney disease (CKD), results in a multitude of deleterious effects. Though the restoration of kidney function following transplantation is generally accompanied by a correction of metabolic acidosis, a subset of transplant recipients remains afflicted by this ailment and its subsequent morbidities. The vulnerability of kidney allografts to metabolic acidosis can be attributed to reasons similar to pathogenesis of acidosis in non-transplant CKD, and to transplant specific causes, including donor related, recipient related, immune mediated factors, and immunosuppressive medications. Correction of metabolic acidosis in kidney transplantation either with alkali therapy or through dietary manipulations may have potential benefits and the results of such clinical trials are eagerly awaited. This review summarizes the published evidence on the pathogenesis and clinical consequences of chronic metabolic acidosis in kidney transplant recipients.
Collapse
Affiliation(s)
- Hafsa Tariq
- Division of Nephrology, University of Rochester Medical Center, Rochester, NY, United States
| | - Mirela Dobre
- Division of Nephrology and Hypertension, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, United States
- *Correspondence: Mirela Dobre,
| |
Collapse
|
25
|
Rhea EM, Banks WA, Raber J. Insulin Resistance in Peripheral Tissues and the Brain: A Tale of Two Sites. Biomedicines 2022; 10:1582. [PMID: 35884888 PMCID: PMC9312939 DOI: 10.3390/biomedicines10071582] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 12/12/2022] Open
Abstract
The concept of insulin resistance has been around since a few decades after the discovery of insulin itself. To allude to the classic Charles Dicken's novel published 62 years before the discovery of insulin, in some ways, this is the best of times, as the concept of insulin resistance has expanded to include the brain, with the realization that insulin has a life beyond the regulation of glucose. In other ways, it is the worst of times as insulin resistance is implicated in devastating diseases, including diabetes mellitus, obesity, and Alzheimer's disease (AD) that affect the brain. Peripheral insulin resistance affects nearly a quarter of the United States population in adults over age 20. More recently, it has been implicated in AD, with the degree of brain insulin resistance correlating with cognitive decline. This has led to the investigation of brain or central nervous system (CNS) insulin resistance and the question of the relation between CNS and peripheral insulin resistance. While both may involve dysregulated insulin signaling, the two conditions are not identical and not always interlinked. In this review, we compare and contrast the similarities and differences between peripheral and CNS insulin resistance. We also discuss how an apolipoprotein involved in insulin signaling and related to AD, apolipoprotein E (apoE), has distinct pools in the periphery and CNS and can indirectly affect each system. As these systems are both separated but also linked via the blood-brain barrier (BBB), we discuss the role of the BBB in mediating some of the connections between insulin resistance in the brain and in the peripheral tissues.
Collapse
Affiliation(s)
- Elizabeth M. Rhea
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA; (E.M.R.); (W.A.B.)
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - William A. Banks
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA; (E.M.R.); (W.A.B.)
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
- Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
26
|
Ho JQ, Abramowitz MK. Clinical Consequences of Metabolic Acidosis-Muscle. Adv Chronic Kidney Dis 2022; 29:395-405. [PMID: 36175077 DOI: 10.1053/j.ackd.2022.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/10/2022] [Accepted: 04/25/2022] [Indexed: 01/25/2023]
Abstract
Metabolic acidosis is common in people with chronic kidney disease and can contribute to functional decline, morbidity, and mortality. One avenue through which metabolic acidosis can result in these adverse clinical outcomes is by negatively impacting skeletal muscle; this can occur through several pathways. First, metabolic acidosis promotes protein degradation and impairs protein synthesis, which lead to muscle breakdown. Second, metabolic acidosis hinders mitochondrial function, which decreases oxidative phosphorylation and reduces energy production. Third, metabolic acidosis directly limits muscle contraction. The purpose of this review is to examine the specific mechanisms of each pathway through which metabolic acidosis affects muscle, the impact of metabolic acidosis on physical function, and the effect of treating metabolic acidosis on functional outcomes.
Collapse
Affiliation(s)
- Jim Q Ho
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Matthew K Abramowitz
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY; Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY; Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
27
|
Douvris A, Viñas J, Burns KD. miRNA-486-5p: signaling targets and role in non-malignant disease. Cell Mol Life Sci 2022; 79:376. [PMID: 35731367 PMCID: PMC9217846 DOI: 10.1007/s00018-022-04406-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 11/30/2022]
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs, highly conserved between species, that are powerful regulators of gene expression. Aberrant expression of miRNAs alters biological processes and pathways linked to human disease. miR-486-5p is a muscle-enriched miRNA localized to the cytoplasm and nucleus, and is highly abundant in human plasma and enriched in small extracellular vesicles. Studies of malignant and non-malignant diseases, including kidney diseases, have found correlations with circulating miR-486-5p levels, supporting its role as a potential biomarker. Pre-clinical studies of non-malignant diseases have identified miR-486-5p targets that regulate major signaling pathways involved in cellular proliferation, migration, angiogenesis, and apoptosis. Validated miR-486-5p targets include phosphatase and tensin homolog (PTEN) and FoXO1, whose suppression activates phosphatidyl inositol-3-kinase (PI3K)/Akt signaling. Targeting of Smad1/2/4 and IGF-1 by miR-486-5p inhibits transforming growth factor (TGF)-β and insulin-like growth factor-1 (IGF-1) signaling, respectively. Other miR-486-5p targets include matrix metalloproteinase-19 (MMP-19), Sp5, histone acetyltransferase 1 (HAT1), and nuclear factor of activated T cells-5 (NFAT5). In this review, we examine the biogenesis, regulation, validated gene targets and biological effects of miR-486-5p in non-malignant diseases.
Collapse
Affiliation(s)
- Adrianna Douvris
- Division of Nephrology, Department of Medicine and Kidney Research Centre, The Ottawa Hospital Research Institute, University of Ottawa, 1967 Riverside Dr., Rm. 535, Ottawa, ON, K1H 7W9, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jose Viñas
- Division of Nephrology, Department of Medicine and Kidney Research Centre, The Ottawa Hospital Research Institute, University of Ottawa, 1967 Riverside Dr., Rm. 535, Ottawa, ON, K1H 7W9, Canada
| | - Kevin D Burns
- Division of Nephrology, Department of Medicine and Kidney Research Centre, The Ottawa Hospital Research Institute, University of Ottawa, 1967 Riverside Dr., Rm. 535, Ottawa, ON, K1H 7W9, Canada. .,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
28
|
Baker LA, O'Sullivan TF, Robinson KA, Graham-Brown MPM, Major RW, Ashford RU, Smith AC, Philp A, Watson EL. Primary skeletal muscle cells from chronic kidney disease patients retain hallmarks of cachexia in vitro. J Cachexia Sarcopenia Muscle 2022; 13:1238-1249. [PMID: 35029054 PMCID: PMC8978027 DOI: 10.1002/jcsm.12802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/23/2021] [Accepted: 08/23/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Skeletal muscle wasting and dysfunction are common characteristics noted in people who suffer from chronic kidney disease (CKD). The mechanisms by which this occurs are complex, and although progress has been made, the key underpinning mechanisms are not yet fully elucidated. With work to date primarily conducted in nephrectomy-based animal models, translational capacity to our patient population has been challenging. This could be overcome if rationale developing work could be conducted in human based models with greater translational capacity. This could be achieved using cells derived from patient biopsies, if they retain phenotypic traits noted in vivo. METHODS Here, we performed a systematic characterization of CKD derived muscle cells (CKD; n = 10; age: 54.40 ± 15.53 years; eGFR: 22.25 ± 13.22 ml/min/1.73 m2 ) in comparison with matched controls (CON; n = 10; age: 58.66 ± 14.74 years; eGFR: 85.81 ± 8.09 ml/min/1.73 m2 ). Harvested human derived muscle cells (HDMCs) were taken through proliferative and differentiation phases and investigated in the context of myogenic progression, inflammation, protein synthesis, and protein breakdown. Follow up investigations exposed HDMC myotubes from each donor type to 0, 0.4, and 100 nM of IGF-1 in order to investigate any differences in anabolic resistance. RESULTS Harvested human derived muscle cells isolated from CKD patients displayed higher rates of protein degradation (P = 0.044) alongside elevated expression of both TRIM63 (2.28-fold higher, P = 0.054) and fbox32 (6.4-fold higher, P < 0.001) in comparison with CONs. No differences were noted in rates of protein synthesis under basal conditions (P > 0.05); however, CKD derived cells displayed a significant degree of anabolic resistance in response to IGF-1 stimulation (both doses) in comparison with matched CONs (0.4 nm: P < 0.001; 100 nM: P < 0.001). CONCLUSIONS In summary, we report for the first time that HDMCs isolated from people suffering from CKD display key hallmarks of the well documented in vivo phenotype. Not only do these findings provide further mechanistic insight into CKD specific cachexia, but they also demonstrate this is a reliable and suitable model in which to perform targeted experiments to begin to develop novel therapeutic strategies targeting the CKD associated decline in skeletal muscle mass and function.
