1
|
Yu F, Chen J, Wang X, Hou S, Li H, Yao Y, He Y, Chen K. Metabolic reprogramming of peritoneal mesothelial cells in peritoneal dialysis-associated fibrosis: therapeutic targets and strategies. Cell Commun Signal 2025; 23:114. [PMID: 40016825 PMCID: PMC11866825 DOI: 10.1186/s12964-025-02113-2] [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: 11/12/2024] [Accepted: 02/17/2025] [Indexed: 03/01/2025] Open
Abstract
Peritoneal dialysis (PD) is considered a life-saving treatment for end-stage renal disease. However, prolonged PD use can lead to the development of peritoneal fibrosis (PF), diminishing its efficacy. Peritoneal mesothelial cells (PMCs) are key initiators of PF when they become damaged. Exposure to high glucose‑based peritoneal dialysis fluids (PDFs) contributes to PF development by directly affecting highly metabolically active PMCs. Recent research indicates that PMCs undergo metabolic reprogramming when exposed to high-glucose PDFs, including enhanced glycolysis, impaired oxidative phosphorylation, abnormal lipid metabolism, and mitochondrial dysfunction. Although this metabolic transition temporarily compensates for the cellular damage and maintains energy levels, its long-term impact on peritoneal tissue is concerning. Multiple studies have identified a close association between this shift in energy metabolism and PF, and may promote the progression of PF through various molecular mechanisms. This review explores recent findings regarding the role and mechanism of PMC metabolic reprogramming in PF progression. Moreover, it provides a summary of potential therapeutic strategies aimed at various metabolic processes, including glucose metabolism, lipid metabolism, and mitochondrial function. The review establishes that targeting metabolic reprogramming in PMCs may be a novel strategy for preventing and treating PD-associated fibrosis.
Collapse
Affiliation(s)
- Fang Yu
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Jia Chen
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Xiaoyue Wang
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Shihui Hou
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Hong Li
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Yaru Yao
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Yani He
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
- State Key Laboratory of Trauma and Chemical poisoning, Burns and Combined Injury, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
| | - Kehong Chen
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
- State Key Laboratory of Trauma and Chemical poisoning, Burns and Combined Injury, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
| |
Collapse
|
2
|
Ning ZH, Wang XH, Tang HF, Hu HJ. The role of SGLT1 in atrial fibrillation and its relationship with endothelial-mesenchymal transition. Biochem Biophys Res Commun 2025; 748:151338. [PMID: 39823893 DOI: 10.1016/j.bbrc.2025.151338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/23/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Atrial fibrillation (AF) is a prevalent arrhythmia closely associated with atrial fibrosis, posing significant challenges to cardiovascular health. Recent studies have identified the sodium-glucose co-transporter 1 (SGLT1) as a potential key player in the pathophysiology of heart diseases, particularly in the context of AF and atrial fibrosis. This review aims to synthesize current knowledge regarding the mechanisms by which SGLT1 influences the development of AF and atrial fibrosis, with a specific focus on its relationship with endothelial-mesenchymal transition (EMT). By analyzing the latest research findings, this paper discusses how SGLT1 may modulate structural and functional changes in the atria, thereby enhancing our understanding of the underlying mechanisms driving AF.
Collapse
Affiliation(s)
- Zhi-Hong Ning
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Xiu-Heng Wang
- The First Affiliated Hospital, Department of Medical-record, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Hui-Fang Tang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Heng-Jing Hu
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China; Department of Cardiovascular Disease and Key Lab for Atherosclerosis of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| |
Collapse
|
3
|
Riedl Khursigara M, Liu P, Kaur R, Mavrakanas TA. Role of SGLT-2 Inhibitors in Ultrafiltration Failure in Peritoneal Dialysis: A Narrative Review. Can J Kidney Health Dis 2024; 11:20543581241293500. [PMID: 39502166 PMCID: PMC11536389 DOI: 10.1177/20543581241293500] [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: 03/22/2024] [Accepted: 08/14/2024] [Indexed: 11/08/2024] Open
Abstract
Purpose of review Sodium-glucose co-transporter-2 (SGLT-2) inhibitors are glucose lowering agents with protective effects on cardiovascular health and the ability to slow chronic kidney disease (CKD) progression. The benefits of SGLT-2 inhibitors have not been studied in patients with advanced CKD or on maintenance dialysis. Ultrafiltration failure is a common reason for failure of peritoneal dialysis (PD). Glucose transporters, such as SGLT-2, are involved in the progression to ultrafiltration failure, and hence, SGLT-2 inhibitors might be beneficial in patients on PD to prevent ultrafiltration failure. Source of information Here, we review data from animal models and ongoing clinical trials of SGLT-2 inhibitors in advanced CKD, as well as considerations for a phase III trial in patients on PD. Methods A literature search was conducted and information on clinical trials was obtained from clinicaltrials.gov. Key findings Animal models of PD have shown upregulation of glucose transporters in the peritoneal membrane and a potential effect of SGLT-2 inhibitors on glucose absorption and ultrafiltration. Several clinical trials are currently ongoing with SGLT-2 inhibitors in patients on PD. We discuss their study designs and propose a mixed-methods, patient-centered approach to studying SGLT-2 inhibitors in PD patients. We also discuss the potential implications of SGLT-2 inhibitors on people living with kidney failure, especially in remote communities.
