1
|
Chu C, Huang Y, Cao L, Ji S, Zhu B, Shen Q. Role of macrophages in peritoneal dialysis-associated peritoneal fibrosis. Ren Fail 2025; 47:2474203. [PMID: 40044628 PMCID: PMC11884102 DOI: 10.1080/0886022x.2025.2474203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
Peritoneal dialysis (PD) can be used as renal replacement therapy when chronic kidney disease (CKD) progresses to end-stage renal disease. However, peritoneal fibrosis (PF) is a major cause of PD failure. Studies have demonstrated that PD fluid contains a significantly larger numbers of macrophages compared with the healthy individuals. During PD, macrophages can secrete cytokines to keep peritoneal tissue in sustained low-grade inflammation, and participate in the regulation of fibrosis-related signaling pathways, such as NF-κB, TGF-β/Smad, IL4/STAT6, and PI3K/AKT. A series of basic pathological changes occurs in peritoneal tissues, including epithelial mesenchymal transformation, overgeneration of neovasculature, and abnormal deposition of extracellular matrix. This review focuses on the role of macrophages in promoting PF during PD, summarizes the targets of macrophage-related inhibition of fibrosis, and provides new ideas for clinical research on delaying PF, maintaining the function and integrity of peritoneum, prolonging duration of PD as a renal replacement modality, and achieving longer survival in CKD patients.
Collapse
Affiliation(s)
- Chenling Chu
- Department of Clinical Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Ying Huang
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
- Department of Public Health and Preventive Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Luxi Cao
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Shuiyu Ji
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Bin Zhu
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Quanquan Shen
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
- Department of Nephrology, Zhejiang Provincial People’s Hospital Bijie Hospital, Bijie, Guizhou, China
| |
Collapse
|
2
|
Pan X, Zhang R, Lu B, Chen S, Chen H, Li M, Qin L, Song Z, Yang Y, Wang Z, Yan F, Sun A, Wu F, Zhang L, Wang J, Guzik TJ, Gao P. SM22α-Lineage Perivascular Stromal Cells Contribute to Abdominal Aortic Aneurysm. Circ Res 2025. [PMID: 40371535 DOI: 10.1161/circresaha.124.325750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 04/24/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Perivascular adipose tissue (PVAT) is a key regulator of vascular dysfunction. Impairment of PVAT phenotypic plasticity with aging may play a role in vascular pathology including abdominal aortic aneurysms (AAAs). Yet, the mechanisms underlying PVAT plasticity in aneurysm pathogenesis remain elusive. METHODS Single-cell RNA sequencing was performed on perivascular stromal cells from young (2- to 3-month-old) and aged (18- to 20-month-old) mice. The expression of PGC-1α (peroxisome proliferator-activated receptor gamma coactivator-1α) was measured in PVAT of aged mice and human aneurysm samples. Loss- and gain-of-function approaches were used to investigate the role of SM22α-lineage perivascular stromal cell-derived PGC-1α in aneurysm development. Molecular mechanisms were explored through transcriptome and functional studies in young and aged mice, SM22αCre; Rosa26RFP/+; PGC1αf/f and SM22αCre; Rosa26RFP/+ mice with Ang II (angiotensin II)-induced and deoxycorticosterone acetate/salt-induced AAA models. RESULTS SM22α+ cells accumulated in PVAT of Ang II-treated aged mice and patients with aortic aneurysms. Single-cell RNA sequencing analysis revealed that aging disrupted the differentiation potential of SM22α-lineage perivascular stromal cells and led to reduced PGC-1α levels. PGC1α downregulation in PVAT was observed in both mouse AAA models and human aneurysm lesions. In mice with SM22α-driven PGC-1α deletion, Ang II-induced AAA formation was accompanied by perivascular stromal cell-to-myofibroblast differentiation. In vitro PGC1α knockdown suppressed nuclear YAP (Yes-associated protein) signaling, reducing adipocyte differentiation, while increasing MMP2 (matrix metalloproteinase 2)-secreting myofibroblasts. Furthermore, PGC-1α overexpression in aged mice or administration of the YAP signaling inhibitor verteporfin in SM22αCre; Rosa26RFP/+; PGC1αf/f mice restored PVAT function and conferred protection against aneurysm formation. Last, we used the radiomics analysis to noninvasively evaluate PVAT in the context of AAA severity in humans. CONCLUSIONS PGC-1α deficiency in SM22α-lineage stromal cells disrupts the balance between adipogenic and myofibrogenic differentiation through regulating YAP signaling, ultimately promoting aneurysm development. Radiomics assessment may present a promising noninvasive approach for PVAT evaluation in aneurysms, offering valuable potential for clinical research.
Collapse
Affiliation(s)
- Xiaoxi Pan
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Run Zhang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Bingling Lu
- School of Biomedical Engineering, Shanghai Jiao Tong University, China (B.L., Z.S., L.Z.)
| | - Siyuan Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Hongjin Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Mengyao Li
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Le Qin
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (L.Q., F.Y.)
| | - Zhiyun Song
- School of Biomedical Engineering, Shanghai Jiao Tong University, China (B.L., Z.S., L.Z.)
| | - Yi Yang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (Y.Y., Z.W.)
| | - Zhe Wang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (Y.Y., Z.W.)
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (L.Q., F.Y.)
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China (A.S.)
| | - Fang Wu
- Shanghai Clinical Research Center for Aging and Medicine, Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (F.W.)
| | - Lichi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, China (B.L., Z.S., L.Z.)
| | - Jiguang Wang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Tomasz J Guzik
- Department of Medicine, Jagiellonian University, Colelgium Medicum, Krakow, Poland (T.J.G.)
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, United Kingdom (T.J.G.)
| | - Pingjin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| |
Collapse
|
3
|
Leone A, Menichetti F, Vignati L, Sileo F, De Amicis R, Foppiani A, Bertoli S, Battezzati A. Relationship between bmi and glomerular filtration rate in a large cohort initiating a weight loss program: differential contributions of fat mass, fat-free mass, and abdominal fat compartments. Nutr J 2025; 24:78. [PMID: 40350415 PMCID: PMC12067886 DOI: 10.1186/s12937-025-01150-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 05/01/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND The relationship between BMI and chronic kidney disease is controversial, likely due to the inability of BMI to accurately define body composition and adipose tissue distribution. Our objective was to evaluate the synergistic contribution of fat-free mass, fat mass, visceral (VAT) and subcutaneous (SAT) adipose tissue, to glomerular filtration rate (GFR) in a large cohort of subjects. METHODS A cross-sectional study of 9704 subjects (72% female, median age 47y, median BMI 28.1 kg/m2) was carried out. Each patient underwent an anthropometric assessment (weight, height, waist circumference, % of body fat by body skinfolds), an ultrasound measurement of VAT and SAT and blood sampling to measure metabolic syndrome (MS) parameters and serum creatinine. GFR was estimated using the EPI-CKD equation. MS was defined according to the harmonized criteria. RESULTS Among 9,704 subjects, 61.1% had a normal renal function, while 29.3% reported a reduction, from slightly to severely. The BMI was initially negatively associated with GFR in the univariate model (β = -0.32, 95% CI: -0.39, -0.25), but after adjusting for %body fat, the association was lost. We then split the BMI into its two components, Fat Mass Index (FMI) and Fat Free Mass Index (FFMI), and observed that FMI (β = -1.23, 95% CI: -1.35, -1.12) and FFMI (β = 0.79, 95% CI: 0.65, 0.92) were associated with a decrease and an increase in GFR, respectively. VAT (β = -1.83, 95% CI: -2.00, -1.67) and SAT (β = 3.21, 95% CI: 2.86, 3.57) were independently associated with a decrease and an increase in GFR, respectively. Similar results were obtained when studying the association between BMI, body composition, adipose tissue distribution, and the risk of reduced GFR (<90 ml/min/1.73 m2). Stratification by sex and MS did not substantially alter the results. A significant association between VAT and reduced GFR was observed only in women. CONCLUSIONS Our study highlights the importance of considering body composition and fat distribution when assessing renal function.
Collapse
Affiliation(s)
- Alessandro Leone
- International Center for the Assessment of Nutritional Status and the development of Dietary Intervention Strategies (ICANS-DIS), Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy.
- IRCCS Istituto Auxologico Italiano, Clinical Nutrition Unit, Department of Endocrine and Metabolic Medicine, Milan, 20100, Italy.
| | - Francesca Menichetti
- International Center for the Assessment of Nutritional Status and the development of Dietary Intervention Strategies (ICANS-DIS), Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Laila Vignati
- IRCCS Istituto Auxologico Italiano, Clinical Nutrition Unit, Department of Endocrine and Metabolic Medicine, Milan, 20100, Italy
| | - Federica Sileo
- International Center for the Assessment of Nutritional Status and the development of Dietary Intervention Strategies (ICANS-DIS), Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
- IRCCS Istituto Auxologico Italiano, Clinical Nutrition Unit, Department of Endocrine and Metabolic Medicine, Milan, 20100, Italy
| | - Ramona De Amicis
- International Center for the Assessment of Nutritional Status and the development of Dietary Intervention Strategies (ICANS-DIS), Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
- IRCCS Istituto Auxologico Italiano, Obesity Unit and Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, Milan, 20145, Italy
| | - Andrea Foppiani
- International Center for the Assessment of Nutritional Status and the development of Dietary Intervention Strategies (ICANS-DIS), Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
- IRCCS Istituto Auxologico Italiano, Clinical Nutrition Unit, Department of Endocrine and Metabolic Medicine, Milan, 20100, Italy
| | - Simona Bertoli
- International Center for the Assessment of Nutritional Status and the development of Dietary Intervention Strategies (ICANS-DIS), Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
- IRCCS Istituto Auxologico Italiano, Obesity Unit and Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, Milan, 20145, Italy
| | - Alberto Battezzati
- International Center for the Assessment of Nutritional Status and the development of Dietary Intervention Strategies (ICANS-DIS), Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
- IRCCS Istituto Auxologico Italiano, Clinical Nutrition Unit, Department of Endocrine and Metabolic Medicine, Milan, 20100, Italy
| |
Collapse
|
4
|
Huang X, Ren X, Zhao L, Hao Y, Zhao Z, Chen F, Zhou J, Bai M, Chen S, Zhou X. Irisin Is a Potential Novel Biomarker and Therapeutic Target Against Kidney Diseases. Cell Biochem Funct 2025; 43:e70075. [PMID: 40318104 DOI: 10.1002/cbf.70075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/15/2025] [Accepted: 04/08/2025] [Indexed: 05/07/2025]
Abstract
Kidney diseases, characterized by renal dysfunction, are the leading causes of death worldwide. It is crucial to prevent and treat kidney diseases to reduce their associated morbidity and mortality. Moderate physical exercise has been recognized to be advantageous for kidney health. Irisin is an exercise-induced myokine that was identified in 2012. It plays an important role in energy and bone metabolism, oxidative stress reduction, anti-inflammatory processes, cell death inhibition, and cardiovascular protection. However, the relationship between irisin and kidney diseases have not been fully elucidated. This review explores the role of irisin as a biomarker for kidney disease diagnosis and its associated complications, as well as the mechanisms through which it participates in various cell death pathways, such as apoptosis, autophagy, pyroptosis, and ferroptosis. Furthermore, irisin secretion levels were discussed to provide a basis for kidney disease prevention and treatment avenues, as well as therapeutic guidance for developing new and promising intervention strategies. Clinical Trial Registration: None.
Collapse
Affiliation(s)
- Xiu Huang
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Xiya Ren
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Limei Zhao
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Yajie Hao
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Zhibo Zhao
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Fahui Chen
- Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Jinxiu Zhou
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Mengqi Bai
- Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Si Chen
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Xiaoshuang Zhou
- The Nephrology Department of Shanxi Provincial People's Hospital, Taiyuan, China
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
5
|
Wu Y, Lee TH, Cheng OH, Peden EK, Li Q, Wang J, Huang F, Melancon MP, Sheikh-Hamad D, Wang T, Truong L, Mitch WE, Liang M, Cheng J. Interplay between Skeletal Muscle Catabolism and Remodeling of Arteriovenous Fistula by Yes-Associated Protein 1 (YAP1) Signaling. J Am Soc Nephrol 2025; 36:845-858. [PMID: 39883520 PMCID: PMC12059102 DOI: 10.1681/asn.0000000605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
Key Points Atrophied muscle–derived myostatin stimulated mesenchymal stem cell differentiation and adverse arteriovenous (AV) fistula remodeling through yes-associated protein 1 (YAP1) activation. Treatment with myostatin peptibody inhibited muscle wasting and blocked mesenchymal stem cell activation and AV fistula fibrosis. A light-sensitive drug-release strategy was engineered for the periadventitial delivery of verteporfin to improve AV fistula patency. Background Arteriovenous (AV) fistulas are the preferred access for dialysis but have a high incidence of failure. The aim of this study was to understand the crosstalk between skeletal muscle catabolism and AV fistula maturation failure. Methods Skeletal muscle metabolism and AV fistula maturation were evaluated in mice with CKD. The roles of myostatin and yes-associated protein 1 (YAP1) in regulating the transdifferentiation of adventitial mesenchymal stem cells (MSCs) and intima hyperplasia in AV fistula were investigated. Nanoparticles carrying a YAP1 inhibitor, verteporfin, with light irradiation–controlled release were synthesized and applied to AV fistula. Results Increased trichrome signals and stenosis were observed in AV fistulas from mice treated with myostatin and from mice with CKD. By contrast, blocking myostatin function with an anti-myostatin peptibody not only improved body weight and muscle size in CKD mice but also decreased neointima formation in AV fistulas. In cultured MSCs, myostatin induced YAP1 expression, promoting the differentiation of MSCs into myofibroblasts and inducing extracellular matrix deposition. Red light irradiation–controlled release of verteporfin from nanoparticles blocked YAP1 activation and alleviated myostatin-induced MSC activation. Periadventitial application and red light irradiation of nanoparticles carrying verteporfin significantly suppressed stiffening and neointima formation in AV fistula. Conclusions CKD induced muscle wasting, leading to increased production of myostatin, which stimulated MSC activation and vascular fibrosis linked to AV fistula stenosis. YAP1 signaling was activated in these processes. Red light irradiation–controlled release of verteporfin offered a feasible approach for local vascular drug intervention to improve AV fistula maturation.