Collapse
Affiliation(s)
- Luke A Baker
- Department of Health Sciences, University of Leicester, Leicester, UK
| | | | | | - Matthew P M Graham-Brown
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK.,Department of Cardiovascular Science, NIHR Leicester Cardiovascular Biomedical Research Unit, Leicester, UK
| | - Rupert W Major
- Department of Health Sciences, University of Leicester, Leicester, UK.,John Walls Renal Unit, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Robert U Ashford
- Leicester Orthopaedics, University Hospitals of Leicester, Leicester, UK.,Department of Cancer Studies, University of Leicester, Leicester, UK
| | - Alice C Smith
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Andrew Philp
- Mitochondrial Metabolism and Ageing Laboratory, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, UNSW Medicine, UNSW, Sydney, NSW, Australia
| | - Emma L Watson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| |
Collapse
|
29
|
Gonzalez P, Lozano P, Solano F. Unraveling the Metabolic Hallmarks for the Optimization of Protein Intake in Pre-Dialysis Chronic Kidney Disease Patients. Nutrients 2022; 14:nu14061182. [PMID: 35334840 PMCID: PMC8954715 DOI: 10.3390/nu14061182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/28/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023] Open
Abstract
The daily amount and quality of protein that should be administered by enteral nutrition in pre-dialysis chronic kidney disease (CKD) patients is a widely studied but still controversial issue. This is due to a compromise between the protein necessary to maintain muscular proteostasis avoiding sarcopenia, and the minimal amount required to prevent uremia and the accumulation of nitrogenous toxic substances in blood because of the renal function limitations. This review underlines some intracellular and extracellular features that should be considered to reconcile those two opposite factors. On one hand, the physiological conditions and usual side effects associated with CKD, mTOR and other proteins and nutrients involved in the regulation of protein synthesis in the muscular tissue are discussed. On the other hand, the main digestive features of the most common proteins used for enteral nutrition formulation (i.e., whey, casein and soy protein) are highlighted, due to the importance of supplying key amino acids to serum and tissues to maintain their concentration above the anabolic threshold needed for active protein synthesis, thereby minimizing the catabolic pathways leading to urea formation.
Collapse
Affiliation(s)
- Patricia Gonzalez
- Project Manager, Fresenius Kabi España, Sociedad Anonima Unipersonal, Marina 16-18, 08005 Barcelona, Spain
- Correspondence: (P.G.); (F.S.)
| | - Pedro Lozano
- Department of Biochemistry and Molecular Biology “B” and Immunology, Faculty of Chemistry, Campus de Espinardo, University of Murcia, 30100 Murcia, Spain;
| | - Francisco Solano
- Department of Biochemistry and Molecular Biology “B” and Immunology, IMIB (Murcian Institute of Health Research), Faculty of Medicine, Campus de Espinardo, University of Murcia, 30100 Murcia, Spain
- Correspondence: (P.G.); (F.S.)
| |
Collapse
|
30
|
Wang XH, Mitch WE, Price SR. Pathophysiological mechanisms leading to muscle loss in chronic kidney disease. Nat Rev Nephrol 2022; 18:138-152. [PMID: 34750550 DOI: 10.1038/s41581-021-00498-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 12/16/2022]
Abstract
Loss of muscle proteins is a deleterious consequence of chronic kidney disease (CKD) that causes a decrease in muscle strength and function, and can lead to a reduction in quality of life and increased risk of morbidity and mortality. The effectiveness of current treatment strategies in preventing or reversing muscle protein losses is limited. The limitations largely stem from the systemic nature of diseases such as CKD, which stimulate skeletal muscle protein degradation pathways while simultaneously activating mechanisms that impair muscle protein synthesis and repair. Stimuli that initiate muscle protein loss include metabolic acidosis, insulin and IGF1 resistance, changes in hormones, cytokines, inflammatory processes and decreased appetite. A growing body of evidence suggests that signalling molecules secreted from muscle can enter the circulation and subsequently interact with recipient organs, including the kidneys, while conversely, pathological events in the kidney can adversely influence protein metabolism in skeletal muscle, demonstrating the existence of crosstalk between kidney and muscle. Together, these signals, whether direct or indirect, induce changes in the levels of regulatory and effector proteins via alterations in mRNAs, microRNAs and chromatin epigenetic responses. Advances in our understanding of the signals and processes that mediate muscle loss in CKD and other muscle wasting conditions will support the future development of therapeutic strategies to reduce muscle loss.
Collapse
Affiliation(s)
- Xiaonan H Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, GA, USA
| | - William E Mitch
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - S Russ Price
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC, USA. .,Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
31
|
Cha RH, Kang SH, Han MY, An WS, Kim SH, Kim JC. Effects of AST-120 on muscle health and quality of life in chronic kidney disease patients: results of RECOVERY study. J Cachexia Sarcopenia Muscle 2022; 13:397-408. [PMID: 34862753 PMCID: PMC8818653 DOI: 10.1002/jcsm.12874] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND The prevalence of sarcopenia is increased with declining renal function. Elevated serum indoxyl sulfate levels are associated with poor skeletal muscle conditions. We aimed to determine the effects of AST-120, the oral adsorbent of indoxyl sulfate, on sarcopenia and sarcopenia-associated factors in chronic kidney disease patients. METHODS This was a 48 week, randomized controlled, parallel group, open-label, multicentre trial (n = 150). The participants were randomly assigned in a 1:1 ratio to the control (CON) and AST-120 (Renamezin®, REN) groups. Outcome measurements were performed at baseline and every 24 weeks for 48 weeks. The primary outcome was gait speed difference ≥0.1 m/s between the two groups, and secondary outcomes included hand grip strength, muscle mass, and health-related quality of life. RESULTS A difference of gait speed ≥0.1 m/s was not observed during the study period. The mean dynamic-start gait speed in the REN group increased from baseline to 48 weeks (1.04 ± 0.31 to 1.08 ± 0.32 m/s, P = 0.019). The static-start gait speed changed by -0.024 and 0.04 m/s (P = 0.049) in the CON and REN groups over 48 weeks, respectively. Hand grip strength decreased during the first 24 weeks and did not significantly change over the next 24 weeks in either group. The proportion of low muscle mass or sarcopenia at baseline was larger in the REN group than in the CON group, but the difference attenuated over the study period [low muscle mass and sarcopenia in the CON and REN groups at baseline, 4.0% vs. 18.9% (P = 0.004) and 2.7% vs. 13.5% (P = 0.017); at 24 weeks, 2.9% vs. 13.6% (P = 0.021) and 1.4% vs. 10.5% (P = 0.029); and at 48 weeks, 7.6% vs. 12.9% (P = 0.319) and 4.5% vs. 8.1% (P = 0.482), respectively]. Bodily pain, vitality, symptoms/problems, and cognitive function in the REN group improved, while the quality of social interactions and the kidney disease effects in the CON group aggravated from baseline to 48 weeks. Interaction between time and group was evident only in symptoms/problems, cognitive function, and kidney disease effects. CONCLUSIONS The addition of AST-120 to standard treatment in chronic kidney disease patients did not make a significant difference in gait speed, although AST-120 modestly had beneficial effects on gait speed change and quality of life and showed the potential to improve sarcopenia. (clinicaltrials.gov: NCT03788252).