Collapse
Affiliation(s)
| | - Ping Liu
- Departments of Medicine and Community Health Sciences, Cumming School of Medicine, University of Calgary, AB, Canada
| | | | - Thomas A. Mavrakanas
- Division of Nephrology, Department of Medicine, Research Institute, McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
4
|
Stepanova N. Dyslipidemia in Peritoneal Dialysis: Implications for Peritoneal Membrane Function and Patient Outcomes. Biomedicines 2024; 12:2377. [PMID: 39457689 PMCID: PMC11505255 DOI: 10.3390/biomedicines12102377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Dyslipidemia is a common metabolic complication in patients undergoing peritoneal dialysis (PD) and has traditionally been viewed primarily in terms of cardiovascular risk. Current guidelines do not recommend initiating lipid-lowering therapy in dialysis patients due to insufficient evidence of its benefits on cardiovascular mortality. However, the impact of dyslipidemia in PD patients may extend beyond cardiovascular concerns, influencing PD-related outcomes such as the peritoneal ultrafiltration rate, residual kidney function, PD technique survival, and overall mortality. This review challenges the traditional perspective by discussing dyslipidemia's potential role in PD-related complications, which may account for the observed link between dyslipidemia and increased all-cause mortality in PD patients. It explores the pathophysiology of dyslipidemia in PD, the molecular mechanisms linking dyslipidemia to peritoneal membrane dysfunction, and summarizes clinical evidence supporting this hypothesis. In addition, this paper examines the potential for therapeutic strategies to manage dyslipidemia to improve peritoneal membrane function and patient outcomes. The review calls for future research to investigate dyslipidemia as a potential contributor to peritoneal membrane dysfunction and to develop targeted interventions for PD patients.
Collapse
Affiliation(s)
- Natalia Stepanova
- State Institution “O.O. Shalimov National Scientific Center of Surgery and Transplantology, National Academy of Medical Science of Ukraine”, 03126 Kyiv, Ukraine;
- Medical Center “Nephrocenter”, 03057 Kyiv, Ukraine
| |
Collapse
|
5
|
Kang Y, Liu Y, Fu P, Ma L. Peritoneal fibrosis: from pathophysiological mechanism to medicine. Front Physiol 2024; 15:1438952. [PMID: 39301425 PMCID: PMC11411570 DOI: 10.3389/fphys.2024.1438952] [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: 05/27/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
Peritoneal dialysis (PD) is currently one of the effective methods for treating end-stage renal disease (ESRD). However, long-term exposure to high concentration glucose in peritoneal dialysis environment could lead to peritoneal fibrosis (PF), impaired peritoneal filtration function, decreased peritoneal dialysis efficiency, and even withdrawal from peritoneal dialysis in patients. Considerable evidence suggests that peritoneal fibrosis after peritoneal dialysis is related to crucial factors such as mesothelial-to-mesenchymal transition (MMT), inflammatory response, and angiogenesis, etc. In our review, we summarize the pathophysiological mechanisms and further illustrate the future strategies against PF.