Collapse
Affiliation(s)
- Yongdong Wu
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Tae Hoon Lee
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Owen H. Cheng
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Eric K. Peden
- Department of Surgery, Houston Methodist Hospital, Houston, Texas
| | - Qingtian Li
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Fengzhang Huang
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Marites P. Melancon
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Sheikh-Hamad
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Tao Wang
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Luan Truong
- Department of Pathology and Genomic medicine, Houston Methodist Hospital, Houston, Texas
| | - William E. Mitch
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Ming Liang
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Jizhong Cheng
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
6
|
Wang Z, Li L, Yang M, Li B, Hu S. From Skeletal Muscle to Myocardium: Molecular Mechanisms of Exercise-Induced Irisin Regulation of Cardiac Fibrosis. Int J Mol Sci 2025; 26:3550. [PMID: 40332022 PMCID: PMC12026460 DOI: 10.3390/ijms26083550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
This study systematically elucidates the regulatory mechanisms and potential therapeutic value of the exercise-induced hormone Irisin in the pathological progression of cardiac fibrosis. Through comprehensive analysis and multidimensional data integration, we constructed a complete regulatory network of Irisin within the cardiovascular system, spanning its secretion, signal transduction, and precise regulatory control. Our findings demonstrate that exercise intervention significantly elevates circulating Irisin levels via the skeletal muscle-peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α)-fibronectin type III domain-containing protein 5 (FNDC5) signaling axis. Irisin establishes a multidimensional molecular barrier against cardiac fibrosis by targeting Sirtuin 1 (Sirt1) activation, inhibiting the transforming growth factor-beta (TGF-β)/Smad3 signaling pathway, and modulating the transcriptional activity of the mitochondrial biogenesis core factors PGC-1α and nuclear respiratory factor 1 (NRF-1). Moreover, the dual regulatory mechanism of the exercise-skeletal muscle-heart axis not only effectively suppresses the aberrant activation of cardiac fibroblasts but also significantly reduces collagen deposition, oxidative stress, and inflammatory infiltration by restoring mitochondrial dynamics balance. Taken together, this study reveals a novel exercise-mediated cardioprotective mechanism at the molecular interaction network level, thereby providing a theoretical basis for the development of non-pharmacological bio-intervention strategies targeting the Irisin signaling pathway and laying a translational foundation for precise exercise prescriptions in cardiovascular diseases.
Collapse
Affiliation(s)
- Zhao Wang
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Z.W.); (L.L.); (M.Y.)
| | - Lin Li
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Z.W.); (L.L.); (M.Y.)
| | - Meng Yang
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Z.W.); (L.L.); (M.Y.)
| | - Biao Li
- School of Physical Science, Hefei Normal University, Hefei 230061, China
| | - Siyuan Hu
- School of Sports & Arts, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
7
|
Kim J, Lee S, Jang H, Jung S, Jung M, Yun J, Jeon H, Kim H, Chang S, Lee E, Kim H. Transcriptional Intermediary Factor 1γ-Induced Irisin in Skeletal Muscle Attenuates Renal Fibrosis in Diabetic Nephropathy. J Cachexia Sarcopenia Muscle 2025; 16:e13810. [PMID: 40235186 PMCID: PMC12000539 DOI: 10.1002/jcsm.13810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 03/06/2025] [Accepted: 03/25/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Transcriptional intermediary factor 1γ (TIF1γ) is a negative regulator of TGF-β1 signalling and has been associated with patient survival in renal cell carcinoma. However, its role in diabetes mellitus (DM), particularly in diabetic nephropathy (DN), remains unclear. DN is the leading cause of chronic kidney disease (CKD). We investigated the potential role of TIF1γ in mitigating multiple DM-related complications. METHODS Mice were divided into four groups: db/m+, db/db and db/db mice treated with cytomegalovirus- or TGF-TIF1γ plasmids (40 μg/mouse; intraperitoneally weekly for 16 weeks). Renal injury, fibrosis, function and gene expression related to fibrosis and epithelial-mesenchymal transition (EMT) in the kidneys were assessed. Muscle atrophy, regeneration markers, myokine levels and exercise capacity were evaluated. C2C12 cells were exposed to palmitate with or without TIF1γ transfection, and irisin expression and secretion were measured. Muscle-kidney crosstalk was analysed using conditioned media (CM) from TIF1γ-transfected C2C12 cells in palmitate-treated human kidney (HK)-2 cells. Additionally, HK-2 cells were incubated in CM from fibronectin type III domain-containing protein (FNDC)5-knockdown C2C12 cells to confirm irisin-mediated kidney crosstalk by TIF1γ. RESULTS TIF1γ treatment in db/db mice resulted in a significant attenuation of renal tubulointerstitial fibrosis (1.5-fold decrease), glomerular injury (1.8-fold improvement), tubular injury (1.6-fold improvement), renal dysfunction (1.7-fold improvement) and a reduction in EMT-related factors (1.8-fold decrease) (p < 0.05). The levels of administered TIF1γ plasmids were higher in skeletal muscle than in renal tissues. TIF1γ expression was significantly elevated in the skeletal muscle of db/db mice treated with TIF1γ plasmids (6.5-fold) (p < 0.05). Mice receiving both plasmids exhibited a 1.8-fold reduction in pathological muscle morphology and atrophy-related gene expression, a 3.0-fold increase in regeneration-related gene expression and a 1.6-fold improvement in muscle function (p < 0.05). Irisin expression increased by 2.1-fold in skeletal muscle and serum (p < 0.05). In TIF1γ-transfected C2C12 cells, irisin secretion was elevated by 1.5-fold (p < 0.05). CM from TIF1γ-transfected C2C12 cells attenuated EMT in palmitate-treated HK-2 cells, compared with medium from nontransfected C2C12 cells (1.9-fold improvement [p < 0.05]). Conversely, FNDC5 knockdown in C2C12 cells accelerated EMT in palmitate-treated HK-2 cells, as evidenced by decreased bone morphogenetic protein-7 (1.6-fold) and increased EMT-related factors (2.1-fold) (p < 0.05), compared with palmitate alone and small interfering RNA control. CONCLUSIONS Our findings emphasize the potential of TIF1γ as a multitargeted therapeutic agent for DN, mitigating both renal and muscular complications through direct fibrosis inhibition and indirect myokine-mediated inter-organ crosstalk.
Collapse
Affiliation(s)
- Jin Hyun Kim
- Biomedical Research InstituteGyeongsang National University HospitalJinjuRepublic of Korea
- Institute of Medical ScienceGyeongsang National UniversityJinjuRepublic of Korea
| | - Seunghye Lee
- Division of Nephrology, Department of Internal MedicineGyeongsang National University College of Medicine and Gyeongsang National University HospitalJinjuRepublic of Korea
| | - Hani Jang
- Institute of Medical ScienceGyeongsang National UniversityJinjuRepublic of Korea
- Division of Nephrology, Department of Internal MedicineGyeongsang National University College of Medicine and Gyeongsang National University HospitalJinjuRepublic of Korea
| | - Sehyun Jung
- Division of Nephrology, Department of Internal MedicineGyeongsang National University College of Medicine and Gyeongsang National University HospitalJinjuRepublic of Korea
| | - Myeong Hee Jung
- Biomedical Research InstituteGyeongsang National University HospitalJinjuRepublic of Korea
| | - Jeong Won Yun
- Biomedical Research InstituteGyeongsang National University HospitalJinjuRepublic of Korea
| | - Haejin Jeon
- Division of Nephrology, Department of Internal MedicineGyeongsang National University College of Medicine and Gyeongsang National University HospitalJinjuRepublic of Korea
| | - Hyun‐Jung Kim
- Institute of Medical ScienceGyeongsang National UniversityJinjuRepublic of Korea
- Division of Nephrology, Department of Internal MedicineGyeongsang National University College of Medicine and Gyeongsang National University HospitalJinjuRepublic of Korea
| | - Se‐Ho Chang
- Institute of Medical ScienceGyeongsang National UniversityJinjuRepublic of Korea
- Division of Nephrology, Department of Internal MedicineGyeongsang National University College of Medicine and Gyeongsang National University HospitalJinjuRepublic of Korea
| | - Eun Ju Lee
- Biomedical Research InstituteSeoul National University HospitalSeoulRepublic of Korea
| | - Hyo‐Soo Kim
- Molecular Medicine & Biopharmaceutical SciencesSeoul National UniversitySeoulRepublic of Korea
- Department of Internal MedicineSeoul National University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
8
|
Sun P, Chen Q, Chen X, Zhou J, Long T, Ma Y, Zhou M, Hu Z, Tian J, Zhu F, Yang Z, Xie L, Wu Q, Nie J. Renal tubular S100A7a impairs fatty acid oxidation and exacerbates renal fibrosis via both intracellular and extracellular pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167656. [PMID: 39778778 DOI: 10.1016/j.bbadis.2025.167656] [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: 09/19/2024] [Revised: 12/16/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025]
Abstract
A couple of S100 family proteins (S100s) have been reported to exert pro-inflammatory functions in the progression of renal fibrosis. Unlike some S100s which are expressed by both epithelial and stromal inflammatory cells, S100A7 is restricted expressed in epithelium. Persistent S100A7 expression occurs in some invasive carcinomas and is associated with poor prognostic factors. Whereas, whether it is implicated in renal tubular epithelial cell injury and kidney disease remains unexplored. In this study, we demonstrate that S100A7 is highly upregulated in tubular cells of both mouse renal fibrotic lesions and kidney biopsies from patients with chronic kidney disease (CKD). The level of renal S100A7 was associated with both the decline of renal function and the progression of renal fibrosis in CKD patients. Overexpressing S100A7a impaired fatty acid oxidation (FAO) and promoted lipid peroxidation in proximal tubular cells (PTCs). Mechanistically, S100A7a interacts with β-catenin, thereby preventing its ubiquitination and degradation by the β-TrCP-SCF complex, and in turn activated β-catenin signaling, downregulated the expression of PGC-1α. Additionally, S100A7a exacerbated lipid peroxidation via RAGE-p-ERK-NOX2 pathway. Specific deletion of S100a7a in tubular cells enhanced FAO and reduced lipid peroxidation, resulting in improved renal function and alleviation of renal fibrosis induced by unilateral ureteral obstruction and unilateral ischemia-reperfusion injury. Collectively, we delineate a previously unrecognized function of S100A7a in the progression of renal fibrosis.
Collapse
Affiliation(s)
- Pengxiao Sun
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qingzhou Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaomei Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiaxin Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Tantan Long
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuanyuan Ma
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Miaomiao Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zheng Hu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jianwei Tian
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fengxin Zhu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhenhua Yang
- Department of Nephrology, The First Affiliated Hospital, Guangxi Medical University, Nanning 530021, China
| | - Liling Xie
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qiaoyuan Wu
- Department of Nephrology, The First Affiliated Hospital, Guangxi Medical University, Nanning 530021, China.
| | - Jing Nie
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Biobank of Peking University First Hospital, Peking University First Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University Health Science Center, Peking University, Beijing 100034, China.
| |
Collapse
|
9
|
Peng L, Li S, Huang Q, Sun Y, Sun J, Luo T, Wang Y, Hu Z, Lai W, Peng H. Irisin-mediated muscle-renal crosstalk as a protective mechanism against contrast-induced acute kidney injury via cGAS-STING signalling inhibition. Clin Transl Med 2025; 15:e70235. [PMID: 40008481 PMCID: PMC11862893 DOI: 10.1002/ctm2.70235] [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: 10/17/2024] [Revised: 01/17/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Contrast-induced acute kidney injury (CI-AKI) continues to pose a pressing clinical challenge during invasive cardiovascular procedures due to the limited availability of preventative strategies. We aimed to demonstrate that irisin, a myokine induced by exercise, protects against CI-AKI by inhibiting the cGAS-STING inflammatory pathway. METHODS AND RESULTS We explored the relationship between serum irisin levels and CI-AKI incidence in patients administered the contrast media iohexol. Notably, lower serum irisin levels were strongly associated with an increased incidence of CI-AKI following contrast media administration. To establish a causal link between serum irisin levels and CI-AKI, we utilised a mouse model that simulates exercise by overexpressing muscle-specific PGC-1α. This approach showed a significant reduction in tubular injury and mitochondrial dysfunction induced by iohexol via cGAS/STING suppression, thereby diminishing inflammation. Mechanistically, irisin was found to inhibit the activation of cGAS/STING, preventing double stranded DNA (dsDNA) leakage and reducing inflammation in tubular epithelial cells (TECs). Pharmacological inhibition of STING further corroborated these observations. Moreover, we identified integrin complex αV/β5 as the irisin receptor on TECs, which is essential for irisin-mediated suppression of cGAS-STING signalling and resolution of inflammation. CONCLUSIONS Our data position irisin as a crucial factor in muscle‒kidney crosstalk, inhibiting cGAS-STING signalling and preventing dsDNA leakage via integrin αV/β5 in TECs, thus mitigating tubular injury and inflammation. These data underscore the potential of irisin as both a predictive biomarker for CI-AKI and a promising candidate for preventative strategies against CI-AKI. HIGHLIGHTS Irisin mediated muscle-kidney crosstalk mitigated tubular injury and inflammation. Irisin inhibited the cGAS-STING signalling activation via integrin αV/β5 in tubular epithelial cells. Irisin was a predictive biomarker and a promising candidate for CI-AKI.
Collapse
Affiliation(s)
- Long Peng
- Division of Cardiovascular Medicine, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Suhua Li
- Division of Cardiovascular Medicine, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Qiang Huang
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yuxiang Sun
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Juan Sun
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Ting Luo
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yanlin Wang
- Division of Nephrology, Department of MedicineUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Zhaoyong Hu
- Division of Nephrology, Department of MedicineBaylor College of MedicineHoustonTexasUSA
| | - Weiyan Lai
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Hui Peng
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
10
|
Liu S, Yang Y, Li Q, Yu L, Zong Z, Zang R, Ji W, Sun S. Ubiquitin-specific peptidase 10 promotes renal interstitial fibrosis progression through deubiquitinating and stabilizing P53 protein. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167660. [PMID: 39788218 DOI: 10.1016/j.bbadis.2025.167660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/30/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Renal interstitial fibrosis is the main factor determining chronic kidney disease (CKD) progression, and renal tubular epithelial cells are the key drivers of this pathological process. Herein, we revealed significantly increased ubiquitin-specific peptidase 10 (USP10) expression in the kidney tissues of both patients with CKD and mice induced by unilateral ureteral obstruction, as well as in transforming growth factor-beta 1 (TGFβ1)-induced renal tubular epithelial cells. In vivo, treatment with the USP10 small molecule inhibitor Spautin-1, which inhibits its deubiquitinating activity, weakened renal interstitial fibrosis progression and alleviated the subsequent inflammatory response and oxidative stress in male mice. In vitro, knocking down USP10 or inhibiting its deubiquitinating activity through Spautin-1 significantly reduced fibronectin expression and ameliorated TGFβ1-induced renal tubular epithelial cell dedifferentiation. Additionally, our results revealed that USP10 directly binds to P53 and removes the K48-linked polyubiquitin chains from P53, thereby affecting its ubiquitination, stability, and nuclear translocation, which subsequently leads to the upregulation of P21 and promotes fibrotic gene expression in injured renal tubular epithelial cells, ultimately exacerbating renal interstitial fibrosis. In conclusion, USP10 is inhibited through the P53 signaling pathway to alleviate the progression of renal interstitial fibrosis and serve as a potential target for treating CKD.