Collapse
Affiliation(s)
- Ran-Hui Cha
- Department of Internal Medicine, National Medical Center, Seoul, Republic of Korea
| | - Seok Hui Kang
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Mi Yeun Han
- Department of Internal Medicine, Hallym University Hangang Sacred Heart Hospital, Seoul, Republic of Korea
| | - Won Suk An
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Su-Hyun Kim
- Department of Internal Medicine, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Jun Chul Kim
- Department of Internal Medicine, CHA Gumi Medical Center, CHA University, Gumi, Republic of Korea
| |
Collapse
|
32
|
Delgado C, Chiang JM, Kittiskulnam P, Sheshadri A, Grimes B, Segal M, Kaysen GA, Johansen KL. Longitudinal Assessment of Body Composition and Its Association With Survival Among Participants of the ACTIVE/ADIPOSE Study. J Ren Nutr 2021; 32:396-404. [PMID: 34930665 DOI: 10.1053/j.jrn.2021.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 08/16/2021] [Accepted: 09/05/2021] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVES The importance of muscle wasting as a predictor of mortality in the hemodialysis population is not clear. Lack of association of muscle mass with survival in some studies could be related to reliance on single measures or to incorporation of excess extracellular water (ECW) into estimates of muscle mass. We examined changes in body composition over a 2-year period and the association of body composition with survival. DESIGN AND METHODS We analyzed data from 325 adults receiving hemodialysis in the Bay Area. We estimated ECW, intracellular water (ICW), and fat mass by whole-body bioimpedance spectroscopy (BIS) at 0, 12, and 24 months from enrollment. We used linear mixed modeling to examine changes in body mass index and BIS-derived estimates of body composition and Cox modeling with BIS-derived estimates as time-varying independent variables to examine associations between body composition and survival in multivariable analyses. RESULTS Body mass index declined over time. Considering individual components of body composition, ICW declined (-0.09 kg/m2 per year, 95% confidence interval -0.14 to -0.04), but fat mass and ECW did not change significantly. There were 120 deaths over a median of 5.2 years. The relationship between ICW and mortality was not linear such that the association was steeper at low values of ICW, whereas higher ICW was associated with better survival that was relatively stable above 9 kg/m2. Higher ECW was associated with higher mortality, and fat mass was not associated with survival. These associations were independent of markers of inflammation and nutritional status. CONCLUSIONS ICW declined over 2 years in this cohort, whereas fat mass and ECW remained relatively stable. Higher ICW was associated with better survival, but higher fat mass was not. Higher ECW was associated with worse survival. These results suggest that muscle mass may predict survival among patients on hemodialysis.
Collapse
Affiliation(s)
- Cynthia Delgado
- Division of Nephrology, University of California, San Francisco and Nephrology Section, San Francisco VA Medical Center, San Francisco, California, USA.
| | - Janet M Chiang
- Division of Endocrinology, University of California, San Francisco and San Francisco VA Medical Center, San Francisco, California, USA
| | | | - Anoop Sheshadri
- Division of Nephrology, University of California, San Francisco and Nephrology Section, San Francisco VA Medical Center, San Francisco, California, USA
| | - Barbara Grimes
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco California, USA
| | - Mark Segal
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco California, USA
| | - George A Kaysen
- Department of Medicine, Division of Nephrology, University of California, Davis, Davis, California, USA; Department of Biochemistry and Molecular Medicine University of California, Davis, Davis, California, USA
| | - Kirsten L Johansen
- Department of Medicine, Division of Nephrology, Hennepin Healthcare, Minneapolis, Minnesota, USA; Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
33
|
Małodobra-Mazur M, Lewoń D, Cierzniak A, Okulus M, Gliszczyńska A. Phospholipid Derivatives of Cinnamic Acid Restore Insulin Sensitivity in Insulin Resistance in 3T3-L1 Adipocytes. Nutrients 2021; 13:3619. [PMID: 34684619 PMCID: PMC8537072 DOI: 10.3390/nu13103619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Insulin resistance (IR) is a condition in which the physiological amount of insulin is insufficient to evoke a proper response of the cell, that is, glucose utilization. Metformin is the first choice for therapy, thanks to its glycemic efficacy and general tolerability. In addition, various natural compounds from plant extracts, spices, and essential oils have been shown to provide health benefits regarding insulin sensitivity. In the present study, we analyzed the effect of phospholipid derivatives of selected natural aromatic acids on insulin action and their potential use to overcome insulin resistance. METHODS The 3T3-L1 fibroblasts were differentiated into mature adipocytes; next, insulin resistance was induced by palmitic acid (16:0). Cells were further cultured with phenophospholipids at appropriate concentrations. To assess insulin sensitivity, we measured the insulin-stimulated glucose uptake, using a glucose uptake test. RESULTS We showed that cinnamic acid (CA) and 3-methoxycinnamic acid (3-OMe-CA) restored the proper insulin response. However, 1,2-dicinnamoyl-sn-glycero-3-phosphocholine (1,2-diCA-PC) and 1-cinnamoyl-2-palmitoyl-sn-glycero-3-phosphocholine (1-CA-2-PA-PC) improved insulin sensitivity in insulin-resistant adipocytes even stronger, exhibiting more beneficial effects. CONCLUSIONS The binding of aromatic acids to phosphatidylcholine increases their beneficial effect on insulin sensitivity in adipocytes and expands their potential practical application as nutraceutical health-promoting agents.
Collapse
Affiliation(s)
- Małgorzata Małodobra-Mazur
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, Sklodowskiej-Curie 52, 50-369 Wrocław, Poland; (D.L.); (A.C.)
| | - Dominika Lewoń
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, Sklodowskiej-Curie 52, 50-369 Wrocław, Poland; (D.L.); (A.C.)
| | - Aneta Cierzniak
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, Sklodowskiej-Curie 52, 50-369 Wrocław, Poland; (D.L.); (A.C.)
| | - Marta Okulus
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375 Wrocław, Poland;
| | - Anna Gliszczyńska
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375 Wrocław, Poland;
| |
Collapse
|
34
|
Rezazadeh H, Sharifi MR, Soltani N. Insulin resistance and the role of gamma-aminobutyric acid. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2021; 26:39. [PMID: 34484371 PMCID: PMC8384006 DOI: 10.4103/jrms.jrms_374_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 12/09/2020] [Accepted: 02/02/2021] [Indexed: 12/17/2022]
Abstract
Insulin resistance (IR) is mentioned to be a disorder in insulin ability in insulin-target tissues. Skeletal muscle (SkM) and liver function are more affected by IR than other insulin target cells. SkM is the main site for the consumption of ingested glucose. An effective treatment for IR has two properties: An inhibition of β-cell death and a promotion of β-cell replication. Gamma-aminobutyric acid (GABA) can improve beta-cell mass and function. Multiple studies have shown that GABA decreases IR probably via increase in glucose transporter 4 (GLUT4) gene expression and prevention of gluconeogenesis pathway in the liver. This review focused on the general aspects of IR in skeletal muscle (SkM), liver; the cellular mechanism(s) lead to the development of IR in these organs, and the role of GABA to reduce insulin resistance.