Collapse
Affiliation(s)
- Yingxi Kang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Yuan Liu
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ping Fu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Liang Ma
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Lho Y, Park Y, Do JY, Kim AY, Park YE, Kang SH. Empagliflozin attenuates epithelial-to-mesenchymal transition through senescence in peritoneal dialysis. Am J Physiol Renal Physiol 2024; 327:F363-F372. [PMID: 38961839 DOI: 10.1152/ajprenal.00028.2024] [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: 01/23/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is considered as one of the senescence processes; reportedly, antisenescence therapies effectively reduce EMT. Some models have shown antisenescence effects with the use of sodium-glucose cotransporter 2 (SGLT2) inhibitor. Therefore, our study investigated the antisenescence effects of empagliflozin as an SGLT2 inhibitor in a peritoneal fibrosis model and their impact on EMT inhibition. For in vitro study, human peritoneal mesothelial cells (HPMCs) were isolated and grown in a 96-well plate. The cell media were exchanged with serum-free M199 medium with d-glucose, with or without empagliflozin. All animal experiments were carried out in male mice. Mice were randomly classified into three treatment groups based on peritoneal dialysis (PD) or empagliflozin. We evaluated changes in senescence and EMT markers in HPMCs and PD model. HPMCs treated with glucose transformed from cobblestone to spindle shape, resulting in EMT. Empagliflozin attenuated these morphological changes. Reactive oxygen species production, DNA damage, senescence, and EMT markers were increased by glucose treatment; however, cotreatment with glucose and empagliflozin attenuated these changes. For the mice with PD, an increase in thickness, collagen deposition, staining for senescence, or EMT markers of the parietal peritoneum was observed, which, however, was attenuated by cotreatment with empagliflozin. p53, p21, and p16 increased in mice with PD compared with those in the control group; however, these changes were decreased by empagliflozin. In conclusion, empagliflozin effectively attenuated glucose-induced EMT in HPMCs through a decrease in senescence. Cotreatment with empagliflozin improved peritoneal thickness and fibrosis in PD.NEW & NOTEWORTHY Epithelial-to-mesenchymal transition (EMT) is considered one of the senescence processes. Antisenescence therapies may effectively reduce EMT in peritoneal dialysis models. Human peritoneal mesothelial cells treated with glucose show an increase in senescence and EMT markers; however, empagliflozin attenuates these changes. Mice undergoing peritoneal dialysis exhibit increased senescence and EMT markers, which are decreased by empagliflozin. These findings suggest that empagliflozin may emerge as a novel strategy for prevention or treatment of peritoneal fibrosis.
Collapse
Affiliation(s)
- Yunmee Lho
- Senotherpy-Based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Yeong Park
- Senotherpy-Based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Jun Young Do
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - A-Young Kim
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Yong-Eun Park
- Department of Surgery, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Seok Hui Kang
- Senotherpy-Based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| |
Collapse
|
7
|
Sheng L, Sun J, Huang L, Yu M, Meng X, Shan Y, Dai H, Wang F, Shi J, Sheng M. Astragalus membranaceus and its monomers treat peritoneal fibrosis and related muscle atrophy through the AR/TGF-β1 pathway. Front Pharmacol 2024; 15:1418485. [PMID: 39239655 PMCID: PMC11374727 DOI: 10.3389/fphar.2024.1418485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/16/2024] [Indexed: 09/07/2024] Open
Abstract
Background: To anticipate the potential molecular mechanism of Astragalus membranaceus (AM) and its monomer, Calycosin, against peritoneal fibrosis (PF) and related muscle atrophy using mRNA-seq, network pharmacology, and serum pharmacochemistry. Methods: Animal tissues were examined to evaluate a CKD-PF mice model construction. mRNA sequencing was performed to find differential targets. The core target genes of AM against PF were screened through network pharmacology analysis, and CKD-PF mice models were given high- and low-dose AM to verify common genes. Serum pharmacochemistry was conducted to clarify which components of AM can enter the blood circulation, and the selected monomer was further validated through cell experiments for the effect on PF and mesothelial mesenchymal transition (MMT) of peritoneal mesothelial cells (PMCs). Results: The CKD-PF mice models were successfully constructed. A total of 31,184 genes were detected in the blank and CKD-PF groups, and 228 transcription factors had significant differences between the groups. Combined with network pharmacology analysis, a total of 228 AM-PF-related targets were identified. Androgen receptor (AR) was the remarkable transcription factor involved in regulating transforming growth factor-β1 (TGF-β1). AM may be involved in regulating the AR/TGF-β1 signaling pathway and may alleviate peritoneal dialysis-related fibrosis and muscle atrophy in CKD-PF mice. In 3% peritoneal dialysis solution-stimulated HMrSV5 cells, AR expression levels were dramatically reduced, whereas TGF-β1/p-smads expression levels were considerably increased. Conclusion: AM could ameliorate PF and related muscle atrophy via the co-target AR and modulated AR/TGF-β1 pathway. Calycosin, a monomer of AM, could partially reverse PMC MMT via the AR/TGF-β1/smads pathway. This study explored the traditional Chinese medicine theory of "same treatment for different diseases," and supplied the pharmacological evidence of "AM can treat flaccidity syndrome."