Collapse
Affiliation(s)
- Suwen Liu
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China.
| | - Yunwen Yang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China; Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Qian Li
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Lichun Yu
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Zihan Zong
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Ruixian Zang
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Wentao Ji
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Shuzhen Sun
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China.
| |
Collapse
|
11
|
Lee K, Kim M. Evolutionary Insights into Irisin/FNDC5: Roles in Aging and Disease from Drosophila to Mammals. Biomolecules 2025; 15:261. [PMID: 40001564 PMCID: PMC11853655 DOI: 10.3390/biom15020261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/03/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
The Irisin/FNDC5 protein family has emerged as a pivotal link between exercise and the prevention of age-associated diseases. Irisin is highly expressed during exercise from skeletal and cardiac muscle cells, playing a critical role in mediating systemic health benefits through its actions on various tissues. However, Irisin levels decline with age, correlating with a heightened incidence of diseases such as muscle weakness, cardiovascular disorders, and neurodegeneration. Notably, the administration of Irisin has shown significant potential in both preventing and treating these conditions. Recently, an Irisin/FNDC5 homolog was identified in an invertebrate Drosophila model, providing valuable insights into its conserved role in exercise physiology. Importantly, Irisin/FNDC5 has been demonstrated to regulate autophagy-a process essential for clearing excessive nutrients, toxic aggregates, and dysfunctional organelles-in both flies and mammals. Dysregulated autophagy is often implicated in age-related diseases, highlighting its relevance to Irisin/FNDC5's functions. These findings deepen our understanding of Irisin/FNDC5's roles and its potential as a therapeutic target for mitigating aging-related health decline. Further studies are needed to elucidate the precise mechanisms by which Irisin regulates autophagy and its broader impact on physiological aging and related diseases.
Collapse
Affiliation(s)
| | - Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
12
|
Li XQ, Jin B, Liu SX, Zhu Y, Li N, Zhang QY, Wan C, Feng Y, Xing YX, Ma KL, Liu J, Jiang CM, Lu J. Neddylation of RhoA impairs its protein degradation and promotes renal interstitial fibrosis progression in diabetic nephropathy. Acta Pharmacol Sin 2025:10.1038/s41401-024-01460-z. [PMID: 39900822 DOI: 10.1038/s41401-024-01460-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/15/2024] [Indexed: 02/05/2025]
Abstract
Diabetic nephropathy (DN) is a common and serious complication of diabetes, characterized by chronic fibro-inflammatory processes with an unclear pathogenesis. Renal fibrosis plays a significant role in the development and progression of DN. While recent research suggests that the neddylation pathway may influence fibrotic processes, its specific dysregulation in DN and the underlying mechanisms remain largely unexplored. This study identified the neddylation of RhoA as a novel post-translational modification that regulates its expression and promotes renal fibrosis in DN. We here demonstrated that two key components of the neddylation pathway-NEDD8-activating enzyme E1 subunit 1 (NAE1) and NEDD8-are significantly upregulated in human chronic kidney disease (CKD) specimens compared to healthy kidneys, implicating neddylation in CKD-associated fibrosis. Our findings further revealed that both pharmacological inhibition of neddylation using MLN4924 and genetic knockdown of NAE1 mitigate renal fibrosis in mouse models of streptozotocin-induced diabetes and unilateral ureteral obstruction (UUO). Immunoprecipitation-mass spectrometry (IP-MS) and subsequent function assays demonstrated a direct interaction between RhoA and NEDD8. Importantly, neddylation inhibition reduced RhoA protein expression, highlighting a potential therapeutic target. Additionally, a positive correlation was noted between elevated NEDD8 mRNA levels and RhoA mRNA expression in human CKD specimens. RhoA overexpression counteracted the antifibrotic effects of neddylation inhibition, underscoring its critical role in fibrosis progression. Mechanistically, we unveiled that neddylation enhances RhoA protein stability by inhibiting its ubiquitination-mediated degradation, which subsequently activates the ERK1/2 pathway. Collectively, this study provides novel insights into NAE1-dependent RhoA neddylation as a key contributor to renal fibrosis in DN. The NAE1 protein mediates RhoA protein hyper-neddylation and subsequent stabilization of the RhoA protein, which, in turn, contributes to the development of renal fibrosis and inflammation through an ERK1/2-dependent mechanism. Consequently, targeting neddylation inhibition represents a viable therapeutic approach for the treatment of renal fibrosis in DN.
Collapse
Affiliation(s)
- Xue-Qi Li
- Institute of Nephrology, Nanjing Drum Tower Hospital, School of Medicine, Southeast University, Nanjing, 210008, China
| | - Bo Jin
- Department of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Si-Xiu Liu
- Department of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yan Zhu
- Department of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Nan Li
- Department of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Qing-Yan Zhang
- Department of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Cheng Wan
- Department of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yuan Feng
- Department of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yue-Xian Xing
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Kun-Ling Ma
- Department of Nephrology, The Second Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, 310009, China
| | - Jing Liu
- Department of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Chun-Ming Jiang
- Department of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Jian Lu
- Department of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
13
|
Wu M, Li H, Sun X, Zhong R, Cai L, Chen R, Madeniyet M, Ren K, Peng Z, Yang Y, Chen W, Tu Y, Lai M, Deng J, Wu Y, Zhao S, Ruan Q, Rao M, Xie S, Ye Y, Wan J. Aerobic exercise prevents renal osteodystrophy via irisin-activated osteoblasts. JCI Insight 2025; 10:e184468. [PMID: 39883525 PMCID: PMC11949034 DOI: 10.1172/jci.insight.184468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
Renal osteodystrophy is commonly seen in patients with chronic kidney disease (CKD) due to disrupted mineral homeostasis. Given the impaired renal function in these patients, common antiresorptive agents, including bisphosphonates, must be used with caution or even contraindicated. Therefore, an alternative therapy without renal burden to combat renal osteodystrophy is urgently needed. Here, we report that clinically relevant aerobic exercise significantly prevents high-turnover renal osteodystrophy in CKD mice and patients with CKD without compromising renal function. Mechanistically, 4-week aerobic exercise in CKD mice increased expression of skeletal muscle PPARγ coactivator-1α (PGC-1α) and circulating irisin. Both exercise and irisin administration significantly activated osteoblasts, but not osteoclasts, via integrin αvβ5, thereby conferring bone quality benefits. Removal of irisin-influenced thermogenic adipose tissues or genetic ablation of uncoupling protein 1 did not alter the irisin-conferred antiosteodystrophy effect. Importantly, in a pilot clinical study, 12-week aerobic exercise in patients with high-grade CKD significantly increased circulating irisin and prevented osteodystrophy progression, without detectable renal burden. The combination of irisin and current antiresorptive agents effectively rescued renal osteodystrophy in mice. Our work provides mechanistic insights into the role of exercise and irisin in renal osteodystrophy, and it highlights a clinically relevant, low-cost, kidney-friendly therapy for patients with this devastating disease.
Collapse
Affiliation(s)
- Meng Wu
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Huilan Li
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoting Sun
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Rongrong Zhong
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Linli Cai
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Ruibo Chen
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Madiya Madeniyet
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kana Ren
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhen Peng
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yujie Yang
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Weiqin Chen
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Yanling Tu
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Miaoxin Lai
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Jinxiu Deng
- Department of Nephrology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Yuting Wu
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Shumin Zhao
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Qingyan Ruan
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Mei Rao
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Sisi Xie
- Department of Cardiology, Basic Scientific Research Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Ying Ye
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Jianxin Wan
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, and
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
14
|
Pinto A, Haytural H, Loss CM, Alvarez C, Ertas A, Curtis O, Williams AR, Murphy G, Salleng K, Gografe S, Altıntaş A, Kafri T, Barres R, Deshmukh AS, van Praag H. Muscle Cathepsin B treatment improves behavioral and neurogenic deficits in a mouse model of Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.20.633414. [PMID: 39896474 PMCID: PMC11785056 DOI: 10.1101/2025.01.20.633414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Muscle secretes factors during exercise that enhance cognition. Myokine Cathepsin B (Ctsb) is linked to memory function, but its role in neurodegenerative disease is unclear. Here we show that AAV-vector-mediated Ctsb overexpression in skeletal muscle in an Alzheimer's Disease (AD) mouse model (APP/PS1), improves motor coordination, memory function and adult hippocampal neurogenesis, while plaque pathology and neuroinflammation remain unchanged. Additionally, in AD mice, Ctsb treatment modifies hippocampal, muscle and plasma proteomic profiles to resemble that of wildtype controls. Conversely, in wildtype mice, Ctsb expression causes memory deficits and results in protein profiles across tissues that are comparable to AD control mice. In AD mice, Ctsb treatment increases the abundance of hippocampal proteins involved in mRNA metabolism and protein synthesis, including those relevant to adult hippocampal neurogenesis and memory function. Furthermore, Ctsb treatment enhances plasma metabolic and mitochondrial processes, and reduces inflammatory responses. In muscle, Ctsb expression elevates protein translation in AD mice, whereas in wildtype mice mitochondrial proteins decrease. Overall, the biological changes in the treatment groups are consistent with effects on memory function. Thus, skeletal muscle Ctsb application has potential as an AD therapeutic intervention.
Collapse
Affiliation(s)
- Alejandro Pinto
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Hazal Haytural
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Cássio Morais Loss
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Claudia Alvarez
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Asude Ertas
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Olivia Curtis
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Alyssa R. Williams
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Grayson Murphy
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Ken Salleng
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Sylvia Gografe
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Ali Altıntaş
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Tal Kafri
- Gene Therapy Center, University of North Carolina at Chapel Hill, Thurston-Bowles, NC 27599, USA
| | - Romain Barres
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur & Centre National pour la Recherche Scientifique (CNRS), 06560 Valbonne, France
| | - Atul S. Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Henriette van Praag
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA
| |
Collapse
|
15
|
Lai TL, Park SY, Nguyen G, Pham PTM, Kang SM, Hong J, Lee JH, Im SS, Choi DH, Cho EH. Irisin Attenuates Hepatic Stellate Cell Activation and Liver Fibrosis in Bile Duct Ligation Mice Model and Improves Mitochondrial Dysfunction. Endocrinol Metab (Seoul) 2024; 39:908-920. [PMID: 39497457 PMCID: PMC11695487 DOI: 10.3803/enm.2024.1984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/11/2024] [Accepted: 06/27/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGRUOUND Liver fibrosis is a common outcome of chronic liver disease and is primarily driven by hepatic stellate cell (HSC) activation. Irisin, a myokine released during physical exercise, is beneficial for metabolic disorders and mitochondrial dysfunction. This study aimed to explore the effects of irisin on liver fibrosis in HSCs, a bile duct ligation (BDL) mouse model, and the associated mitochondrial dysfunction. METHODS In vitro experiments utilized LX-2 cells, a human HSC line, stimulated with transforming growth factor-β1 (TGF-β1), a major regulator of HSC fibrosis, with or without irisin. Mitochondrial function was assessed using mitochondrial fission markers, transmission electron microscopy, mitochondrial membrane potential, and adenosine triphosphate (ATP) production. In vivo, liver fibrosis was induced in mice via BDL, followed by daily intraperitoneal injections of irisin (100 μg/kg/day) for 10 days. RESULTS In vitro, irisin mitigated HSC activation and reduced reactive oxygen species associated with the TGF-β1/Smad signaling pathway. Irisin restored TGF-β1-induced increases in fission markers (Fis1, p-DRP1) and reversed the decreased expression of TFAM and SIRT3. Additionally, irisin restored mitochondrial membrane potential and ATP production lowered by TGF-β1 treatment. In vivo, irisin ameliorated the elevated liver-to-body weight ratio induced by BDL and alleviated liver fibrosis, as evidenced by Masson's trichrome staining. Irisin also improved mitochondrial dysfunction induced by BDL surgery. CONCLUSION Irisin effectively attenuated HSC activation, ameliorated liver fibrosis in BDL mice, and improved associated mitochondrial dysfunction. These findings highlight the therapeutic potential of irisin for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Thuy Linh Lai
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - So Young Park
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Giang Nguyen
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Phuc Thi Minh Pham
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Seon Mee Kang
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Jeana Hong
- Department of Pediatrics, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Jae-Ho Lee
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Dae-Hee Choi
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Eun-Hee Cho
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| |
Collapse
|
16
|
Honda M, Inoue R, Nishiyama K, Ueda T, Komuro A, Amano H, Sugisawa R, Dash S, Shirakawa J, Okada H. Vgll2 as an integrative regulator of mitochondrial function and contractility specific to skeletal muscle. J Cell Physiol 2024; 239:e31436. [PMID: 39286968 DOI: 10.1002/jcp.31436] [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: 05/13/2024] [Revised: 07/26/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
During skeletal muscle adaptation to physiological or pathophysiological signals, contractile apparatus and mitochondrial function are coordinated to alter muscle fiber type. Although recent studies have identified various factors involved in modifying contractile proteins and mitochondrial function, the molecular mechanisms coordinating contractile and metabolic functions during muscle fiber transition are not fully understood. Using a gene-deficient mouse approach, our previous studies uncovered that vestigial-like family member 2 (Vgll2), a skeletal muscle-specific transcription cofactor activated by exercise, is essential for fast-to-slow adaptation of skeletal muscle. The current study provides evidence that Vgll2 plays a role in increasing muscle mitochondrial mass and oxidative capacity. Transgenic Vgll2 overexpression in mice altered muscle fiber composition toward the slow type and enhanced exercise endurance, which contradicted the outcomes observed with Vgll2 deficiency. Vgll2 expression was positively correlated with the expression of genes related to mitochondrial function in skeletal muscle, mitochondrial DNA content, and protein abundance of oxidative phosphorylation complexes. Additionally, Vgll2 overexpression significantly increased the maximal respiration of isolated muscle fibers and enhanced the suppressive effects of endurance training on weight gain. Notably, no additional alteration in expression of myosin heavy chain genes was observed after exercise, suggesting that Vgll2 plays a direct role in regulating mitochondrial function, independent of its effect on contractile components. The observed increase in exercise endurance and metabolic efficiency may be attributed to the acute upregulation of genes promoting fatty acid utilization as a direct consequence of Vgll2 activation facilitated by endurance exercise. Thus, the current study establishes that Vgll2 is an integrative regulator of mitochondrial function and contractility in skeletal muscle.
Collapse
Affiliation(s)
- Masahiko Honda
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Ryota Inoue
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Gunma, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Kuniyuki Nishiyama
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Gunma, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, Yokohma, Kanagawa, Japan
| | - Takeshi Ueda
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Akiyoshi Komuro
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Hisayuki Amano
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Ryoichi Sugisawa
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Suman Dash
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Jun Shirakawa
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Gunma, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Hitoshi Okada
- Department of Biochemistry, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Graduate School of Medical Sciences, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
- Antiaging Center, Kindai University, Higashi-Osaka, Osaka, Japan
| |
Collapse
|
17
|
Hoshino J, Ohigashi T, Tsunoda R, Ito Y, Kai H, Saito C, Okada H, Narita I, Wada T, Maruyama S, Pisoni R, Pecoits-Filho R, Yamagata K. Physical activity and renal outcome in diabetic and non-diabetic patients with chronic kidney disease stage G3b to G5. Sci Rep 2024; 14:26378. [PMID: 39487292 PMCID: PMC11530613 DOI: 10.1038/s41598-024-77497-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024] Open
Abstract
The association of physical activity with renal outcome and mortality in advanced chronic kidney disease (CKD; i.e., estimated glomerular filtration rate [eGFR] < 45 ml/min/1.73m2) is poorly studied. We examined this association in patients with advanced CKD in Japan. We used the Rapid Assessment of Physical Activity to assess baseline physical activity and classify patients as active or inactive. CKD progression was defined as 40% decline in eGFR, eGFR < 10, or requiring dialysis or transplantation. Among the 1,808 eligible patients, after adjusting for possible confounders, hazard ratios (HRs) for poor renal outcome in the active group were 0.68 (95% CI, 0.44-1.04), 1.09 (0.86-1.38), and 1.01 (0.82-1.25) in CKD stage G3b, G4, and G5, respectively, suggesting a renal benefit of exercise in CKD stage G3b. Adjusted HRs for death were 0.79 (0.40-1.57), 0.55 (0.38-0.80), and 0.75 (0.44-1.26) in stage G3b, G4, and G5, respectively. While the adjusted HRs of death were 0.84 (0.52-1.38) and 0.60 (0.43-0.83) in diabetic and non-diabetic patients, suggesting that exercise may reduce mortality in non-diabetic patients. Our study suggests that exercise is associated with better survival in non-diabetic patients with CKD stage G3b-5, and better renal outcome in diabetic and non-diabetic CKD stage G3b.