Collapse
Affiliation(s)
- Hossein Rezazadeh
- Department of Physiology, School of Medicine, Isfahan University of Medical Science, Isfahan Iran
| | - Mohammad Reza Sharifi
- Department of Physiology, School of Medicine, Isfahan University of Medical Science, Isfahan Iran
| | - Nepton Soltani
- Department of Physiology, School of Medicine, Isfahan University of Medical Science, Isfahan Iran
| |
Collapse
|
35
|
Zhang YY, Gu LJ, Zhu N, Wang L, Cai MC, Jia JS, Rong S, Yuan WJ. Calpain 6 inhibits autophagy in inflammatory environments: A preliminary study on myoblasts and a chronic kidney disease rat model. Int J Mol Med 2021; 48:194. [PMID: 34435644 PMCID: PMC8416137 DOI: 10.3892/ijmm.2021.5027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/30/2021] [Indexed: 02/06/2023] Open
Abstract
A non-classical calpain, calpain 6 (CAPN6), can inhibit skeletal muscle differentiation and regeneration. In the present study, the role of CAPN6 in the regulation of the autophagy of myoblasts in vitro was investigated. The underlying molecular events and the CAPN6 level in atrophic skeletal muscle in a rat model of chronic kidney disease (CKD) were also investigated. In vitro, CAPN6 was overexpressed, or knocked down, in rat L6 myoblasts to assess autophagy and related gene expression and co-localization. Subsequently, myoblasts were treated with a mixture of cytokines, and relative gene expression and autophagy were assessed. A rat model of CKD for muscle atrophy was established, and blood chemical level and the expression of CAPN6 in muscle were assessed. The data revealed that the knockdown of CAPN6 in rat myoblasts resulted in increased microtubule-associated protein 1 light chain 3 (LC3) levels, while its overexpression decreased LC3 levels and impaired autophagy. Additionally, it was observed that the co-localization of mammalian target of rapamycin (mTOR) and lysosomal-associated membrane protein 1 (LAMP1), a lysosomal marker, proteins was increased. In addition, mTOR, Raptor and α-tubulin (a marker of microtubules) increased in the CAPN6 overexpression group. However, inflammatory cytokines, such as interleukin (IL)-6, tumor necrosis factor (TNF)-α, interferon (INF)-γ and lipopolysaccharides upregulated CAPN6 expression, inhibited L6 myoblast autophagy and stabilized mTOR activity. Furthermore, the animal model successfully mimicked human disease as regards an increase in body weight, and a reduction in muscle mass, cross-sectional area and blood biomarker concentrations; a slight increase in CAPN6 mRNA and protein levels in muscles was observed. Finally, the data of the present study suggested that CAPN6 reduced autophagy via the maintenance of mTOR signaling, which may play a role in CKD-related muscle atrophy. However, future studies are required to determine whether CAPN6 may be used as an intervention target for CKD-related skeletal muscle atrophy.
Collapse
Affiliation(s)
- Yue Yue Zhang
- Division of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Li Jie Gu
- Division of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Nan Zhu
- Division of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Ling Wang
- Division of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Min Chao Cai
- Division of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Jie Shuang Jia
- Division of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Shu Rong
- Division of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Wei Jie Yuan
- Division of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| |
Collapse
|
36
|
Shiomi K, Saito C, Nagai K, Kosaki K, Kawamura T, Kaneko S, Kai H, Morito N, Usui J, Yanagi H, Yamagata K. Ratio of serum creatinine to cystatin C is related to leg strength in predialysis CKD patients. Clin Exp Nephrol 2021; 25:1079-1086. [PMID: 34342777 DOI: 10.1007/s10157-021-02050-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 03/08/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND OBJECTIVES Chronic kidney disease (CKD) patients have lower levels of physical function. Especially, leg strength is important for daily living and preventing falls. However, physical function screenings are difficult to perform at clinical sites. To find clinically useful method to evaluate physical function in predialysis CKD patients, we tried to evaluate the relationship between the ratio of serum creatinine to serum cystatin C (Cre/CysC), and knee extensor muscle strength/body weight (KEMS) which reflects their leg strength. DESIGN, SETTING, PARTICIPANTS, AND MEASUREMENTS We recruited 147 outpatients with CKD (87 men; mean age, 61.6 ± 9.8 years; mean eGFRcreat, 40.7 ± 12.9 mL/min/1.73m2) in this cross-sectional study. KEMS was assessed using a wire strain gauge dynamometer. Skeletal muscle mass and body fat mass were assessed by bioelectrical impedance analysis. RESULTS The mean value of Cre/CysC was 1.01 ± 0.18. The mean value of KEMS was 1.60 ± 0.47 Nm/kg. In multivariate linear regression analysis, skeletal muscle mass (p < 0.01), body fat mass (p < 0.01), hemoglobin (p = 0.01), and Cre/CysC (p < 0.01) was independently related to KEMS. The correlation between Cre/CysC and KEMS is stronger in high quantile of Cre/CysC. CONCLUSIONS In predialysis CKD patients, KEMS showed lower as CKD stage advanced. Cre/CysC is significantly related to KEMS independently. Cre/CysC may be an alternative marker for leg strength in CKD patients and even more valuable to utilize in cases with high Cre/CysC.
Collapse
Affiliation(s)
- Kohei Shiomi
- Degree Programs in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Chie Saito
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kei Nagai
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Keisei Kosaki
- Faculty of Sports Science, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Tetsuya Kawamura
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Shuzo Kaneko
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hirayasu Kai
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Naoki Morito
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Joichi Usui
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hisako Yanagi
- Department of Medical Science and Welfare, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kunihiro Yamagata
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
37
|
Cha RH, Lee GS, Yoo JY, Rhee OB, Jeon YD. Hand Grip and Leg Muscle Strength in Hemodialysis Patients and Its Determinants. J Korean Med Sci 2021; 36:e76. [PMID: 33754509 PMCID: PMC7985287 DOI: 10.3346/jkms.2021.36.e76] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/10/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Chronic kidney disease is associated with chronic inflammation and progressive loss of peripheral muscle strength and the ability to exercise, and these changes are highly pronounced in patients receiving hemodialysis (HD). We evaluated hand grip strength (HGS) and leg muscle strength (LMS) in patients receiving HD and attempted to identify factors associated with muscle strength. METHODS We screened HGS (opposite the fistula side) and LMS (both sides) in HD patients at a single center (n = 112) by using digital hand and leg dynamometers (T.K.K. 5401 and 5710e/5715, Takei Scientific Instruments Co. Ltd., Niigata, Japan). RESULTS The mean age of patients was 62.6 years, and 73.2% of the patients were male. Diabetes was the cause of kidney failure in 50% of the patients, and the median HD vintage was 34 months. A total of 77.7% of patients reported that they participated in regular home-based exercise, and 29.5% of patients regularly participated in hospital-based resistance exercise. HGS and LMS showed good correlation (r = 0.715, P < 0.001). HGS (25.1 vs. 17.0 kg) and LMS (30.1 vs. 20.4 kg) were greater in males (P < 0.001 and P < 0.001, respectively) than in females. Older patients (≥ 60 years) showed less LMS than younger patients in both males and females (P = 0.012 and P = 0.037, respectively), but HGS did not differ according to age. Patients performing regular home- or hospital-based exercise showed higher HGS than those who did not exercise (24.2 vs. 18.6 kg, P = 0.011), but LMS was not significantly different (29.3 vs. 23.6 kg, P = 0.185). Multiple linear regression analysis proved that male sex, younger age, and any type of exercise were factors associated with improved HGS and LMS. Groups of older age (≥ 60 years), male sex, and shorter duration of HD (< median) benefitted more from exercise. CONCLUSION Sex, age, and exercise were the most important determinants of muscle strength in HD patients. We need to encourage patients to engage in regular home or group exercise from the beginning of dialysis and introduce new feasible forms of exercise for HD patients.
Collapse
Affiliation(s)
- Ran Hui Cha
- Department of Internal Medicine, National Medical Center, Seoul, Korea.
| | - Geum Sil Lee
- Department of Internal Medicine, National Medical Center, Seoul, Korea
| | - Ju Yeon Yoo
- Department of Internal Medicine, National Medical Center, Seoul, Korea
| | - Oe Bog Rhee
- Department of Internal Medicine, National Medical Center, Seoul, Korea
| | - Yong Duk Jeon
- Department of Internal Medicine, National Medical Center, Seoul, Korea
| |
Collapse
|
38
|
Genders AJ, Marin EC, Bass JJ, Kuang J, Saner NJ, Smith K, Atherton PJ, Bishop DJ. Ammonium chloride administration prior to exercise has muscle-specific effects on mitochondrial and myofibrillar protein synthesis in rats. Physiol Rep 2021; 9:e14797. [PMID: 33769716 PMCID: PMC7995552 DOI: 10.14814/phy2.14797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 02/01/2021] [Accepted: 02/14/2021] [Indexed: 12/04/2022] Open
Abstract
AIM Exercise is able to increase both muscle protein synthesis and mitochondrial biogenesis. However, acidosis, which can occur in pathological states as well as during high-intensity exercise, can decrease mitochondrial function, whilst its impact on muscle protein synthesis is disputed. Thus, the aim of this study was to determine the effect of a mild physiological decrease in pH, by administration of ammonium chloride, on myofibrillar and mitochondrial protein synthesis, as well as associated molecular signaling events. METHODS Male Wistar rats were given either a placebo or ammonium chloride prior to a short interval training session. Rats were killed before exercise, immediately after exercise, or 3 h after exercise. RESULTS Myofibrillar (p = 0.036) fractional protein synthesis rates was increased immediately after exercise in the soleus muscle of the placebo group, but this effect was absent in the ammonium chloride group. However, in the gastrocnemius muscle NH4 Cl increased myofibrillar (p = 0.044) and mitochondrial protein synthesis (0 h after exercise p = 0.01; 3 h after exercise p = 0.003). This was accompanied by some small differences in protein phosphorylation and mRNA expression. CONCLUSION This study found ammonium chloride administration immediately prior to a single session of exercise in rats had differing effects on mitochondrial and myofibrillar protein synthesis rates in soleus (type I) and gastrocnemius (type II) muscle in rats.