Collapse
Affiliation(s)
- Li Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
- Medical Experimental Research Center, First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinyi Sun
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liyan Huang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Manshu Yu
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaohui Meng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Shan
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Huibo Dai
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Funing Wang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jun Shi
- School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meixiao Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
8
|
Moral Berrio E, De La Flor JC, Arambarri Segura M, Rodríguez-Doyágüez P, Martínez Calero A, Zamora R, Cieza-Terrones M, Yuste-Lozano C, Sánchez de la Nieta García MD, Nieto Iglesias J, Vozmediano Poyatos C. Effects of Sodium-Glucose Cotransporter 2 Inhibitors in Diabetic and Non-Diabetic Patients with Advanced Chronic Kidney Disease in Peritoneal Dialysis on Residual Kidney Function: In Real-World Data. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1198. [PMID: 39202480 PMCID: PMC11356563 DOI: 10.3390/medicina60081198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024]
Abstract
Background and Objectives: Peritoneal dialysis (PD) is a renal replacement therapy modality in which the dialysis dose can be individually adapted according to the patients' residual kidney function (RKF). RKF is a crucial factor for technique and patient survival. Pharmacological strategies aimed at slowing the loss of RKF in patients on PD are limited. Therefore, we aimed to assess the potential effects and safety of sodium-glucose cotransporter 2 (SGLT-2) inhibitors on the preservation of RKF in patients with and without type 2 diabetes mellitus (T2DM) on PD during an average follow-up of 6 months. Materials and Methods: In this retrospective observational, single-center study on real-world data, we included patients from the Peritoneal Dialysis Unit of the Hospital General Universitario de Ciudad Real, who started treatment with SGLT-2 inhibitors during the period from December 2022 to December 2023. Data on analytical and clinical parameters, RKF, and peritoneal membrane transport function were retrospectively collected at months 0, 3, and 6. Results: Out of 31 patients in our unit, 16 prevalent patients initiated treatment with SGLT-2 inhibitors (13 empagliflozin and 3 dapagliflozin). A total of 62.5% were male and the mean age was 67.3 years. The baseline peritoneal ultrafiltration was higher in the non-diabetic patient (NDMP) group than in the diabetic patient (DMP) group. However, the residual diuresis volume, 24 h residual renal clearance rate of urea in urine, and 24 h proteinuria were higher in the DMP group than in the NDMP group. At the sixth month, patients in both groups preserved RKF and diuresis, with a trend towards a non-significant reduction in proteinuria and blood pressure. Only two patients of the DMP group presented adverse effects. Conclusions: The use of SGLT-2 inhibitors in our sample of patients with and without T2DM on PD appears to be safe and effective to preserve RKF.
Collapse
Affiliation(s)
- Esperanza Moral Berrio
- Department of Nephrology, Hospital General Universitario de Ciudad Real, 13005 Ciudad Real, Spain; (E.M.B.); (M.A.S.); (A.M.C.); (M.D.S.d.l.N.G.); (J.N.I.); (C.V.P.)
| | - José C. De La Flor
- Department of Nephrology, Hospital Central de la Defensa Gómez Ulla, 28047 Madrid, Spain
- Department of Medicine and Medical Specialties, Faculty of Medicine, Alcala University, 28805 Madrid, Spain
| | - Minerva Arambarri Segura
- Department of Nephrology, Hospital General Universitario de Ciudad Real, 13005 Ciudad Real, Spain; (E.M.B.); (M.A.S.); (A.M.C.); (M.D.S.d.l.N.G.); (J.N.I.); (C.V.P.)
| | | | - Alberto Martínez Calero
- Department of Nephrology, Hospital General Universitario de Ciudad Real, 13005 Ciudad Real, Spain; (E.M.B.); (M.A.S.); (A.M.C.); (M.D.S.d.l.N.G.); (J.N.I.); (C.V.P.)