Collapse
Affiliation(s)
- Junichi Hoshino
- Department of Nephrology, Tokyo Women's Medical University, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan.
- Department of Nephrology, Faculty of Medicine, Institute of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Tomohiro Ohigashi
- Tsukuba Clinical Research and Development Organization (T-CReDO), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Ryoya Tsunoda
- Department of Nephrology, Faculty of Medicine, Institute of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yukiko Ito
- Tsukuba Clinical Research and Development Organization (T-CReDO), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hirayasu Kai
- Ibaraki Clinical Education and Training Center, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Nephrology, Ibaraki Prefectural Central Hospital, Kasama, Ibaraki, Japan
| | - Chie Saito
- Department of Nephrology, Faculty of Medicine, Institute of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hirokazu Okada
- Department of Nephrology, Saitama Medical University, Saitama, Japan
| | - Ichiei Narita
- Niigata Institute for Health and Sports Medicine, Niigata, Japan
| | - Takashi Wada
- Department of Nephrology and Rheumatology, Kanazawa University, Ishikawa, Japan
| | | | - Ronald Pisoni
- Arbor Research Collaborative for Health, Ann Arbor, MI, USA
| | | | - Kunihiro Yamagata
- Department of Nephrology, Faculty of Medicine, Institute of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
18
|
Alsawaf Y, Maksimovic I, Zheng J, Zhang S, Vuckovic I, Dzeja P, Macura S, Irazabal MV. A brief harvesting-freezing delay significantly alters the kidney metabolome and leads to false positive and negative results. Am J Physiol Renal Physiol 2024; 327:F697-F711. [PMID: 39205659 PMCID: PMC11563588 DOI: 10.1152/ajprenal.00131.2024] [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: 04/30/2024] [Revised: 07/25/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Abnormalities in distinct metabolic pathways have been associated with the pathogenesis and progression of many forms of kidney disease. Metabolomics analyses can be used to determine organ-specific metabolic fingerprints and, ideally, should represent the metabolic state of the organ at the exact moment the sample is harvested. However, conventional harvesting methods depend on posteuthanasia tissue harvest, which results in ischemia conditions and metabolome changes that could potentially introduce artifacts into the final studies. We recently optimized a modified clamp-freezing technique for rodent kidney harvesting and freezing, significantly reducing ischemia and freezing times and granting a closer snapshot of in vivo metabolism. In this study, we characterized and compared the metabolome of kidneys harvested using our modified approach versus traditional techniques to determine which metabolites are preferentially affected by a brief lapse of ischemia and freezing delay and which are more stable. We used Sprague-Dawley rats as a model of wild-type (WT) kidneys and PCK [polycystic kidney disease (PKD)] rats as a model of chronic kidney disease kidneys. Finally, we compared the metabolic profile of clamp-frozen and delayed WT and PKD kidneys to determine which metabolic changes are most likely observed in vivo in PKD and which could be subjected to false positive or negative results. Our data indicate that a short harvesting-freezing delay is sufficient to impart profound metabolic changes in WT and PKD kidneys, leading to false positive and negative differences when comparing these genotypes. In addition, we identified a group of metabolites that were more stable. Interestingly, while the delay had a similar effect between WT and PKD, there were notable differences. The data obtained indicate that the quick clamp-freezing technique for kidney metabolomics provides a more accurate interpretation of the in vivo metabolic changes associated with the disease state. NEW & NOTEWORTHY Our study shows that a brief harvesting-freezing delay associated with organ collection and freezing can significantly alter the kidney metabolic profile of both male and female wild-type and a genetic model of chronic kidney disease. Importantly, given that the effect of this delay differs among genotypes, it is not safe to assume that equally delaying harvesting-freezing in wild-type and polycystic kidney disease kidneys adequately controls this effect, ultimately leading to false positive and negative results among different renal diseases.
Collapse
Affiliation(s)
- Yahya Alsawaf
- Mayo Translational PKD Center, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Igor Maksimovic
- Mayo Translational PKD Center, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Jamie Zheng
- Mayo Translational PKD Center, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Song Zhang
- Metabolomics Core, Mayo Clinic, Rochester, Minnesota, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Ivan Vuckovic
- Metabolomics Core, Mayo Clinic, Rochester, Minnesota, United States
| | - Petras Dzeja
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Slobodan Macura
- Department of Biochemistry, Mayo Clinic, Rochester, Minnesota, United States
| | - Maria V Irazabal
- Mayo Translational PKD Center, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
19
|
Hayden CM, Begue G, Gamboa JL, Baar K, Roshanravan B. Review of Exercise Interventions to Improve Clinical Outcomes in Nondialysis CKD. Kidney Int Rep 2024; 9:3097-3115. [PMID: 39534200 PMCID: PMC11551061 DOI: 10.1016/j.ekir.2024.07.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 06/22/2024] [Accepted: 07/26/2024] [Indexed: 11/16/2024] Open
Abstract
Exercise interventions in chronic kidney disease (CKD) have received growing interest, with over 30 meta-analyses published in the past 5 years. The potential benefits of exercise training in CKD range from slowing disease progression to improving comorbidities and quality of life. Nevertheless, there is a lack of large, randomized control trials in diverse populations, particularly regarding exercise in nondialysis-dependent CKD (NDD). When exercise interventions are implemented, they often lack fundamental features of exercise training such as progressive overload, personalization, and specificity. Furthermore, the physiology of exercise and CKD-specific barriers appear poorly understood. This review explores the potential benefits of exercise training in NDD, draws lessons from previous interventions and other fields, and provides several basic tools that may help improve interventions in research and practice.
Collapse
Affiliation(s)
- Christopher M.T. Hayden
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California, USA
| | - Gwénaëlle Begue
- Kinesiology Department, California State University, Sacramento, California, USA
| | - Jorge L. Gamboa
- Department of Medicine, Division of Clinical Pharmacology. Vanderbilt University. Nashville, Tennessee, USA
| | - Keith Baar
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California, USA
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California, USA
| | - Baback Roshanravan
- Department of Medicine, Division of Nephrology. University of California Davis. Sacramento, California, USA
| |
Collapse
|
20
|
Maden M, Ider M, Or ME, Dokuzeylül B, Gülersoy E, Kılıçkaya MC, Bilgiç B, Durgut MK, İzmirli S, Iyigün SS, Telci DZ, Naseri A. The clinical efficacy of cGMP-specific sildenafil on mitochondrial biogenesis induction and renal damage in cats with acute on chronic kidney disease. BMC Vet Res 2024; 20:499. [PMID: 39478527 PMCID: PMC11526613 DOI: 10.1186/s12917-024-04345-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Mitochondrial biogenesis (MB) induction has recently emerged as potential therapeutic approaches in kidney pathology and the mitochondria-targeted therapies should be investigated to improve treatment of animals with kidney diseases. This study aimed to investigate the effects of MB induction with sildenafil citrate on the cGMP/NO pathway, glomerular filtration, and reduction of kidney damage and fibrosis (TGF-β/SMAD pathway) in cats with acute on chronic kidney disease (ACKD). Thirty-three cats were divided into the non-azotemic (healthy) group (n:8) and the ACKD group (n:25), comprising different breeds, sexes, and ages. Sildenafil citrate was administered to the non-azotemic and ACKD groups (2.5 mg/kg, PO, q12 hours) for 30 days. Serum and urine NO, MDA, NGAL, KIM-1, TGF-β1, IL-18, FGF 23, PGC-1α and cGMP concentrations were measured. RESULTS Serum cGMP concentrations increased (P < 0.05) in the non-azotemic group during the 2nd (median 475.99 pmol/mL) and 3rd (median 405.01 pmol/mL) weeks of the study, whereas serum cGMP concentrations decreased in the ACKD group during the 4th(median 188.52 pmol/mL) week compared to the non-azotemic group (P < 0.05). No difference was observed in serum biomarker concentrations except NO, which increased in the 4th week (P < 0.05). The urinary concentrations of NO, MDA, PGC-1α, TGF-β1, NGAL, KIM-1, IL-18, and FGF 23 in the ACKD group were found to be higher compared to those in the non-azotemic group from the 1st to the 4th week (P < 0.05). In the ACKD group, the urine PGC-1α concentration in the 2nd (median 6.10 ng/mL) week was lower compared to that in the 0 and 1st (median 7.65 and 7.21 ng/mL, respectively) week, and the NO concentration in the 3rd (median 28.94 µmol/mL) week was lower than that in the 0th (median 37.43 µmol/mL) week (P < 0.05). CONCLUSIONS While sildenafil citrate has been determined to induce a low level of MB and to have a beneficial effect on glomerular filtration, it is observed to be ineffective in mitigating renal damage and fibrosis via the TGF-β/SMAD pathway in cats with ACKD.
Collapse
Affiliation(s)
- Mehmet Maden
- Faculty of Veterinary Medicine, Department of Internal Medicine, Selcuk University, Konya, 42250, Türkiye.
| | - Merve Ider
- Faculty of Veterinary Medicine, Department of Internal Medicine, Selcuk University, Konya, 42250, Türkiye
| | - Mehmet Erman Or
- Faculty of Veterinary Medicine, Internal Medicine Department, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Banu Dokuzeylül
- Faculty of Veterinary Medicine, Internal Medicine Department, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Erdem Gülersoy
- Faculty of Veterinary Medicine, Department of Internal Medicine, Harran University, Şanlıurfa, Türkiye
| | - Merve Cansu Kılıçkaya
- Faculty of Veterinary Medicine, Department of Internal Medicine, Selcuk University, Konya, 42250, Türkiye
| | - Bengü Bilgiç
- Faculty of Veterinary Medicine, Internal Medicine Department, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Murat Kaan Durgut
- Faculty of Veterinary Medicine, Department of Internal Medicine, Selcuk University, Konya, 42250, Türkiye
| | - Semih İzmirli
- Faculty of Veterinary Medicine, Internal Medicine Department, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Suleyman Serhat Iyigün
- Faculty of Veterinary Medicine, Department of Internal Medicine, Selcuk University, Konya, 42250, Türkiye
| | - Deniz Zeynep Telci
- Faculty of Veterinary Medicine, Internal Medicine Department, Istanbul University-Cerrahpasa, Istanbul, Türkiye
| | - Amir Naseri
- Faculty of Veterinary Medicine, Department of Internal Medicine, Selcuk University, Konya, 42250, Türkiye
| |
Collapse
|
21
|
Chiba I, Nakaya N, Kogure M, Hatanaka R, Nakaya K, Tokioka S, Nakamura T, Nagaie S, Ogishima S, Obara T, Sato T, Fuse N, Izumi Y, Kuriyama S, Hozawa A. Associations between leisure time, non-leisure time physical activity, and kidney function in Japanese adults: a cross-sectional study. BMC Nephrol 2024; 25:354. [PMID: 39415119 PMCID: PMC11484116 DOI: 10.1186/s12882-024-03813-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) contributes to decreased life expectancy. We examined the association between leisure-time physical activity (LTPA), non-leisure-time physical activity (non-LTPA) and kidney function. METHODS This was a cross-sectional study including 32 162 community-dwelling adults aged ≥ 20 years from the Tohoku Medical MegaBank community-based cohort study. Kidney function was evaluated using cystatin C-based estimated glomerular filtration rate (eGFR) as well as self-reported LTPA and non-LTPA. CKD was defined as either eGFR decline (≤ 60 mL/min/1.73 m2) or presence of albuminuria (albumin-creatinine ≥ 30 mg/g). The association between domain-specific physical activity and kidney function, and CKD prevalence was examined using multivariable-adjusted ordinary least squares and modified Poisson models. RESULTS The mean eGFR was 98.1 (± 13.2) mL/min/1.73 m2. 3 185 (9.9%) participants were classified as having CKD. The mean LTPA and non-LTPA levels were 2.9 (± 4.2) and 16.6 (± 14.2) METs-hour/day, respectively. For LTPA, in the adjusted model, the quartile groups with higher levels had a higher kidney function (β, 0.36; 95% confidence intervals [CI], [0.06, 0.66]; p = 0.019 for the 2nd quartile, β, 0.82; 95% CI, [0.51, 1.14]; p < 0.001 for the 3rd quartile, and β, 1.16; 95% CI, [0.83, 1.49]; p < 0.001 for the 4th quartile), whereas there were no apparent associations for prevalence of CKD. For non-LTPA, 4th quartile was associated with decreased eGFR (β, -0.42; 95% CI, [-0.72, -0.11]; p = 0.007) and higher prevalence of CKD prevalence (Prevalence ratio, 1.12; 95% CI, [1.02, 1.24]; p = 0.022). These associations with kidney function remained consistent in the subgroup analyses divided by demographic and biological variables. CONCLUSIONS We observed a positive association between higher LTPA levels and better kidney function, but not association with CKD prevalence. In contrast, higher non-LTPA was negatively associated with both kidney function and CKD prevalence. These findings suggest that promoting LTPA is beneficial for kidney function.
Collapse
Affiliation(s)
- Ippei Chiba
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan.
- Graduate School of Medicine, Tohoku University, Sendai, Japan.
| | - Naoki Nakaya
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Mana Kogure
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Rieko Hatanaka
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Kumi Nakaya
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Sayuri Tokioka
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Tomohiro Nakamura
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Faculty of Data Science, Kyoto Women's University, Kyoto, Japan
| | - Satoshi Nagaie
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Soichi Ogishima
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Taku Obara
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Toshimi Sato
- Department of Physical Therapy, Fukushima Medical University, Fukushima, Japan
| | - Nobuo Fuse
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yoko Izumi
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Shinichi Kuriyama
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Atsushi Hozawa
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
22
|
Michou V, Tsamos G, Vasdeki D, Deligiannis A, Kouidi E. Unraveling of Molecular Mechanisms of Cognitive Frailty in Chronic Kidney Disease: How Exercise Makes a Difference. J Clin Med 2024; 13:5698. [PMID: 39407758 PMCID: PMC11476541 DOI: 10.3390/jcm13195698] [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: 09/14/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
As our population ages, the medical challenges it faces become increasingly acute, with chronic kidney disease (CKD) becoming more prevalent among older adults. Frailty is alarmingly more common in CKD patients than in the general populace, putting the elderly at high risk of both physical and cognitive decline. CKD not only accelerates physical deterioration, but also heightens vascular dysfunction, calcification, arterial rigidity, systemic inflammation, oxidative stress, and cognitive impairment. Cognitive frailty, a distinct syndrome marked by cognitive deficits caused by physiological causes (excluding Alzheimer's and other dementias), is a critical concern. Although cognitive impairment has been well-studied, the molecular mechanisms driving cognitive frailty remain largely uncharted. Comprehensive interventions, including cutting-edge pharmaceuticals and lifestyle changes, are pivotal and effective, especially in the early stages of CKD. Recent research suggests that systematic exercise could counteract cognitive decline by improving brain blood flow, boosting neuroplasticity through the brain-derived neurotrophic factor (BDNF), and by triggering the release of neurotrophic factors such as insulin-like growth factor (IGF-1). This review delves into the molecular pathways of cognitive frailty in CKD, identifies key risk factors, and highlights therapeutic approaches, particularly the potent role of exercise in enhancing cognitive health.