Collapse
Affiliation(s)
- Amanda J. Genders
- Institute for Health and Sport (iHeS)Victoria UniversityMelbourneVictoriaAustralia
| | - Evelyn C. Marin
- Institute for Health and Sport (iHeS)Victoria UniversityMelbourneVictoriaAustralia
- Department of Medicine (Austin Health)The University of MelbourneMelbourneVictoriaAustralia
| | - Joseph J. Bass
- MRC/ARUK Centre for Musculoskeletal Ageing ResearchNottingham Biomedical Research Centre (BRC)National Institute for Health Research (NIHR)School of MedicineUniversity of NottinghamNottinghamUK
| | - Jujiao Kuang
- Institute for Health and Sport (iHeS)Victoria UniversityMelbourneVictoriaAustralia
| | - Nicholas J. Saner
- Institute for Health and Sport (iHeS)Victoria UniversityMelbourneVictoriaAustralia
| | - Ken Smith
- MRC/ARUK Centre for Musculoskeletal Ageing ResearchNottingham Biomedical Research Centre (BRC)National Institute for Health Research (NIHR)School of MedicineUniversity of NottinghamNottinghamUK
| | - Philip J. Atherton
- MRC/ARUK Centre for Musculoskeletal Ageing ResearchNottingham Biomedical Research Centre (BRC)National Institute for Health Research (NIHR)School of MedicineUniversity of NottinghamNottinghamUK
| | - David J. Bishop
- Institute for Health and Sport (iHeS)Victoria UniversityMelbourneVictoriaAustralia
| |
Collapse
|
39
|
Uchiyama K, Wakino S, Irie J, Miyamoto J, Matsui A, Tajima T, Itoh T, Oshima Y, Yoshifuji A, Kimura I, Itoh H. Contribution of uremic dysbiosis to insulin resistance and sarcopenia. Nephrol Dial Transplant 2021; 35:1501-1517. [PMID: 32535631 DOI: 10.1093/ndt/gfaa076] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 03/14/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) leads to insulin resistance (IR) and sarcopenia, which are associated with a high mortality risk in CKD patients; however, their pathophysiologies remain unclear. Recently, alterations in gut microbiota have been reported to be associated with CKD. We aimed to determine whether uremic dysbiosis contributes to CKD-associated IR and sarcopenia. METHODS CKD was induced in specific pathogen-free mice via an adenine-containing diet; control animals were fed a normal diet. Fecal microbiota transplantation (FMT) was performed by oral gavage in healthy germ-free mice using cecal bacterial samples obtained from either control mice (control-FMT) or CKD mice (CKD-FMT). Vehicle mice were gavaged with sterile phosphate-buffered saline. Two weeks after inoculation, mice phenotypes, including IR and sarcopenia, were evaluated. RESULTS IR and sarcopenia were evident in CKD mice compared with control mice. These features were reproduced in CKD-FMT mice compared with control-FMT and vehicle mice with attenuated insulin-induced signal transduction and mitochondrial dysfunction in skeletal muscles. Intestinal tight junction protein expression and adipocyte sizes were lower in CKD-FMT mice than in control-FMT mice. Furthermore, CKD-FMT mice showed systemic microinflammation, increased concentrations of serum uremic solutes, fecal bacterial fermentation products and elevated lipid content in skeletal muscle. The differences in gut microbiota between CKD and control mice were mostly consistent between CKD-FMT and control-FMT mice. CONCLUSIONS Uremic dysbiosis induces IR and sarcopenia, leaky gut and lipodystrophy.
Collapse
Affiliation(s)
- Kiyotaka Uchiyama
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, Tokyo, Japan
| | - Shu Wakino
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, Tokyo, Japan
| | - Junichiro Irie
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Junki Miyamoto
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ayumi Matsui
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, Tokyo, Japan
| | - Takaya Tajima
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, Tokyo, Japan
| | - Tomoaki Itoh
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, Tokyo, Japan
| | - Yoichi Oshima
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, Tokyo, Japan
| | - Ayumi Yoshifuji
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Ikuo Kimura
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hiroshi Itoh
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
40
|
Association between the uremic toxins indoxyl-sulfate and p-cresyl-sulfate with sarcopenia and malnutrition in elderly patients with advanced chronic kidney disease. Exp Gerontol 2021; 147:111266. [PMID: 33529747 DOI: 10.1016/j.exger.2021.111266] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/04/2021] [Accepted: 01/29/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND in patients with chronic kidney disease (CKD) indoxyl sulfate (IS) and p-cresyl sulfate (PCs) may induce sarcopenia either directly or via systemic inflammation. We evaluated whether IS and PCs were associated with: sarcopenia, systemic inflammation and nutritional status. METHODS we examined cross sectionally 93 patients with advanced CKD. Sarcopenia was identified according to EWGSOP2 definition. Malnutrition was assessed by Malnutrition Inflammation Score (MIS) and Protein Energy Wasting syndrome (PEW). Inflammatory status was assessed by dosing: CRP, IL6, TNFα, MCP1, IL10, IL17, IL12p70. RESULTS we did not find any association of sarcopenia with IS and PCs. IS was associated with LogTNFα and LogMCP-1 in the overall cohort (r = 0.30, p = 0.0043; r = 0.22 p = 0.047) and in not sarcopenic patients (r = 0.32, p = 0.0077; r = 0.25, p = 0.041). PCs was associated with LogIL10 and LogIL12p70 in sarcopenic patients (r = 0.58, p = 0.0042; r = 0.52, p = 0.013). IS was higher in patients without PEW (p = 0.029), while PCs was higher in patients with PEW (p = 0.0040). IS and PCs were not different in patients with normal or increased MIS. CONCLUSIONS IS and PCs were not associated with sarcopenia, although they were both associated with some inflammatory pathways. Notably, we found a positive association of PCs with PEW syndrome.
Collapse
|
41
|
Li J, Liu D, Liu Z. Serum Total Bilirubin and Progression of Chronic Kidney Disease and Mortality: A Systematic Review and Meta-Analysis. Front Med (Lausanne) 2021; 7:549. [PMID: 33569386 PMCID: PMC7868400 DOI: 10.3389/fmed.2020.00549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/31/2020] [Indexed: 12/04/2022] Open
Abstract
Background: Previous studies have suggested that serum total bilirubin (STB) levels are associated with heightened chronic kidney disease (CKD) and mortality in both the general population and nephropathy patients. However, these results remain inconsistent. The aim of our study was to investigate whether STB was a predictor for progression of CKD and mortality by meta-analysis. Methods: We performed a systematic literature search in PubMed, Web of Science, MEDLINE, EMBASE, Google Scholar, and Cochrane Library's database up to June 30, 2019. Pooled risk ratios (RR) and corresponding 95% confidence intervals (CI) were extracted for the highest vs. lowest category STB levels within the physiological range, and a random-effects model was applied to calculate the dose–response relationships. A pooled hazard ratio (HR) was used to investigate the association between STB levels and mortality in dialysis patients. Results: A total of 16 studies, wherein participants were followed from 21 months to 7 years, were eligible for inclusion in the study. For the categorized STB, 11 studies with 41,188 participants were identified and analyzed. Patients with the highest STB levels were associated with a lower risk of CKD (RR = 0.64; 95% CI 0.55–0.73) compared to those with the lowest STB levels. Furthermore, based on seven studies, a pooled RR of 0.89, 95% CI (0.80–0.99) was observed for the continuous STB levels (per 0.2 mg/dL increase). Four studies that included 51,764 participants illustrated that there was no association between STB levels and all-cause mortality (HR = 0.77; 95% CI 0.42–1.41). A prominent negative linear relationship (X2 = 14.70; P = 0.0001) was found between STB levels and risk of CKD. Subgroup analyses showed that there were no significant differences in the subgroup adjustment factor except for sample size. Conclusions: Elevated STB levels within a physiological range are associated with lower risk of CKD regardless of the study characteristics and coincide with a liner dose–response relationship. However, whether high STB levels are a protective factor against mortality remains inconclusive. Large-scale randomized controlled trails are needed to target STB levels for predicting renal outcomes.