| | - Rocío Zamora
- Department of Nephrology, Hospital Universitario General Villalba, 28400 Madrid, Spain;
| | | | | | - María Dolores Sánchez de la Nieta García
- Department of Nephrology, Hospital General Universitario de Ciudad Real, 13005 Ciudad Real, Spain; (E.M.B.); (M.A.S.); (A.M.C.); (M.D.S.d.l.N.G.); (J.N.I.); (C.V.P.)
| | - Javier Nieto Iglesias
- Department of Nephrology, Hospital General Universitario de Ciudad Real, 13005 Ciudad Real, Spain; (E.M.B.); (M.A.S.); (A.M.C.); (M.D.S.d.l.N.G.); (J.N.I.); (C.V.P.)
| | - Carmen Vozmediano Poyatos
- Department of Nephrology, Hospital General Universitario de Ciudad Real, 13005 Ciudad Real, Spain; (E.M.B.); (M.A.S.); (A.M.C.); (M.D.S.d.l.N.G.); (J.N.I.); (C.V.P.)
| |
Collapse
|
9
|
Doi Y, Shinzawa M, Arisato T, Oka H, Matsumoto A, Kitamura H, Nakazono Y, Nishiya Y, Ueda Y, Kamimura T, Hayashi T, Yoshihara F, Isaka Y. Effects of sodium-glucose co-transporter 2 inhibitors on ultrafiltration in patients with peritoneal dialysis: a protocol for a randomized, double-blind, placebo-controlled, crossover trial (EMPOWERED). Clin Exp Nephrol 2024; 28:629-635. [PMID: 38402502 PMCID: PMC11189947 DOI: 10.1007/s10157-024-02467-w] [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: 10/30/2023] [Accepted: 01/17/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Volume overload is common and associated with high mortality in patients on peritoneal dialysis (PD). Traditional strategies including diuretics, water/salt restriction, and icodextrin-based solutions cannot always fully correct this condition, necessitating novel alternative strategies. Recent studies confirmed the expression of sodium-glucose cotransporter 2 (SGLT2) in the human peritoneum. Experimental data suggest that SGLT2 inhibitors decrease glucose absorption from the PD solution, thereby increasing the ultrafiltration volume. This trial aims to assess whether SGLT2 inhibitors increase the ultrafiltration volume in patients on PD. METHODS The EMPOWERED trial (trial registration: jRCTs051230081) is a multicenter, randomized, double-blind, placebo-controlled, crossover trial. Patients with clinically diagnosed chronic heart failure are eligible regardless of the presence of diabetes if they use at least 3 L/day glucose-based PD solutions. Participants will be randomly assigned (1:1) to receive empagliflozin 10 mg once daily and then placebo or vice versa. Each treatment period will last 8 weeks with a 4-week washout period. This study will recruit at least 36 randomized participants. The primary endpoint is the change in the daily ultrafiltration volume from baseline to week 8 in each intervention period. The key secondary endpoints include changes in the biomarkers of drained PD solutions, renal residual function, and anemia-related parameters. CONCLUSIONS This trial aims to assess the benefit of SGLT2 inhibitors in fluid management with a novel mechanism of action in patients on PD. It will also provide insights into the effects of SGLT2 inhibitors on solute transport across the peritoneal membrane and residual renal function.
Collapse
Affiliation(s)
- Yohei Doi
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2-D11, Yamada-oka, Suita, Osaka, 565-0871, Japan.
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan.