Collapse
Affiliation(s)
- Vasiliki Michou
- Sports Medicine Laboratory, School of Physical Education & Sport Science, Aristotle University, 57 001 Thessaloniki, Greece; (A.D.); (E.K.)
| | - Georgios Tsamos
- Division of Endocrinology and Metabolism and Diabetes Centre, First Department of Internal Medicine, Medical School, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece; (G.T.); (D.V.)
| | - Dimitra Vasdeki
- Division of Endocrinology and Metabolism and Diabetes Centre, First Department of Internal Medicine, Medical School, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece; (G.T.); (D.V.)
| | - Asterios Deligiannis
- Sports Medicine Laboratory, School of Physical Education & Sport Science, Aristotle University, 57 001 Thessaloniki, Greece; (A.D.); (E.K.)
| | - Evangelia Kouidi
- Sports Medicine Laboratory, School of Physical Education & Sport Science, Aristotle University, 57 001 Thessaloniki, Greece; (A.D.); (E.K.)
| |
Collapse
|
23
|
Pan M, Li H, Shi X. A New Target for Hepatic Fibrosis Prevention and Treatment: The Warburg Effect. FRONT BIOSCI-LANDMRK 2024; 29:321. [PMID: 39344326 DOI: 10.31083/j.fbl2909321] [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: 05/13/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 10/01/2024]
Abstract
Hepatic fibrosis is a major public health problem that endangers human wellbeing. In recent years, a number of studies have revealed the important impact of metabolic reprogramming on the occurrence and development of hepatic fibrosis. Among them, the Warburg effect, as an intracellular glucose metabolism reprogramming, can promote the occurrence and development of hepatic fibrosis by promoting the activation of hepatic stellate cells (HSCs) and inducing the polarization of liver macrophages (KC). Understanding the Warburg effect and its important role in the progression of hepatic fibrosis will assist in developing new strategies for the prevention and treatment of hepatic fibrosis. This review focuses on the Warburg effect and the specific mechanism by which it affects the progression of hepatic fibrosis by regulating HSCs activation and KC polarization. In addition, we also summarize and discuss the related experimental drugs and their mechanisms that inhibit the Warburg effect by targeting key proteins of glycolysis in order to improve hepatic fibrosis in the hope of providing more effective strategies for the clinical treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Meng Pan
- College of Basic Medical Sciences, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| | - Huanyu Li
- Second Clinical Medical College, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| | - Xiaoyan Shi
- College of Basic Medical Sciences, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| |
Collapse
|
24
|
Rahman MM, Hwang SM, Go EJ, Kim YH, Park CK. Irisin alleviates CFA-induced inflammatory pain by modulating macrophage polarization and spinal glial cell activation. Biomed Pharmacother 2024; 178:117157. [PMID: 39042964 DOI: 10.1016/j.biopha.2024.117157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024] Open
Abstract
Although the potent anti-inflammatory effects of irisin have been documented in various inflammatory disorders, its efficacy against inflammatory pain remains unexplored. Herein, we examined the therapeutic effects of irisin in a mouse model of inflammatory pain induced by complete Freund's adjuvant (CFA). Mice were divided into three groups: normal control, CFA-injected (CFA), and CFA plus irisin-treated (CFA+Irisin). The irisin-treated group exhibited a gradual reduction in mechanical allodynia and thermal hyperalgesia when compared with the CFA group. Moreover, treatment with irisin significantly upregulated the expression of M2 macrophage markers (interleukin [IL]-4 and IL-10) and downregulated M1 macrophage markers (IL-1β, IL-6, and tumor necrosis factor-α) in the local paw tissue, dorsal root ganglion, and spinal cord tissue. However, there was no significant difference in the total number of F4/80+ macrophages in the paw tissue and dorsal root ganglion, indicating phenotypic exchange. Treatment with irisin also downregulated the expression of the glial cell activation-related markers Iba-1 and GFAP in the spinal cord tissue. To elucidate the underlying mechanisms, we detected the expression of Toll-like receptor 4 (TLR4), MyD88, and interferon regulatory factor 5 (IRF5) in paw tissues, dorsal root ganglion, and spinal tissues, revealing that irisin could downregulate the expression of these proteins. Irisin alleviated inflammatory pain by modulating local tissue inflammation and peripheral and central neuroinflammation and reducing glial cell activation and M2 macrophage polarization by modulating the TLR4-MyD88-IRF5 signaling pathway. Accordingly, irisin is a promising candidate for treating inflammatory pain in various diseases.
Collapse
Affiliation(s)
- Md Mahbubur Rahman
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, the Republic of Korea
| | - Sung-Min Hwang
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, the Republic of Korea
| | - Eun Jin Go
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, the Republic of Korea
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, the Republic of Korea.
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, the Republic of Korea.
| |
Collapse
|
25
|
Cui Y, Yu L, Cong W, Jiang S, Qiu X, Wei C, Zheng G, Mao J, Liu R, Patzak A, Persson PB, Chen J, Zhao L, Lai EY. Irisin preserves mitochondrial integrity and function in tubular epithelial cells after ischemia-reperfusion-induced acute kidney injury. Acta Physiol (Oxf) 2024; 240:e14211. [PMID: 39073055 DOI: 10.1111/apha.14211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 06/15/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024]
Abstract
AIMS A myokine secreted by skeletal muscles during exercise called irisin mitigates ischemia-reperfusion (I/R) injury in epithelial cells of various organs by limiting damage to mitochondria. We test whether irisin may preserve the mitochondrial integrity and function in renal tubular epithelial cells and protect against ischemia-reperfusion-induced acute kidney injury (AKI). METHODS We correlated serum irisin levels with serum creatinine and BUN levels from both AKI patients and healthy individuals. In mice with irisin administration, various renal injury markers such as serum creatinine, BUN, kidney injury molecule-1 (Kim-1), and neutrophil gelatinase-associated lipocalin (NGAL), and renal histopathology were assessed after I/R. To identify the potential mechanisms of the protective of irisin's protective effect, we perfused proximal tubules under confocal microscopy and analyzed kidney tissues by qPCR, western blot, and immunohistochemistry. RESULTS Serum irisin correlated inversely with serum creatinine and BUN levels were significantly lower in AKI patients than in healthy subjects. Administering irisin to mice after I/R decreased biomarker levels for AKI including serum creatinine, BUN, Kim-1, NAGL and lessened histological changes. In kidney tissues of mice, irisin upregulated the mitochondrial autophagy marker protein microtubule-associated protein 1 light chain 3 (LC3), the mitochondrial autophagy pathway-related proteins PTEN-induced putative kinase 1 (PINK1) and Parkinson's disease 2 parkin (PARK2) and downregulated the reactive substrate protein sequestosome 1 (P62) and mitochondrial membrane proteins translocase of outer mitochondrial membrane 20 (TOM20) and translocase of inner mitochondrial membrane 23 (TIM23). CONCLUSION Irisin protects against renal I/R injury, which may involve the preservation of mitochondrial integrity and function.
Collapse
Affiliation(s)
- Yu Cui
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Lu Yu
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Wenqi Cong
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Shan Jiang
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingyu Qiu
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunchun Wei
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Gui Zheng
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Jianhua Mao
- Provincial Key Laboratory of Neonatal Diseases, Department of Nephrology, National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruisheng Liu
- Department of Molecular Pharmacology & Physiology, Hypertension and Kidney Research Center, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Andreas Patzak
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pontus B Persson
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jianghua Chen
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Liang Zhao
- Provincial Key Laboratory of Neonatal Diseases, Department of Nephrology, National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - En Yin Lai
- Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Physiology, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
26
|
Nath KA, Griffin MD. CKD: A Potentially Self-Perpetuating Cycle of Injury to the Kidney and Other Organs. Mayo Clin Proc 2024; 99:1356-1359. [PMID: 39232621 DOI: 10.1016/j.mayocp.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 09/06/2024]
Affiliation(s)
- Karl A Nath
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Ireland.
| | - Matthew D Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Ireland
| |
Collapse
|
27
|
Wang Q, Sun X, Fang X, Wang Z, Wang H, Sun S, Wang S, Li T, Zhang P, Cheng Z. Dual-molecular targeting nanomedicine upregulates synergistic therapeutic efficacy in preclinical hepatoma models. Acta Biomater 2024; 183:306-317. [PMID: 38838902 DOI: 10.1016/j.actbio.2024.05.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/11/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
Advanced hepatocellular carcinoma (HCC) is one of the most challenging cancers because of its heterogeneous and aggressive nature, precluding the use of curative treatments. Sorafenib (SOR) is the first approved molecular targeting agent against the mitogen-activated protein kinase (MAPK) pathway for the noncurative therapy of advanced HCC; yet, any clinically meaningful benefits from the treatment remain modest, and are accompanied by significant side effects. Here, we hypothesized that using a nanomedicine platform to co-deliver SOR with another molecular targeting drug, metformin (MET), could tackle these issues. A micelle self-assembled with amphiphilic polypeptide methoxy poly(ethylene glycol)-block-poly(L-phenylalanine-co-l-glutamic acid) (mPEG-b-P(LP-co-LG)) (PM) was therefore designed for combinational delivery of two molecular targeted drugs, SOR and MET, to hepatomas. Compared with free drugs, the proposed, dual drug-loaded micelle (PM/SOR+MET) enhanced the drugs' half-life in the bloodstream and drug accumulation at the tumor site, thereby inhibiting tumor growth effectively in the preclinical subcutaneous, orthotopic and patient-derived xenograft hepatoma models without causing significant systemic and organ toxicity. Collectively, these findings demonstrate an effective dual-targeting nanomedicine strategy for treating advanced HCC, which may have a translational potential for cancer therapeutics. STATEMENT OF SIGNIFICANCE: Treatment of advanced hepatocellular carcinoma (HCC) remains a formidable challenge due to its aggressive nature and the limitations inherent to current therapies. Despite advancements in molecular targeted therapies, such as Sorafenib (SOR), their modest clinical benefits coupled with significant adverse effects underscore the urgent need for more efficacious and less toxic treatment modalities. Our research presents a new nanomedicine platform that synergistically combines SOR with metformin within a specialized diblock polypeptide micelle, aiming to enhance therapeutic efficacy while reducing systemic toxicity. This innovative approach not only exhibits marked antitumor efficacy across multiple HCC models but also significantly reduces the toxicity associated with current treatments. Our dual-molecular targeting approach unveils a promising nanomedicine strategy for the molecular treatment of advanced HCC, potentially offering more effective and safer treatment alternatives with significant translational potential.
Collapse
Affiliation(s)
- Qilong Wang
- Department of Vascular Surgery, General Surgery Center, the First Hospital of Jilin University, Changchun, 130061, PR China
| | - Xiwei Sun
- Department of Vascular Surgery, General Surgery Center, the First Hospital of Jilin University, Changchun, 130061, PR China
| | - Xizhu Fang
- Department of Immunology and Pathogenic Biology, College of Medicine, Yanbian University, Yanji, 133002, PR China
| | - Zhongying Wang
- Department of Vascular Surgery, General Surgery Center, the First Hospital of Jilin University, Changchun, 130061, PR China
| | - Haodong Wang
- Department of Vascular Surgery, General Surgery Center, the First Hospital of Jilin University, Changchun, 130061, PR China
| | - Siqiao Sun
- Department of Vascular Surgery, General Surgery Center, the First Hospital of Jilin University, Changchun, 130061, PR China
| | - Shuai Wang
- Department of Vascular Surgery, General Surgery Center, the First Hospital of Jilin University, Changchun, 130061, PR China
| | - Tingting Li
- Department of Vascular Surgery, General Surgery Center, the First Hospital of Jilin University, Changchun, 130061, PR China
| | - Ping Zhang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, the First Hospital of Jilin University, Changchun, 130061, PR China.
| | - Zhihua Cheng
- Department of Vascular Surgery, General Surgery Center, the First Hospital of Jilin University, Changchun, 130061, PR China.
| |
Collapse
|
28
|
Li H, Qin S, Tang J, Wang T, Ren W, Di L, Bo W, Ma Y, Wu F, Xu Z, Song W, Cai M, Xi Y, Tian Z. Resistance exercise upregulates Irisin expression and suppresses myocardial fibrosis following myocardial infarction via activating AMPK-Sirt1 and inactivating TGFβ1-Smad2/3. Acta Physiol (Oxf) 2024; 240:e14163. [PMID: 38752665 DOI: 10.1111/apha.14163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Abstract
AIM To reveal the contribution of Irisin in the beneficial effects of resistance exercise on myocardial fibrosis (MF) and cardiac function in the mice with myocardial infarction (MI). METHODS The MI model was built by ligating the left anterior descending coronary artery in Fndc5 knockout mice (Fndc5-/-). Resistance exercise was started one week after surgery and continued for four weeks. In addition, H2O2, AICAR, recombinant human Irisin protein (rhIRISIN), and Sirt1 shRNA lentivirus (LV-Sirt1 shRNA) were used to intervene primary isolated cardiac fibroblasts (CFs). MF was observed through Masson staining, and apoptosis was assessed using TUNEL staining. MDA and T-SOD contents were detected by biochemical kits. The expression of proteins and genes was detected by Western blotting and RT-qPCR. RESULTS Resistance exercise increased Fndc5 mRNA level, inhibited the activation of TGFβ1-TGFβR2-Smad2/3 pathway, activated AMPK-Sirt1 pathway, reduced the levels of oxidative stress, apoptosis, and MF in the infarcted heart, and promoted cardiac function. However, Fndc5 knockout attenuated the protective effects of resistance exercise on the MI heart. Results of the in vitro experiments showed that AICAR and rhIRISIN intervention activated the AMPK-Sirt1 pathway and inactivated the TGFβ1-Smad2/3 pathway, and promoted apoptosis in H2O2-treated CFs. Notably, these effects of rhIRISIN intervention, except for the TGFβR2 expression, were attenuated by LV-Sirt1 shRNA. CONCLUSION Resistance exercise upregulates Fndc5 expression, activates AMPK-Sirt1 pathway, inhibits the activation of TGFβ1-Smad2/3 pathway, attenuates MF, and promotes cardiac function after MI.