Collapse
Affiliation(s)
- Jia Li
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China.,Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, China
| | - Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China.,Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China.,Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, China
| |
Collapse
|
42
|
Thome T, Kumar RA, Burke SK, Khattri RB, Salyers ZR, Kelley RC, Coleman MD, Christou DD, Hepple RT, Scali ST, Ferreira LF, Ryan TE. Impaired muscle mitochondrial energetics is associated with uremic metabolite accumulation in chronic kidney disease. JCI Insight 2020; 6:139826. [PMID: 33290279 PMCID: PMC7821598 DOI: 10.1172/jci.insight.139826] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 11/25/2020] [Indexed: 01/01/2023] Open
Abstract
Chronic kidney disease (CKD) causes progressive skeletal myopathy involving atrophy, weakness, and fatigue. Mitochondria have been thought to contribute to skeletal myopathy; however, the molecular mechanisms underlying muscle metabolism changes in CKD are unknown. We employed a comprehensive mitochondrial phenotyping platform to elucidate the mechanisms of skeletal muscle mitochondrial impairment in mice with adenine-induced CKD. CKD mice displayed significant reductions in mitochondrial oxidative phosphorylation (OXPHOS), which was strongly correlated with glomerular filtration rate, suggesting a link between kidney function and muscle mitochondrial health. Biochemical assays uncovered that OXPHOS dysfunction was driven by reduced activity of matrix dehydrogenases. Untargeted metabolomics analyses in skeletal muscle revealed a distinct metabolite profile in CKD muscle including accumulation of uremic toxins that strongly associated with the degree of mitochondrial impairment. Additional muscle phenotyping found CKD mice experienced muscle atrophy and increased muscle protein degradation, but only male CKD mice had lower maximal contractile force. CKD mice had morphological changes indicative of destabilization in the neuromuscular junction. This study provides the first comprehensive evaluation of mitochondrial health in murine CKD muscle to our knowledge and uncovers several unknown uremic metabolites that strongly associate with the degree of mitochondrial impairment.
Collapse
Affiliation(s)
- Trace Thome
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Ravi A Kumar
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Sarah K Burke
- Department of Physical Therapy, College of Public Health and Health Professions
| | - Ram B Khattri
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Zachary R Salyers
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Rachel C Kelley
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Madeline D Coleman
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance
| | - Demetra D Christou
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance.,Center for Exercise Science, College of Health & Human Performance, and
| | - Russell T Hepple
- Department of Physical Therapy, College of Public Health and Health Professions
| | - Salvatore T Scali
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida, USA.,Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida, USA
| | - Leonardo F Ferreira
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance.,Center for Exercise Science, College of Health & Human Performance, and
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, College of Health & Human Performance.,Center for Exercise Science, College of Health & Human Performance, and
| |
Collapse
|
43
|
Draicchio F, van Vliet S, Ancu O, Paluska SA, Wilund KR, Mickute M, Sathyapalan T, Renshaw D, Watt P, Sylow L, Burd NA, Mackenzie RW. Integrin-associated ILK and PINCH1 protein content are reduced in skeletal muscle of maintenance haemodialysis patients. J Physiol 2020; 598:5701-5716. [PMID: 32969494 DOI: 10.1113/jp280441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/09/2020] [Indexed: 12/17/2022] Open
Abstract
KEY POINTS Patients with renal failure undergoing maintenance haemodialysis are associated with insulin resistance and protein metabolism dysfunction. Novel research suggests that disruption to the transmembrane protein linkage between the cytoskeleton and the extracellular matrix in skeletal muscle may contribute to reduced amino acid metabolism and insulin resistance in haemodialysis. ILK, PINCH1 and pFAKTyr397 were significantly decreased in haemodialysis compared to controls, whereas Rac1 and Akt2 showed no different between groups. Rac1 deletion in the Rac1 knockout model did not alter the expression of integrin-associated proteins. Phenylalanine kinetics were reduced in the haemodialysis group at 30 and 60 min post meal ingestion compared to controls; both groups showed similar levels of insulin sensitivity and β-cell function. Key proteins in the integrin-cytoskeleton linkage are reduced in haemodialysis patients, suggesting for the first time that integrin-associated proteins dysfunction may contribute to reduced phenylalanine flux without affecting insulin resistance in haemodialysis patients. ABSTRACT Muscle atrophy, insulin resistance and reduced muscle phosphoinositide 3-kinase-Akt signalling are common characteristics of patients undergoing maintenance haemodialysis (MHD). Disruption to the transmembrane protein linkage between the cytoskeleton and the extracellular matrix in skeletal muscle may contribute to reduced amino acid metabolism and insulin resistance in MHD patients. Eight MHD patients (age: 56 ± 5 years: body mass index: 32 ± 2 kg m-2 ) and non-diseased controls (age: 50 ± 2 years: body mass index: 31 ± 1 kg m-2 ) received primed continuous l-[ring-2 H5 ]phenylalanine before consuming a mixed meal. Phenylalanine metabolism was determined using two-compartment modelling. Muscle biopsies were collected prior to the meal and at 300 min postprandially. In a separate experiment, skeletal muscle tissue from muscle-specific Rac1 knockout (Rac1 mKO) was harvested to investigate whether Rac1 depletion disrupted the cytoskeleton-integrin linkage, allowing for cross-model examination of proteins of interest. ILK, PINCH1 and pFAKTyr397 were significantly lower in MHD (P < 0.01). Rac1 and Akt showed no difference between groups for the human trial. Rac1 deletion in the Rac1 mKO model did not alter the expression of integrin-associated proteins. Phenylalanine rates of appearance and disappearance, as well as metabolic clearance rates, were lower in the MHD group at 30 and 60 min post meal ingestion compared to controls (P < 0.05). Both groups showed similar levels of insulin sensitivity and β-cell function. Key proteins in the integrin-cytoskeleton linkage are reduced in MHD patients, suggesting for the first time that integrin-associated proteins dysfunction may contribute to reduced phenylalanine flux without affecting insulin resistance in haemodialysis patients.
Collapse
Affiliation(s)
- Fulvia Draicchio
- Department of Life Sciences, Sport and Exercise Science Research Center, University of Roehampton, London, UK
| | - Stephan van Vliet
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Oana Ancu
- Department of Life Sciences, Sport and Exercise Science Research Center, University of Roehampton, London, UK
| | - Scott A Paluska
- Department of Family Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Kenneth R Wilund
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Monika Mickute
- Leicester Diabetes Center, Leicester General Hospital, Leicester, UK
| | | | - Derek Renshaw
- Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry, UK
| | - Peter Watt
- Sport and Exercise Science and Sports Medicine research and enterprise group, Welkin Laboratories, University of Brighton, Eastbourne, UK
| | - Lykke Sylow
- Department of Nutrition, Exercise and Sport, August Krogh Bygningen, University of Copenhagen, Denmark
| | - Nicholas A Burd
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Richard Wa Mackenzie
- Department of Life Sciences, Sport and Exercise Science Research Center, University of Roehampton, London, UK
| |
Collapse
|
44
|
Posa DK, Baba SP. Intracellular pH Regulation of Skeletal Muscle in the Milieu of Insulin Signaling. Nutrients 2020; 12:nu12102910. [PMID: 32977552 PMCID: PMC7598285 DOI: 10.3390/nu12102910] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 12/18/2022] Open
Abstract
Type 2 diabetes (T2D), along with obesity, is one of the leading health problems in the world which causes other systemic diseases, such as cardiovascular diseases and kidney failure. Impairments in glycemic control and insulin resistance plays a pivotal role in the development of diabetes and its complications. Since skeletal muscle constitutes a significant tissue mass of the body, insulin resistance within the muscle is considered to initiate the onset of diet-induced metabolic syndrome. Insulin resistance is associated with impaired glucose uptake, resulting from defective post-receptor insulin responses, decreased glucose transport, impaired glucose phosphorylation, oxidation and glycogen synthesis in the muscle. Although defects in the insulin signaling pathway have been widely studied, the effects of cellular mechanisms activated during metabolic syndrome that cross-talk with insulin responses are not fully elucidated. Numerous reports suggest that pathways such as inflammation, lipid peroxidation products, acidosis and autophagy could cross-talk with insulin-signaling pathway and contribute to diminished insulin responses. Here, we review and discuss the literature about the defects in glycolytic pathway, shift in glucose utilization toward anaerobic glycolysis and change in intracellular pH [pH]i within the skeletal muscle and their contribution towards insulin resistance. We will discuss whether the derangements in pathways, which maintain [pH]i within the skeletal muscle, such as transporters (monocarboxylate transporters 1 and 4) and depletion of intracellular buffers, such as histidyl dipeptides, could lead to decrease in [pH]i and the onset of insulin resistance. Further we will discuss, whether the changes in [pH]i within the skeletal muscle of patients with T2D, could enhance the formation of protein aggregates and activate autophagy. Understanding the mechanisms by which changes in the glycolytic pathway and [pH]i within the muscle, contribute to insulin resistance might help explain the onset of obesity-linked metabolic syndrome. Finally, we will conclude whether correcting the pathways which maintain [pH]i within the skeletal muscle could, in turn, be effective to maintain or restore insulin responses during metabolic syndrome.
Collapse
Affiliation(s)
- Dheeraj Kumar Posa
- Diabetes and Obesity Center, University of Louisville, Louisville, KY 40202, USA
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA
| | - Shahid P Baba
- Diabetes and Obesity Center, University of Louisville, Louisville, KY 40202, USA
- Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
45
|
Zhang L, Chen Q, Chen Z, Wang Y, Gamboa JL, Ikizler TA, Garibotto G, Mitch WE. Mechanisms Regulating Muscle Protein Synthesis in CKD. J Am Soc Nephrol 2020; 31:2573-2587. [PMID: 32764136 DOI: 10.1681/asn.2019121277] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 06/21/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND CKD induces loss of muscle proteins partly by suppressing muscle protein synthesis. Muscles of mice with CKD have increased expression of nucleolar protein 66 (NO66), as do muscle biopsy specimens from patients with CKD or those undergoing hemodialysis. Inflammation stimulates NO66 expression and changes in NF-κB mediate the response. METHODS Subtotal nephrectomy created a mouse model of CKD with BUN >80 mg/dl. Crossing NO66flox/flox with MCK-Cre mice bred muscle-specific NO66 (MCK-NO66) knockout mice. Experiments assessed the effect of removing NO66. RESULTS Muscle-specific NO66 knockout in mice blocks CKD-induced loss of muscle mass and improves protein synthesis. NO66 suppression of ribosomal biogenesis via demethylase activity is the mechanism behind these responses. In muscle cells, expression of NO66, but not of demethylase-dead mutant NO66, decreased H3K4me3 and H3K36me3 and suppressed pre-rRNA expression. Knocking out NO66 increased the enrichment of H3K4me3 and H3K36me3 on ribosomal DNA. In primary muscle cells and in muscles of mice without NO66, ribosomal RNA, pre-rRNA, and protein synthesis all increased. CONCLUSIONS CKD suppresses muscle protein synthesis via epigenetic mechanisms that NO66 mediates. Blocking NO66 could suggest strategies that counter CKD-induced abnormal muscle protein catabolism.
Collapse
Affiliation(s)
- Liping Zhang
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Qin Chen
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zihong Chen
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Ying Wang
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Jorge L Gamboa
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Talat Alp Ikizler
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Giacomo Garibotto
- Nephrology Division, Department of Internal Medicine, Genoa University, Scientific Hospitalization and Treatment Institute Policlinico San Martino Hospital, Genoa, Italy
| | - William E Mitch
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
46
|
Zhang L, Wang Y, Dong Y, Chen Z, Eckols TK, Kasembeli MM, Tweardy DJ, Mitch WE. Pharmacokinetics and pharmacodynamics of TTI-101, a STAT3 inhibitor that blocks muscle proteolysis in rats with chronic kidney disease. Am J Physiol Renal Physiol 2020; 319:F84-F92. [PMID: 32475130 DOI: 10.1152/ajprenal.00603.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Loss of muscle proteins increases the morbidity and mortality of patients with chronic kidney disease (CKD), and there are no reliable preventive treatments. We uncovered a STAT3/CCAAT-enhancer-binding protein-δ to myostatin signaling pathway that activates muscle protein degradation in mice with CKD or cancer; we also identified a small-molecule inhibitor of STAT3 (TTI-101) that blocks this pathway. To evaluate TTI-101 as a treatment for CKD-induced cachexia, we measured TTI-101 pharmacokinetics and pharmacodynamics in control and CKD rats that were orally administered TTI-101or its diluent. The following two groups of gavage-fed rats were studied: sham-operated control rats and CKD rats. Plasma was collected serially (0, 0.25, 0.5, 1, 2, 4, 8, and 24 h) following TTI-101 administration (at oral doses of 0, 10, 30, or 100 mg/kg). Plasma levels of TTI-101 were measured by LC-MS/MS, and pharmacokinetic results were analyzed with the PKSolver program. Plasma TTI-101 levels increased linearly with doses; the maximum plasma concentrations and time to maximal plasma levels (~1 h) were similar in sham-operated control rats and CKD rats. Notably, gavage treatment of TTI-101 for 3 days produced TTI-101 muscle levels in sham control rats and CKD rats that were not significantly different. CKD rats that received TTI-101 for 7 days had suppression of activated STAT3 and improved muscle grip strength; there also was a trend for increasing body and muscle weights. TTI-101 was tolerated at doses of 100 mg·kg-1·day-1 for 7 days. These results with TTI-101 in rats warrant its development as a treatment for cachexia in humans.
Collapse
Affiliation(s)
- Liping Zhang
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Ying Wang
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Yanlan Dong
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Zihong Chen
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Thomas K Eckols
- Division of Internal Medicine, Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Moses M Kasembeli
- Division of Internal Medicine, Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David J Tweardy
- Division of Internal Medicine, Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - William E Mitch
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
47
|
Zhang L, Chen Z, Wang Y, Tweardy DJ, Mitch WE. Stat3 activation induces insulin resistance via a muscle-specific E3 ubiquitin ligase Fbxo40. Am J Physiol Endocrinol Metab 2020; 318:E625-E635. [PMID: 32101031 PMCID: PMC7272729 DOI: 10.1152/ajpendo.00480.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cellular mechanisms causing insulin resistance (IR) in chronic kidney disease (CKD) are poorly understood. One potential mechanism is that CKD-induced inflammation activates the signal transducer and activator of transcription 3 (Stat3) in muscle. We uncovered increased p-Stat3 in muscles of mice with CKD or mice fed high-fat diet (HFD). Activated Stat3 stimulates the expression of Fbxo40, a muscle-specific E3 ubiquitin ligase that stimulates ubiquitin conjugation leading to degradation of insulin receptor substrate 1 (IRS1). Evidence that Stat3 activates Fbxo40 includes 1) potential Stat3 binding sites in Fbxo40 promoters; 2) Stat3 binding to the Fbxo40 promoter; and 3) constitutively active Stat3 stimulating both Fbxo40 expression and its promoter activity. We found that IL-6 activates Stat3 in myotubes, increasing Fbxo40 expression with reduced IRS1 and p-Akt. Knockdown Fbxo40 using siRNA from myotubes results in higher levels of IRS1 and p-Akt despite the presence of IL-6. We treated mice with a small-molecule inhibitor of Stat3 (TTI-101) and found improved glucose tolerance and insulin signaling in skeletal muscles of mice with CKD or fed an HFD. Finally, we uncovered improved glucose tolerance in mice with muscle-specific Stat3 KO versus results in Stat3f/f mice in response to the HFD. Thus Stat3 activation in muscle increases IR in mice. Inhibition of Stat3 by TTI-101 could be developed into clinical strategies to improve muscle insulin signaling in inflammation and other catabolic diseases.