Collapse
Affiliation(s)
- Hangzhuo Li
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Shuguang Qin
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Jie Tang
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Tao Wang
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Wujing Ren
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Lingyun Di
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Wenyan Bo
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Yixuan Ma
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Fangnan Wu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Zujie Xu
- The Department of Physical Education, School of Physical Education, Taiyuan University of Technology, Taiyuan, China
| | - Wei Song
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Mengxin Cai
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Yue Xi
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Zhenjun Tian
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
29
|
Wang Z, Dong S, Zhou W. Pancreatic stellate cells: Key players in pancreatic health and diseases (Review). Mol Med Rep 2024; 30:109. [PMID: 38695254 PMCID: PMC11082724 DOI: 10.3892/mmr.2024.13233] [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: 01/31/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
As a pluripotent cell, activated pancreatic stellate cells (PSCs) can differentiate into various pancreatic parenchymal cells and participate in the secretion of extracellular matrix and the repair of pancreatic damage. Additionally, PSCs characteristics allow them to contribute to pancreatic inflammation and carcinogenesis. Moreover, a detailed study of the pathogenesis of activated PSCs in pancreatic disease can offer promise for the development of innovative therapeutic strategies and improved patient prognoses. Therefore, the present study review aimed to examine the involvement of activated PSCs in pancreatic diseases and elucidate the underlying mechanisms to provide a viable therapeutic strategy for the management of pancreas‑related diseases.
Collapse
Affiliation(s)
- Zhengfeng Wang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Shi Dong
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Wence Zhou
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
30
|
Wang Y, Deng X, Wei J, Yang Z, Du Y, Song S, Shi Y, Wu H. Irisin ameliorates UUO-induced renal interstitial fibrosis through TGF-β1/periostin/MMP-2 signaling pathway. PLoS One 2024; 19:e0299389. [PMID: 38870184 PMCID: PMC11175535 DOI: 10.1371/journal.pone.0299389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/08/2024] [Indexed: 06/15/2024] Open
Abstract
Renal fibrosis is the most common pathway in progressive kidney diseases. The unilateral ureteral obstruction (UUO) model is used to induce progressive renal fibrosis. We evaluated the effects of irisin on renal interstitial fibrosis in UUO mice. The GSE121190, GSE36496, GSE42303, and GSE96101 datasets were downloaded from the Gene Expression Omnibus (GEO) database. In total, 656 differentially expressed genes (DEGs) were identified in normal and UUO mouse renal samples. Periostin and matrix metalloproteinase-2 (MMP-2) were selected to evaluate the effect of irisin on renal fibrosis in UUO mice. In UUO mice, irisin ameliorated renal function, decreased the expression of periostin and MMP-2, and attenuated epithelial-mesenchymal transition and extracellular matrix deposition in renal tissues. In HK-2 cells, irisin treatment markedly attenuated TGF-β1-induced expression of periostin and MMP-2. Irisin treatment also inhibited TGF-β1-induced epithelial-mesenchymal transition, extracellular matrix formation, and inflammatory responses. These protective effects of irisin were abolished by the overexpression of periostin and MMP-2. In summary, irisin treatment can improve UUO-induced renal interstitial fibrosis through the TGF-β1/periostin/MMP-2 signaling pathway, suggesting that irisin may be used for the treatment of renal interstitial fibrosis.
Collapse
Affiliation(s)
- Yashu Wang
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Xinna Deng
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Jinying Wei
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Zhaohua Yang
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Yunxia Du
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Shan Song
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Yonghong Shi
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Haijiang Wu
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
31
|
Malheiro LFL, Fernandes MM, Oliveira CA, Barcelos IDS, Fernandes AJV, Silva BS, Ávila JS, Soares TDJ, Amaral LSDB. Renoprotective mechanisms of exercise training against acute and chronic renal diseases - A perspective based on experimental studies. Life Sci 2024; 346:122628. [PMID: 38614303 DOI: 10.1016/j.lfs.2024.122628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 03/22/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Regular exercise training can lead to several health benefits, reduce mortality risk, and increase life expectancy. On the other hand, a sedentary lifestyle is a known risk factor for chronic diseases and increased mortality. Acute kidney injury (AKI) and chronic kidney disease (CKD) represent a significant global health problem, affecting millions of people worldwide. The progression from AKI to CKD is well-recognized in the literature, and exercise training has emerged as a potential renoprotective strategy. Thus, this article aims to review the main molecular mechanisms underlying the renoprotective actions of exercise training in the context of AKI and CKD, focusing on its antioxidative, anti-inflammatory, anti-apoptotic, anti-fibrotic, and autophagy regulatory effects. For that, bibliographical research was carried out in Medline/PubMed and Scielo databases. Although the pathophysiological mechanisms involved in renal diseases are not fully understood, experimental studies demonstrate that oxidative stress, inflammation, apoptosis, and dysregulation of fibrotic and autophagic processes play central roles in the development of tissue damage. Increasing evidence has suggested that exercise can beneficially modulate these mechanisms, potentially becoming a safe and effective non-pharmacological strategy for kidney health protection and promotion. Thus, the evidence base discussed in this review suggests that an adequate training program emerges as a valuable tool for preserving renal function in experimental animals, mainly through the production of antioxidant enzymes, nitric oxide (NO), irisin, IL-10, and IL-11. Future research can continue to explore these mechanisms to develop specific guidelines for the prescription of exercise training in different populations of patients with kidney diseases.
Collapse
Affiliation(s)
- Lara Fabiana Luz Malheiro
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil; Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Brazil
| | - Mariana Masimessi Fernandes
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil
| | - Caroline Assunção Oliveira
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil; Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Brazil
| | - Isadora de Souza Barcelos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil
| | - Ana Jullie Veiga Fernandes
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil
| | - Bruna Santos Silva
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil
| | - Júlia Spínola Ávila
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil
| | - Telma de Jesus Soares
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil; Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Brazil; Programa de Pós-Graduação em Biociências, Brazil
| | - Liliany Souza de Brito Amaral
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Bahia 45029-094, Brazil; Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Brazil; Programa de Pós-Graduação em Biociências, Brazil.
| |
Collapse
|
32
|
Picciotto D, Macciò L, Verzola D, Baciga F, Momentè C, Russo E, Viazzi F, Battaglia Y, Esposito P. Pathophysiology of Physical Exercise in Kidney Patients: Unveiling New Players - The Role of Myokines. Kidney Blood Press Res 2024; 49:457-471. [PMID: 38815556 DOI: 10.1159/000539489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is a progressive systemic condition characterized by numerous complications. Among these, alterations in skeletal muscle physiology, such as sarcopenia, are particularly significant, as they are associated with poor outcomes and reduced quality of life. SUMMARY Various interventions, including pharmacological approaches and lifestyle modifications have been investigated to slow CKD progression and prevent or treat its complications. Physical exercise, in particular, has emerged as a promising intervention with multiple beneficial effects. These include improvements in physical functioning, increased muscle mass, modulation of metabolic abnormalities, and reduced cardiovascular risk. However, the pathophysiology of physical exercise in patients with kidney disease is complex and remains only partially understood. A crucial advancement in understanding this phenomenon has been the identification of myokines - molecules expressed and released by skeletal muscle in response to physical activity. These myokines can exert both paracrine and systemic effects, influencing not only skeletal muscle physiology but also other processes such as energy metabolism and lipid regulation. KEY MESSAGES The interplay among skeletal muscle, physical activity, and myokines may act as a pivotal regulator in various physiological processes, including aging, as well as in pathological conditions like cachexia and sarcopenia, frequently observed in CKD patients at different stages, including patients on dialysis. Despite the potential importance of this relationship, only a limited number of studies have explored the relationship between exercise and myokine, and the effect of this interaction on experimental models or individuals with kidney disease. In the following sections, we review and discuss this topic.
Collapse
Affiliation(s)
- Daniela Picciotto
- Nephrology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lucia Macciò
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genoa, Italy
| | - Daniela Verzola
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genoa, Italy
| | - Federica Baciga
- Department of Medicine, University of Verona, Verona, Italy
- Nephrology and Dialysis Unit, Pederzoli Hospital, Peschiera del Garda, Italy
| | | | - Elisa Russo
- Nephrology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genoa, Italy
| | - Francesca Viazzi
- Nephrology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genoa, Italy
| | - Yuri Battaglia
- Department of Medicine, University of Verona, Verona, Italy
- Nephrology and Dialysis Unit, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Pasquale Esposito
- Nephrology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genoa, Italy
| |
Collapse
|
33
|
Cho JM, Koh JH, Kim SG, Lee S, Kim Y, Cho S, Kim K, Kim YC, Han SS, Lee H, Lee JP, Joo KW, Lim CS, Kim YS, Kim DK, Park S. Associations of MRI-derived kidney volume, kidney function, body composition and physical performance in ≈38 000 UK Biobank participants: a population-based observational study. Clin Kidney J 2024; 17:sfae068. [PMID: 38660121 PMCID: PMC11040514 DOI: 10.1093/ckj/sfae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Indexed: 04/26/2024] Open
Abstract
Background Kidney volume is used as a predictive and therapeutic marker for several clinical conditions. However, there is a lack of large-scale studies examining the relationship between kidney volume and various clinicodemographic factors, including kidney function, body composition and physical performance. Methods In this observational study, MRI-derived kidney volume measurements from 38 526 UK Biobank participants were analysed. Major kidney volume-related measures included body surface area (BSA)-adjusted total kidney volume (TKV) and the difference in bilateral kidneys. Multivariable-adjusted linear regression and cubic spline analyses were used to explore the association between kidney volume-related measures and clinicodemographic factors. Cox or logistic regression was used to identify the risks of death, non-kidney cancer, myocardial infarction, ischaemic stroke and chronic kidney disease (CKD). Results The median of BSA-adjusted TKV and the difference in kidney volume were 141.9 ml/m2 [interquartile range (IQR) 128.1-156.9] and 1.08-fold (IQR 1.04-1.15), respectively. Higher BSA-adjusted TKV was significantly associated with higher estimated glomerular filtration rate {eGFR; β = 0.43 [95% confidence interval (CI) 0.42-0.44]; P < .001}, greater muscle volume [β = 0.50 (95% CI 0.48-0.51); P < .001] and greater mean handgrip strength [β = 0.15 (95% CI 0.13-0.16); P < .001] but lower visceral adipose tissue volume [VAT; β = -0.09 (95% CI -0.11 to -0.07); P < .001] in adjusted models. A greater difference in bilateral kidney volumes was associated with lower eGFR, muscle volume and physical performance but with higher proteinuria and VAT. Higher BSA-adjusted TKV was significantly associated with a reduced risk of CKD [odds ratio (OR) 0.7 (95% CI 0.63-0.77); P < .001], while a greater difference in kidney volume was significantly associated with an increased risk of CKD [OR 1.13 (95% CI 1.07-1.20); P < .001]. Conclusion Higher BSA-adjusted TKV and lower differences in bilateral kidney volumes are associated with higher kidney function, muscle volume and physical performance and a reduced risk of CKD.
Collapse
Affiliation(s)
- Jeong Min Cho
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Chung-Ang University Gwangmyeong Hospital, Gyeonggi-do, Korea
| | - Jung Hun Koh
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seong Geun Kim
- Department of Internal Medicine, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - Soojin Lee
- Department of Internal Medicine, Uijeongbu Eulji University Medical Center, Uijeongbu, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yaerim Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Semin Cho
- Department of Internal Medicine, Chung-Ang University Gwangmyeong Hospital, Gyeonggi-do, Korea
| | - Kwangsoo Kim
- Transdisciplinary Department of Medicine & Advanced Technology, Seoul National University Hospital, Seoul, Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Sehoon Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
34
|
Zhang J, Li H, Zhong H, Chen X, Hu ZX. Omega-3 polyunsaturated fatty acids protect peritoneal mesothelial cells from hyperglycolysis and mesothelial-mesenchymal transition through the FFAR4/CaMKKβ/AMPK/mTOR signaling pathway. Int Immunopharmacol 2024; 128:111561. [PMID: 38262160 DOI: 10.1016/j.intimp.2024.111561] [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: 12/24/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/25/2024]
Abstract
Peritoneal fibrosis is a severe clinical complication associated with peritoneal dialysis (PD) and impacts its efficacy and patient outcomes. The process of mesothelial-mesenchymal transition (MMT) in peritoneal mesothelial cells plays a pivotal role in fibrogenesis, whereas metabolic reprogramming, characterized by excessive glycolysis, is essential in MMT development. No reliable therapies are available despite substantial progress made in understanding the mechanisms underlying peritoneal fibrosis. Protective effect of omega-3 polyunsaturated fatty acids (ω3 PUFAs) has been described in PD-induced peritoneal fibrosis, although the detailed mechanisms remain unknown. It is known that ω3 PUFAs bind to and activate the free fatty acid receptor 4 (FFAR4). However, the expression and role of FFAR4 in the peritoneum have not been investigated. Thus, we hypothesized that ω3 PUFAs would alleviate peritoneal fibrosis by inhibiting hyperglycolysis and MMT through FFAR4 activation. First, we determined FFAR4 expression in peritoneal mesothelium in humans and mice. FFAR4 expression was abnormally decreased in patients on PD and mice and HMrSV5 mesothelial cells exposed to PD fluid (PDF); this change was restored by the ω3 PUFAs (EPA and DHA). ω3 PUFAs significantly inhibited peritoneal hyperglycolysis, MMT, and fibrosis in PDF-treated mice and HMrSV5 mesothelial cells; these changes induced by ω3 PUFAs were blunted by treatment with the FFAR4 antagonist AH7614 and FFAR4 siRNA. Additionally, ω3 PUFAs induced FFAR4, Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ), and AMPK and suppressed mTOR, leading to the inhibition of hyperglycolysis, demonstrating that the ω3 PUFAs-mediated FFAR4 activation ameliorated peritoneal fibrosis by inhibiting hyperglycolysis and MMT via CaMKKβ/AMPK/mTOR signaling. As natural FFAR4 agonists, ω3 PUFAs may be considered for the treatment of PD-associated peritoneal fibrosis.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Li
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Zhong
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhang-Xue Hu
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China; National Clinical Research Center for Geriatrics and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
35
|
Wang S, Hu S, Pan Y. The emerging roles of irisin in vascular calcification. Front Endocrinol (Lausanne) 2024; 15:1337995. [PMID: 38405155 PMCID: PMC10884194 DOI: 10.3389/fendo.2024.1337995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/19/2024] [Indexed: 02/27/2024] Open
Abstract
Vascular calcification is a common accompanying pathological change in many chronic diseases, which is caused by calcium deposition in the blood vessel wall and leads to abnormal blood vessel function. With the progress of medical technology, the diagnosis rate of vascular calcification has explosively increased. However, due to its mechanism's complexity, no effective drug can relieve or even reverse vascular calcification. Irisin is a myogenic cytokine regulating adipose tissue browning, energy metabolism, glucose metabolism, and other physiological processes. Previous studies have shown that irisin could serve as a predictor for vascular calcification, and protect against hypertension, diabetes, chronic kidney disease, and other risk factors for vascular calcification. In terms of mechanism, it improves vascular endothelial dysfunction and phenotypic transformation of vascular smooth muscle cells. All the above evidence suggests that irisin plays a predictive and protective role in vascular calcification. In this review, we summarize the association of irisin to the related risk factors for vascular calcification and mainly explore the role of irisin in vascular calcification.