Collapse
Affiliation(s)
- Liping Zhang
- Baylor College of Medicine, Department of Medicine, Nephrology Division, Houston, Texas
| | - Zihong Chen
- Baylor College of Medicine, Department of Medicine, Nephrology Division, Houston, Texas
| | - Ying Wang
- Baylor College of Medicine, Department of Medicine, Nephrology Division, Houston, Texas
| | - David J Tweardy
- University of Texas MD Anderson Cancer Center, Division of Internal Medicine, Houston, Texas
- University of Texas MD Anderson Cancer Center, Department of Infectious Diseases, Infection Control and Employee Health, Houston, Texas
- University of Texas MD Anderson Cancer Center, Department of Molecular and Cellular Oncology, Houston, Texas
| | - William E Mitch
- Baylor College of Medicine, Department of Medicine, Nephrology Division, Houston, Texas
| |
Collapse
|
48
|
Garibotto G, Picciotto D, Saio M, Esposito P, Verzola D. Muscle protein turnover and low-protein diets in patients with chronic kidney disease. Nephrol Dial Transplant 2020; 35:741-751. [PMID: 32378720 DOI: 10.1093/ndt/gfaa072] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/28/2020] [Indexed: 02/07/2023] Open
Abstract
Adaptation to a low-protein diet (LPD) involves a reduction in the rate of amino acid (AA) flux and oxidation, leading to more efficient use of dietary AA and reduced ureagenesis. Of note, the concept of 'adaptation' to low-protein intakes has been separated from the concept of 'accommodation', the latter term implying a decrease in protein synthesis, with development of wasting, when dietary protein intake becomes inadequate, i.e. beyond the limits of the adaptive mechanisms. Acidosis, insulin resistance and inflammation are recognized mechanisms that can increase protein degradation and can impair the ability to activate an adaptive response when an LPD is prescribed in a chronic kidney disease (CKD) patient. Current evidence shows that, in the short term, clinically stable patients with CKD Stages 3-5 can efficiently adapt their muscle protein turnover to an LPD containing 0.55-0.6 g protein/kg or a supplemented very-low-protein diet (VLPD) by decreasing muscle protein degradation and increasing the efficiency of muscle protein turnover. Recent long-term randomized clinical trials on supplemented VLPDs in patients with CKD have shown a very good safety profile, suggesting that observations shown by short-term studies on muscle protein turnover can be extrapolated to the long-term period.
Collapse
Affiliation(s)
- Giacomo Garibotto
- Division of Nephrology, Dialysis and Transplantation, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Daniela Picciotto
- Division of Nephrology, Dialysis and Transplantation, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Michela Saio
- Division of Nephrology, Dialysis and Transplantation, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Pasquale Esposito
- Division of Nephrology, Dialysis and Transplantation, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Daniela Verzola
- Division of Nephrology, Dialysis and Transplantation, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| |
Collapse
|
49
|
Kestenbaum B, Gamboa J, Liu S, Ali AS, Shankland E, Jue T, Giulivi C, Smith LR, Himmelfarb J, de Boer IH, Conley K, Roshanravan B. Impaired skeletal muscle mitochondrial bioenergetics and physical performance in chronic kidney disease. JCI Insight 2020; 5:133289. [PMID: 32161192 PMCID: PMC7141399 DOI: 10.1172/jci.insight.133289] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/10/2019] [Indexed: 11/17/2022] Open
Abstract
The maintenance of functional independence is the top priority of patients with chronic kidney disease (CKD). Defects in mitochondrial energetics may compromise physical performance and independence. We investigated associations of the presence and severity of kidney disease with in vivo muscle energetics and the association of muscle energetics with physical performance. We performed measures of in vivo leg and hand muscle mitochondrial capacity (ATPmax) and resting ATP turnover (ATPflux) using 31phosphorus magnetic resonance spectroscopy and oxygen uptake (O2 uptake) by optical spectroscopy in 77 people (53 participants with CKD and 24 controls). We measured physical performance using the 6-minute walk test. Participants with CKD had a median estimated glomerular filtration rate (eGFR) of 33 ml/min per 1.73 m2. Participants with CKD had a -0.19 mM/s lower leg ATPmax compared with controls but no difference in hand ATPmax. Resting O2 uptake was higher in CKD compared with controls, despite no difference in ATPflux. ATPmax correlated with eGFR and serum bicarbonate among participants with GFR <60. ATPmax of the hand and leg correlated with 6-minute walking distance. The presence and severity of CKD associate with muscle mitochondrial capacity. Dysfunction of muscle mitochondrial energetics may contribute to reduced physical performance in CKD.
Collapse
Affiliation(s)
- Bryan Kestenbaum
- Division of Nephrology, Department of Medicine, and
- Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | - Jorge Gamboa
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sophia Liu
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Amir S. Ali
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Eric Shankland
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Thomas Jue
- Department of Biochemistry and Molecular Medicine, School of Medicine
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, and
| | - Lucas R. Smith
- Department of Physical Medicine and Rehabilitation, School of Medicine, UCD, Davis, California, USA
| | - Jonathan Himmelfarb
- Division of Nephrology, Department of Medicine, and
- Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | - Ian H. de Boer
- Division of Nephrology, Department of Medicine, and
- Kidney Research Institute, University of Washington, Seattle, Washington, USA
- Puget Sound Veterans Administration Healthcare System, Seattle, Washington, USA
| | - Kevin Conley
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Baback Roshanravan
- Division of Nephrology, Department of Medicine, School of Medicine, UCD, Sacramento, California, USA
| |
Collapse
|
50
|
Uchiyama K, Washida N, Morimoto K, Muraoka K, Nakayama T, Adachi K, Kasai T, Miyashita K, Wakino S, Itoh H. Effects of exercise on residual renal function in patients undergoing peritoneal dialysis: A post-hoc analysis of a randomized controlled trial. Ther Apher Dial 2020; 24:668-676. [PMID: 31997526 DOI: 10.1111/1744-9987.13481] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/03/2020] [Accepted: 01/27/2020] [Indexed: 12/11/2022]
Abstract
We aimed to investigate the effects of exercise on renal outcomes in patients undergoing peritoneal dialysis (PD). In a post-hoc analysis of a randomized controlled trial of a 12-week home-based exercise program involving 47 patients undergoing PD, we excluded 18 patients with anuria and analyzed 13 and 16 patients in the usual care and exercise groups, respectively. The primary outcomes were weekly renal creatinine clearance (CCr) and urinary biomarkers: liver-type fatty acid-binding protein (L-FABP) and the microalbumin-to-creatinine ratio (ACR). Although the maintenance of weekly renal CCr in the exercise group was not significantly different compared with that in the usual care group (P = .09), urinary L-FABP levels (P = .02) and ACR (P = .04) were significantly decreased in the exercise group. To the best of our knowledge, this is the first study to demonstrate the beneficial effects of exercise on renal outcomes in patients undergoing PD.
Collapse
Affiliation(s)
- Kiyotaka Uchiyama
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Naoki Washida
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.,Department of Nephrology, International University of Health and Welfare School of Medicine, Chiba, Japan
| | - Kohkichi Morimoto
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kaori Muraoka
- Department of Rehabilitation Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takashin Nakayama
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Keika Adachi
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takahiro Kasai
- Department of Nephrology, International University of Health and Welfare School of Medicine, Chiba, Japan
| | - Kazutoshi Miyashita
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shu Wakino
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Itoh
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|