Collapse
Affiliation(s)
- Shuangshuang Wang
- Department of Cardiology, The First People’s Hospital of Wenling (The Affiliated Wenling Hospital of Wenzhou Medical University), Wenling, Zhejiang, China
| | - Siwang Hu
- The Orthopaedic Center, The First People’s Hospital of Wenling (The Affiliated Wenling Hospital of Wenzhou Medical University), Wenling, Zhejiang, China
| | - Yuping Pan
- Department of Internal Medicine, Yuhuan Second People’s Hospital, Yuhuan, Zhejiang, China
| |
Collapse
|
36
|
Li X, Lindholm B. The role of irisin in kidney diseases. Clin Chim Acta 2024; 554:117756. [PMID: 38218331 DOI: 10.1016/j.cca.2023.117756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 12/31/2023] [Accepted: 12/31/2023] [Indexed: 01/15/2024]
Abstract
Irisin is a hormone that is produced mainly by skeletal muscles in response to exercise. It has been found to have a close correlation with obesity and diabetes mellitus for its energy expenditure and metabolic properties. Recent research has revealed that irisin also possesses anti-inflammatory, anti-oxidative and anti-apoptotic properties, which make it associated with major chronic diseases, such as chronic kidney disease (CKD), liver diseases, osteoporosis, atherosclerosis and Alzheimer s disease. The identification of irisin has not only opened up new possibilities for monitoring metabolic and non-metabolic diseases but also presents a promising therapeutic target due to its multiple biological functions. Studies have shown that circulating irisin levels are lower in CKD patients than in non-CKD patients and decrease with increasing CKD stage. Furthermore, irisin also plays a role in many CKD-related complications like protein energy wasting (PEW), cardiovascular disease (CVD) and chronic kidney disease-mineral and bone disorder (CKD-MBD). In this review, we present the current knowledge on the role of irisin in kidney diseases and their complications.
Collapse
Affiliation(s)
- Xiejia Li
- Department of Nephrology, The 2nd Xiangya Hospital, Central South University, Changsha, Hunan, China; Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Bengt Lindholm
- Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Zhao ZW, Lin XX, Guo YZ, He X, Zhang XT, Huang Y. Irisin alleviates hyperoxia-induced bronchopulmonary dysplasia through activation of Nrf2/HO-1 pathway. Peptides 2023; 170:171109. [PMID: 37804931 DOI: 10.1016/j.peptides.2023.171109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/19/2023] [Accepted: 10/02/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a common pulmonary injury among premature infants, which is often caused by hyperoxia exposure. Irisin is a novel hormone-like myokine derived mainly from skeletal muscles as well as adipose tissues. Many studies have indicated that Irisin exert a variety of properties against hyperoxia-induced inflammation and oxidative stress (OS). We aimed to evaluate the effects of irisin on hyperoxia-induced lung injury explore the underlying mechanisms. METHODS BPD model was established after exposing newborn mouse to 85% oxygen. BPD mouse received continuous intraperitoneal injection of irisin at a dose of 25 μg/kg/day. Lung tissues were collected for histological examination at 7 and 14 days after birth. The alveolarization and alveolar vascularization of each animal was assessed. Levels of oxidative stress indicators, and the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) in lung tissues were detected at 14 days after birth. RESULTS Hyperoxia exposure induced a markedly alveolar simplification and a disrupted alveolar angiogenesis, which was ameliorated by irisin treatment. The hyperoxia-induced increase in these oxidative stress indicators was significantly reversed by irisin treatment. The Nrf2/HO-1 pathway is inducted in the hyperoxia-induced BPD mouse model, which is further activated by irisin treatment. CONCLUSION Our results demonstrated the beneficial effects of irisin in reducing the OS, enhancing alveolarization, and promoting vascular development through activation of Nrf2/HO-1 axis in a hyperoxia-induced experimental model of BPD.
Collapse
Affiliation(s)
- Zi-Wen Zhao
- Department of Cardiology, Fujian Heart Medical Center, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, PR China; Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Xiao-Xia Lin
- Department of Pediatrics, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, PR China
| | - Yong-Zhe Guo
- Department of Cardiology, Fujian Heart Medical Center, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, PR China
| | - Xi He
- Department of Cardiology, Fujian Heart Medical Center, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, PR China
| | - Xin-Tao Zhang
- Department of Cardiology, Fujian Heart Medical Center, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, PR China
| | - Yu Huang
- Department of Cardiology, Fujian Heart Medical Center, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, PR China.
| |
Collapse
|
38
|
Price SR, Mitch WE, Garibotto G. Muscle Atrophy in CKD: A Historical Perspective of Advancements in Its Understanding. J Ren Nutr 2023; 33:S88-S92. [PMID: 36183901 DOI: 10.1053/j.jrn.2022.09.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/21/2022] [Accepted: 09/21/2022] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVE This perspective reviews the seminal clinical and experimental observations that led to today's current mechanistic model of muscle protein loss (wasting) in patients with chronic kidney disease (CKD). RESULTS AND CONCLUSION Early International Society of Renal Nutrition and Metabolism (ISRNM) meetings facilitated discussions and hypotheses about the causes of muscle wasting in CKD. It became widely recognized that wasting is common and correlated with increased risks of mortality and morbidity. Although anorexia and dietary restrictions contribute to muscle loss, several features of CKD-associated wasting cannot be explained by malnutrition alone. The protein catabolism-inducing actions of metabolic acidosis, inflammation, insulin resistance, endocrine disorders and uremic toxins were progressively identified. Continued research to understand the interactions of inflammation, anabolic resistance, mitochondrial dysfunction, exercise, and nutrition on muscle protein turnover in patients with CKD will hopefully accelerate discoveries and treatments to ameliorate muscle wasting as well as the progression of CKD.
Collapse
Affiliation(s)
- S Russ Price
- Department of Biochemistry & Molecular Biology, Brody School of Medicine at East Carolina University, Greenville, North Carolina; Department of Internal Medicine, Brody School of Medicine at East Carolina University, Greenville, North Carolina.
| | - William E Mitch
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | | |
Collapse
|
39
|
Si M, Yu R, Lin H, Li F, Jung S, Thomas SS, Danesh FS, Wang Y, Peng H, Hu Z. ROCK1 activates mitochondrial fission leading to oxidative stress and muscle atrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.22.563469. [PMID: 37905139 PMCID: PMC10614981 DOI: 10.1101/2023.10.22.563469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Chronic kidney disease (CKD) is often associated with protein-energy wasting (PEW), which is characterized by a reduction in muscle mass and strength. Although mitochondrial dysfunction and oxidative stress have been implicated to play a role in the pathogenesis of muscle wasting, the underlying mechanisms remain unclear. In this study, we used transcriptomics, metabolomics analyses and mouse gene manipulating approaches to investigate the effects of mitochondrial plasticity and oxidative stress on muscle wasting in mouse CKD models. Our results showed that the expression of oxidative stress response genes was increased, and that of oxidative phosphorylation genes was decreased in the muscles of mice with CKD. This was accompanied by reduced oxygen consumption rates, decreased levels of mitochondrial electron transport chain proteins, and increased cellular oxidative damage. Excessive mitochondrial fission was also observed, and we found that the activation of ROCK1 was responsible for this process. Inducible expression of muscle-specific constitutively active ROCK1(mROCK1ca)exacerbated mitochondrial fragmentation and muscle wasting in CKD mice. Conversely, ROCK1 depletion (ROCK1-/-) alleviated these phenomena. Mechanistically, ROCK1 activation promoted the recruitment of Drp1 to mitochondria, thereby facilitating fragmentation. Notably, the pharmacological inhibition of ROCK1 mitigated muscle wasting by suppressing mitochondrial fission and oxidative stress. Our findings demonstrate that ROCK1 participates in CKD-induced muscle wasting by promoting mitochondrial fission and oxidative stress, and pharmacological suppression of ROCK1 could be a therapeutic strategy for combating muscle wasting in CKD conditions.
Collapse
Affiliation(s)
- Meijun Si
- Nephrology Division, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Nephrology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences; Guangzhou, China
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Rizhen Yu
- Nephrology Division, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Hangzhou, Zhejiang, China
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Hongchun Lin
- Nephrology Division, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Feng Li
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Sungyun Jung
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Sandhya S. Thomas
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Farhard S Danesh
- Nephrology Division, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yanlin Wang
- Division of Nephrology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Hui Peng
- Nephrology Division, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Zhaoyong Hu
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
40
|
Nørregaard R, Mutsaers HAM, Frøkiær J, Kwon TH. Obstructive nephropathy and molecular pathophysiology of renal interstitial fibrosis. Physiol Rev 2023; 103:2827-2872. [PMID: 37440209 PMCID: PMC10642920 DOI: 10.1152/physrev.00027.2022] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/14/2023] Open
Abstract
The kidneys play a key role in maintaining total body homeostasis. The complexity of this task is reflected in the unique architecture of the organ. Ureteral obstruction greatly affects renal physiology by altering hemodynamics, changing glomerular filtration and renal metabolism, and inducing architectural malformations of the kidney parenchyma, most importantly renal fibrosis. Persisting pathological changes lead to chronic kidney disease, which currently affects ∼10% of the global population and is one of the major causes of death worldwide. Studies on the consequences of ureteral obstruction date back to the 1800s. Even today, experimental unilateral ureteral obstruction (UUO) remains the standard model for tubulointerstitial fibrosis. However, the model has certain limitations when it comes to studying tubular injury and repair, as well as a limited potential for human translation. Nevertheless, ureteral obstruction has provided the scientific community with a wealth of knowledge on renal (patho)physiology. With the introduction of advanced omics techniques, the classical UUO model has remained relevant to this day and has been instrumental in understanding renal fibrosis at the molecular, genomic, and cellular levels. This review details key concepts and recent advances in the understanding of obstructive nephropathy, highlighting the pathophysiological hallmarks responsible for the functional and architectural changes induced by ureteral obstruction, with a special emphasis on renal fibrosis.
Collapse
Affiliation(s)
- Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Jørgen Frøkiær
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea
| |
Collapse
|
41
|
Tsutsui Y, Mori T, Yoshio S, Sato M, Sakata T, Yoshida Y, Kawai H, Yoshikawa S, Yamazoe T, Matsuda M, Kakazu E, Osawa Y, Oyama C, Tamura-Nakano M, Kawaguchi T, Yoshizumi T, Kanto T. Exercise changes the intrahepatic immune cell profile and inhibits the progression of nonalcoholic steatohepatitis in a mouse model. Hepatol Commun 2023; 7:e0236. [PMID: 37756046 PMCID: PMC10531194 DOI: 10.1097/hc9.0000000000000236] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 06/10/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND NASH is an increasingly common cause of chronic liver disease and can progress to cirrhosis and HCC. Although exercise suppresses inflammation during acute hepatitis, its impact on the progression of chronic liver disease remains unclear. Here, we investigated the effects of exercise on disease progression and intrahepatic immune cell composition in a mouse model of NASH. METHOD Mice were assigned to 4 groups: 2 control groups (normal diet) and 2 NASH groups (western diet and low-dose carbon tetrachloride injection). One of each group remained sedentary and one was exercised on a treadmill for 12 weeks (60 min/d, 5 times/wk). All mice were then analyzed for liver histomorphology, steatosis, inflammation, and fibrosis; liver, adipose tissue, and skeletal muscle expression of genes related to metabolism and inflammation; and intrahepatic immune cell composition. RESULT Compared with the normal diet mice, NASH mice exhibited enhanced liver steatosis, inflammation, and fibrosis; upregulated expression of liver lipogenesis-related and inflammation-related genes; and increased frequencies of intrahepatic F4/80 int CD11b hi bone marrow-derived macrophages and programmed death receptor-1 (PD-1) + CD8 + T cells. Expression of inflammatory cytokines and the frequencies of bone marrow-derived macrophages and PD-1 + CD8 + T cells correlated positively with liver steatosis, inflammation, and fibrosis. Exercise was shown to reduce NASH-induced hepatic steatosis, liver inflammation, and fibrosis; induce alterations in metabolism-related genes and inflammatory cytokines in the liver; and suppress accumulation of liver bone marrow-derived macrophages and PD-1 + CD8 + T cells. In addition, we showed that exercise induced increased expression of IL-15 in muscle and its deficiency exacerbated the pathology of NASH. CONCLUSIONS Exercise alters the intrahepatic immune cell profile and protects against disease progression in a mouse model of NASH.
Collapse
Affiliation(s)
- Yuriko Tsutsui
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taizo Mori
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Sachiyo Yoshio
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Miku Sato
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Toshihiro Sakata
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Yuichi Yoshida
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Hironari Kawai
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Shiori Yoshikawa
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Taiji Yamazoe
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Michitaka Matsuda
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Eiji Kakazu
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Yosuke Osawa
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
- Department of Gastroenterology, International University of Health and Welfare Hospital, Tochigi, Japan
| | - Chinatsu Oyama
- Communal Laboratory, National Center for Global Health and Medicine, Tokyo, Japan
| | - Miwa Tamura-Nakano
- Communal Laboratory, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takumi Kawaguchi
- Department of Medicine, Division of Gastroenterology, Kurume University School of Medicine, Kurume, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsuya Kanto
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| |
Collapse
|
42
|
Lai W, Luo D, Li Y, Li Y, Wang Q, Hu Z, Ye Z, Peng H. Irisin ameliorates diabetic kidney disease by restoring autophagy in podocytes. FASEB J 2023; 37:e23175. [PMID: 37742293 DOI: 10.1096/fj.202300420r] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 07/21/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023]
Abstract
Many studies have highlighted the importance of moderate exercise. While it can attenuate diabetic kidney disease, its mechanism has remained unclear. The level of myokine irisin in plasma increases during exercise. We found that irisin was decreased in diabetic patients and was closely related to renal function, proteinuria, and podocyte autophagy injury. Muscle-specific overexpression of PGC-1α (mPGC-1α) in a mouse model is known to increase plasma irisin levels. The mPGC-1α mice were crossed with db/m mice to obtain db/db mPGC-1α+ mice in the present study. Compared to db/db mice without mPGC-1α, plasma irisin was increased, and albuminuria and glomerular pathological damage were both alleviated in db/db mPGC-1α+ mice. Impaired autophagy in podocytes was restored as well. Irisin inhibited the activation of the PI3K/AKT/mTOR signaling pathway in cultured human podocytes and improved damaged autophagy induced by high glucose levels. Then, db/db mice were treated with recombinant irisin, which had similar beneficial effects on the kidney as those in db/db mPGC-1α+ mice, with alleviated glomerular injury and albuminuria. Moreover, the autophagy in podocytes was also significantly restored. These results suggest that irisin secreted by skeletal muscles protects the kidney from diabetes mellitus damage. It also restores autophagy in podocytes by inhibiting the abnormal activation of the PI3K/AKT/mTOR signaling pathway. Thus, irisin may become a new drug for the prevention and treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Weiyan Lai
- Nephrology Division, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dan Luo
- Nephrology Division, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yin Li
- Nephrology Division, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuanqing Li
- Nephrology Division, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qianqian Wang
- Nephrology Division, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhaoyong Hu
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Zengchun Ye
- Nephrology Division, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hui Peng
- Nephrology Division, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
43
|
Hori T, Yokobori K, Moore R, Negishi M, Sueyoshi T. CAR requires Gadd45β to promote phenobarbital-induced mouse liver tumors in early stage. Front Oncol 2023; 13:1217847. [PMID: 37746289 PMCID: PMC10516603 DOI: 10.3389/fonc.2023.1217847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
Phenobarbital (PB) is an archetypal substance used as a mouse hepatocellular carcinoma (HCC) promotor in established experimental protocols. Our previous results showed CAR is the essential factor for PB induced HCC promotion. Subsequent studies suggested Gadd45β, which is induced by PB through CAR activation, is collaborating with CAR to repress TNF-α induced cell death. Here, we used Gadd45β null mice (Gadd45β KO) treated with N-diethylnitrosamine (DEN) at 5 weeks of age and kept the mice with PB supplemented drinking water from 7 to 57 weeks old. Compared with wild type mice, Gadd45β KO mice developed no HCC in the PB treated group. Increases in liver weight were more prominent in wild type mice than KO mice. Microarray analysis of mRNA derived from mouse livers found multiple genes specifically up or down regulated in wild type mice but not null mice in DEN + PB groups. Further qPCR analysis confirmed two genes, Tgfbr2 and irisin/Fndc5, were up-regulated in PB treated wild type mice but no significant increase was observed in Gadd45β KO mice. We focused on these two genes because previous reports showed that hepatic Irisin/Fndc5 expression was significantly higher in HCC patients and that irisin binds to TGF-β receptor complex that includes TGFBR2 subunit. Our results revealed irisin peptide in cell culture media increased the growth rate of mouse hepatocyte-derived AML12 cells. Microarray analysis revealed that irisin-regulated genes in AML12 cells showed a significant association with the genes in the TGF-β pathway. Expression of irisin/Fndc5 and Tgfbr2 induced growth of human HCC cell line HepG2. Thus, Gadd45β plays an indispensable role in mouse HCC development regulating the irisin/Fndc5 and Tgfbr2 genes.
Collapse
Affiliation(s)
- Takeshi Hori
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health,
Research Triangle Park, NC, United States
- Department of Biomechanics, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kosuke Yokobori
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health,
Research Triangle Park, NC, United States
| | - Rick Moore
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health,
Research Triangle Park, NC, United States
| | - Masahiko Negishi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health,
Research Triangle Park, NC, United States
| | - Tatsuya Sueyoshi
- Pharmacogenetics Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health,
Research Triangle Park, NC, United States
| |
Collapse
|
44
|
Conticini E, Naveen R, Sen P, Singh M, Rathore U, Anuja AK, Rai MK, Yadav B, Prasad N, Agarwal V, Gupta L. Renal injury, biomarkers, and myositis, an understudied aspect of disease: prospective study in the MyoCite cohort. Front Med (Lausanne) 2023; 10:1127657. [PMID: 37425322 PMCID: PMC10325640 DOI: 10.3389/fmed.2023.1127657] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/30/2023] [Indexed: 07/11/2023] Open
Abstract
INTRODUCTION The mechanisms leading to chronic kidney disease (CKD) in patients with idiopathic inflammatory myopathies (IIMs) are poorly understood. We assessed the prevalence of subclinical renal injury in patients with IIMs, through elevation in biomarker levels of tubular injury and fibrosis (NGAL, KIM1, Activin A, CD163, and Cys-c), and assessed differences between subtypes of IIMs, and the effect of disease activity and duration. MATERIALS AND METHODS Clinical data, core set measures, sera and urine were prospectively collected from all patients enrolled in the MyoCite cohort from 2017 to 2021. Twenty healthy subjects (HC) and 16 patients with acute kidney injury (AKI) were included as controls. Baseline and follow up data for IIMs were included. Enzyme-linked immunosorbent assay (ELISA) was used to measure urine NGAL (Human Lipocalin-2/NGAL Duoset ELISA, Cat no: DY1757), KIM1 (Human TIM-1/KIM 1/HAVCR Duoset ELISA, Cat.no: DY1750B), Activin A (Human Activin A Duoset ELISA, Cat no: DY338), CD163 (Human CD163 Duoset ELISA,Cat no: DY1607-05), and Cys-c (Human Cystatin C Duoset ELISA, Cat. no.: DY1196) levels, while eGFR (unit mL/min/1.73 m2) was calculated by the Cockcroft-Gault formula and CKD-EPI formula. RESULTS Analysis of 201 visits of 110 adult patients with IIMs indicated higher normalized biomarker levels compared to HCs, and comparable to patients with AKI, with the exception of NGAL, which was higher in the AKI group. Notably 72 (49%) patients with IIMs had eGFR<90; the levels of the 5 biomarkers were comparable between active and inactive IIMs, and different subtypes of IIMs. Similarly, a poor correlation between urine biomarker levels and core set measures of activity and damage was found. Changes in biomarker levels on follow-up did not correlate with eGFR changes. DISCUSSION This exploratory analysis of urinary biomarkers identified low eGFR and elevated biomarkers of CKD in nearly half of the patients with IIMs, comparable to patients with AKI and higher than HCs, indicative of potential renal damage in IIMs that may have a lead to complications in other systems.
Collapse
Affiliation(s)
- Edoardo Conticini
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - R. Naveen
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | | | - Mantabya Singh
- Department of Nephrology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Upendra Rathore
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Anamika Kumari Anuja
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Mohit Kumar Rai
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Brijesh Yadav
- Department of Nephrology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Narayan Prasad
- Department of Nephrology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Vikas Agarwal
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Latika Gupta
- Department of Rheumatology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, United Kingdom
- Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
- Department of Rheumatology, City Hospital, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, United Kingdom
| |
Collapse
|
45
|
Srivastava A, Tomar B, Sharma D, Rath SK. Mitochondrial dysfunction and oxidative stress: Role in chronic kidney disease. Life Sci 2023; 319:121432. [PMID: 36706833 DOI: 10.1016/j.lfs.2023.121432] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023]
Abstract
Chronic kidney disease (CKD) is associated with a variety of distinct disease processes that permanently change the function and structure of the kidney across months or years. CKD is characterized as a glomerular filtration defect or proteinuria that lasts longer than three months. In most instances, CKD leads to end-stage kidney disease (ESKD), necessitating kidney transplantation. Mitochondrial dysfunction is a typical response to damage in CKD patients. Despite the abundance of mitochondria in the kidneys, variations in mitochondrial morphological and functional characteristics have been associated with kidney inflammatory responses and injury during CKD. Despite these variations, CKD is frequently used to define some classic signs of mitochondrial dysfunction, including altered mitochondrial shape and remodeling, increased mitochondrial oxidative stress, and a marked decline in mitochondrial biogenesis and ATP generation. With a focus on the most significant developments and novel understandings of the involvement of mitochondrial remodeling in the course of CKD, this article offers a summary of the most recent advances in the sources of procured mitochondrial dysfunction in the advancement of CKD. Understanding mitochondrial biology and function is crucial for developing viable treatment options for CKD.
Collapse
Affiliation(s)
- Anjali Srivastava
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Bhawna Tomar
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divyansh Sharma
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Srikanta Kumar Rath
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
46
|
Matsuura R, Doi K, Rabb H. Acute kidney injury and distant organ dysfunction-network system analysis. Kidney Int 2023; 103:1041-1055. [PMID: 37030663 DOI: 10.1016/j.kint.2023.03.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 04/10/2023]
Abstract
Acute kidney injury (AKI) occurs in about half of critically ill patients and associates with high in-hospital mortality, increased long-term mortality post-discharge and subsequent progression to chronic kidney disease. Numerous clinical studies have shown that AKI is often complicated by dysfunction of distant organs, which is a cause of the high mortality associated with AKI. Experimental studies have elucidated many mechanisms of AKI-induced distant organ injury, which include inflammatory cytokines, oxidative stress and immune responses. This review will provide an update on evidence of organ crosstalk and potential therapeutics for AKI-induced organ injuries, and present the new concept of a systemic organ network to balance homeostasis and inflammation that goes beyond kidney-crosstalk with a single distant organ.
Collapse
Affiliation(s)
- Ryo Matsuura
- Department of Nephrology and Endocrinology, the University of Tokyo Hospital
| | - Kent Doi
- Department of Emergency and Critical Care Medicine, the University of Tokyo Hospital.
| | - Hamid Rabb
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine
| |
Collapse
|
47
|
DNA-dependent protein kinase catalytic subunit (DNA-PKcs) drives chronic kidney disease progression in male mice. Nat Commun 2023; 14:1334. [PMID: 36906617 PMCID: PMC10008567 DOI: 10.1038/s41467-023-37043-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/28/2023] [Indexed: 03/13/2023] Open
Abstract
Kidney injury initiates epithelial dedifferentiation and myofibroblast activation during the progression of chronic kidney disease. Herein, we find that the expression of DNA-PKcs is significantly increased in the kidney tissues of both chronic kidney disease patients and male mice induced by unilateral ureteral obstruction and unilateral ischemia-reperfusion injury. In vivo, knockout of DNA-PKcs or treatment with its specific inhibitor NU7441 hampers the development of chronic kidney disease in male mice. In vitro, DNA-PKcs deficiency preserves epithelial cell phenotype and inhibits fibroblast activation induced by transforming growth factor-beta 1. Additionally, our results show that TAF7, as a possible substrate of DNA-PKcs, enhances mTORC1 activation by upregulating RAPTOR expression, which subsequently promotes metabolic reprogramming in injured epithelial cells and myofibroblasts. Taken together, DNA-PKcs can be inhibited to correct metabolic reprogramming via the TAF7/mTORC1 signaling in chronic kidney disease, and serve as a potential target for treating chronic kidney disease.
Collapse
|
48
|
Liao K, Cui Z, Wang Z, Peng Y, Tang S, Chen J. Hyperosmolar Potassium Inhibits Corneal Myofibroblast Transformation and Prevent Corneal Scar. Curr Eye Res 2023; 48:238-250. [PMID: 36149345 DOI: 10.1080/02713683.2022.2129072] [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] [Indexed: 11/03/2022]
Abstract
PURPOSE Corneal myofibroblasts play a crucial role in the process of corneal scarring. Potassium has been documented to reduce skin scar tissue formation. Herein, we investigated the ability of potassium to prevent corneal fibrosis in cell culture and in vivo. METHODS Corneal fibroblasts (CFs) were isolated from the corneal limbus and treated with TGF-β1 to transform into corneal myofibroblasts. Corneal myofibroblast markers were detected by quantitative real-time PCR, Western blot, and immunofluorescence. The contractive functions of corneal myofibroblast were evaluated by the scratch assay and the collagen gel contraction assay. RNA sequencing in corneal fibroblasts was performed to explore the mechanisms underlying hyperosmolar potassium treatment. GO and KEGG analysis were performed to explore the underlying mechanism by hyperosmolar potassium treatment. The ATP detection assay assessed the level of cell metabolism. KCl eye drops four times per day were administered to mice models of corneal injury to evaluate the ability to prevent corneal scar formation. Corneal opacity area was evaluated by Image J software. RESULTS Treatment with hyperosmolar potassium could suppress corneal myofibroblast transformation and collagen I synthesis induced by TGF-β1 in cell culture. Hyperosmolar potassium could inhibit wound healing and gel contraction in CFs. RNA sequencing results suggested that genes involved in the metabolic pathway were downregulated after KCl treatment. ATP levels were significantly decreased in the KCl group compared with the control group. Hyperosmolar potassium could prevent corneal myofibroblast transformation after corneal injury and corneal scar formation in mice. CONCLUSION Potassium can suppress corneal myofibroblast transformation and collagen I protein synthesis. Moreover, given that KCl eye drops can prevent corneal scar formation, it has been suggested to have huge prospects as a novel treatment approach during clinical practice.
Collapse
Affiliation(s)
- Kai Liao
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
- Aier Eye Institute, Changsha, Hunan Province, China
| | - Zekai Cui
- Aier Eye Institute, Changsha, Hunan Province, China
| | - Zhijie Wang
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
- Aier Eye Institute, Changsha, Hunan Province, China
| | - Yu Peng
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
- Aier Eye Institute, Changsha, Hunan Province, China
| | - Shibo Tang
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
- Aier Eye Institute, Changsha, Hunan Province, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jiansu Chen
- Aier School of Ophthalmology, Central South University, Changsha, Hunan, China
- Aier Eye Institute, Changsha, Hunan Province, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| |
Collapse
|
49
|
Abstract
Muscle wasting (ie, atrophy) is a serious consequence of chronic kidney disease (CKD) that reduces muscle strength and function. It reduces the quality of life for CKD patients and increases the risks of comorbidities and mortality. Current treatment strategies to prevent or reverse skeletal muscle loss are limited owing to the broad and systemic nature of the initiating signals and the multifaceted catabolic mechanisms that accelerate muscle protein degradation and impair protein synthesis and repair pathways. Recent evidence has shown how organs such as muscle, adipose, and kidney communicate with each other through interorgan exchange of proteins and RNAs during CKD. This crosstalk changes cell functions in the recipient organs and represents an added dimension in the complex processes that are responsible for muscle atrophy in CKD. This complexity creates challenges for the development of effective therapies to ameliorate muscle wasting and weakness in patients with CKD.
Collapse
Affiliation(s)
- Xiaonan H Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, GA
| | - S Russ Price
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC; Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC.
| |
Collapse
|
50
|
Li G, Qin H, Zhou M, Zhang T, Zhang Y, Ding H, Xu L, Song J. Knockdown of SIRT3 perturbs protective effects of irisin against bone loss in diabetes and periodontitis. Free Radic Biol Med 2023; 200:11-25. [PMID: 36863620 DOI: 10.1016/j.freeradbiomed.2023.02.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/19/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023]
Abstract
A well-recognized risk factor for periodontitis, diabetes mellitus (DM) aggravates periodontal disease with increasing alveolar bone loss. As a novel myokine, irisin is closely linked with bone metabolism. Nonetheless, the effects of irisin on periodontitis under diabetic conditions and the underlying mechanisms remain poorly understood. Here, we showed that local irisin treatment ameliorates alveolar bone loss and oxidative stress, increases SIRT3 expression within periodontal tissues of our experimentally-induced diabetes and periodontitis (DP) rat models. By culturing the periodontal ligament cells (PDLCs) in vitro, we found that irisin could partially rescue inhibited cell viability, mitigate accumulated intracellular oxidative stress, ameliorate mitochondrial dysfunctions, and restore disturbed osteogenic and osteoclastogenic capacities of PDLCs when exposed to high glucose and pro-inflammatory stimulation. Furthermore, lentivirus-mediated SIRT3 knockdown was employed to unravel the underlying mechanism by which SIRT3 mediated irisin's beneficial effects on PDLCs. Meanwhile, in SIRT3-deficient mice, irisin treatment did not protect against alveolar bone destruction and oxidative stress accumulation in DP models, which underlined the crucial role of SIRT3 in mediating the positive effects of irisin on DP. Our findings, for the first time, revealed that irisin attenuates alveolar bone loss and oxidative stress via activation of the SIRT3 signaling cascade, and highlighted its therapeutic potential for the treatment of DP.
Collapse
Affiliation(s)
- Guangyue Li
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China; College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Han Qin
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China; College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Mengjiao Zhou
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China; College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Tingwei Zhang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China; College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Yang Zhang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China; College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Huifen Ding
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China; College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Ling Xu
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China; College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Jinlin Song
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China; College of Stomatology, Chongqing Medical University, Chongqing, China.
| |
Collapse
|