| | - Maki Shinzawa
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2-D11, Yamada-oka, Suita, Osaka, 565-0871, Japan
- Health and Counseling Center, Osaka University, Toyonaka, Osaka, Japan
| | - Tetsuya Arisato
- Division of Nephrology and Hypertension, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Hideaki Oka
- Division of Kidney Center, Matsuyama Red Cross Hospital, Matsuyama, Ehime, Japan
| | - Ayumi Matsumoto
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2-D11, Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Harumi Kitamura
- Department of Clinical Quality Management, Osaka University Hospital, Suita, Osaka, Japan
| | - Yumi Nakazono
- Medicine Division, Nippon Boehringer Ingelheim Co., Ltd., Shinagawa-ku, Tokyo, Japan
| | - Yoichi Nishiya
- Medicine Division, Nippon Boehringer Ingelheim Co., Ltd., Shinagawa-ku, Tokyo, Japan
| | - Yoshiyasu Ueda
- Department of Kidney Disease and Hypertension, Osaka General Medical Center, Osaka, Osaka, Japan
| | - Taro Kamimura
- Division of Kidney Center, Matsuyama Red Cross Hospital, Matsuyama, Ehime, Japan
| | - Terumasa Hayashi
- Department of Kidney Disease and Hypertension, Osaka General Medical Center, Osaka, Osaka, Japan
| | - Fumiki Yoshihara
- Division of Nephrology and Hypertension, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2-D11, Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
10
|
Huang Q, Chen J, Liao S, Long J, Fang R, He Y, Chen P, Liu D. The SGLT2 inhibitor empagliflozin inhibits skeletal muscle fibrosis in naturally aging male mice through the AMPKα/MMP9/TGF-β1/Smad pathway. Biogerontology 2024; 25:567-581. [PMID: 38403802 DOI: 10.1007/s10522-024-10093-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/03/2024] [Indexed: 02/27/2024]
Abstract
ABSTACT With advancing age, the incidence of sarcopenia increases, eventually leading to a cascade of adverse events. However, there is currently a lack of effective pharmacological treatment for sarcopenia. Sodium-glucose co-transporter 2 inhibitor (SGLT2i) empagliflozin demonstrates anti-fibrotic capabilities in various organs. This study aims to determine whether empagliflozin can improve skeletal muscle fibrosis induced by sarcopenia in naturally aging mice. A natural aging model was established by feeding male mice from 13 months of age to 19 months of age. A fibrosis model was created by stimulating skeletal muscle fibroblasts with TGF-β1. The Forelimb grip strength test assessed skeletal muscle function, and expression levels of COL1A1, COL3A1, and α-SMA were analyzed by western blot, qPCR, and immunohistochemistry. Additionally, levels of AMPKα/MMP9/TGFβ1/Smad signaling pathways were examined. In naturally aging mice, skeletal muscle function declines, expression of muscle fibrosis markers increases, AMPKα expression is downregulated, and MMP9/TGFβ1/Smad signaling pathways are upregulated. However, treatment with empagliflozin reverses this phenomenon. At the cellular level, empagliflozin exhibits similar anti-fibrotic effects, and these effects are attenuated by Compound C and siAMPKα. Empagliflozin exhibits anti-fibrotic effects, possibly associated with the AMPK/MMP9/TGFβ1/Smad signaling pathways.
Collapse
Affiliation(s)
- Qixuan Huang
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Jie Chen
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Department of Endocrinology, The Ninth People's Hospital of Chongqing, Chongqing, 400700, China
| | - Siqi Liao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Jiangchuan Long
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Ronghua Fang
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yusen He
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Peiyun Chen
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Dongfang Liu
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
11
|
Sheng L, Shan Y, Dai H, Yu M, Sun J, Huang L, Wang F, Sheng M. Intercellular communication in peritoneal dialysis. Front Physiol 2024; 15:1331976. [PMID: 38390449 PMCID: PMC10882094 DOI: 10.3389/fphys.2024.1331976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Long-term peritoneal dialysis (PD) causes structural and functional alterations of the peritoneal membrane. Peritoneal deterioration and fibrosis are multicellular and multimolecular processes. Under stimulation by deleterious factors such as non-biocompatibility of PD solution, various cells in the abdominal cavity show differing characteristics, such as the secretion of different cytokines, varying protein expression levels, and transdifferentiation into other cells. In this review, we discuss the role of various cells in the abdominal cavity and their interactions in the pathogenesis of PD. An in-depth understanding of intercellular communication and inter-organ communication in PD will lead to a better understanding of the pathogenesis of this disease, enabling the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Li Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Shan
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Huibo Dai
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Manshu Yu
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinyi Sun
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liyan Huang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Funing Wang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meixiao Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
12
|
Stepanova N. SGLT2 inhibitors in peritoneal dialysis: a promising frontier toward improved patient outcomes. RENAL REPLACEMENT THERAPY 2024; 10:5. [DOI: 10.1186/s41100-024-00523-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/14/2024] [Indexed: 01/12/2025] Open
Abstract
AbstractPeritoneal dialysis (PD) stands as an important modality among kidney replacement therapies for end-stage kidney disease, offering patients remarkable flexibility and autonomy. Despite its widespread use, challenges such as glucose-related complications, peritoneal membrane fibrosis, declining renal function, and cardiovascular risks persist, necessitating innovative therapeutic approaches. Sodium–glucose cotransporter 2 (SGLT2) inhibitors, originally developed for treating type 2 diabetes mellitus, have recently shown promise as add-on therapy for patients with diabetic and non-diabetic chronic kidney disease (CKD), even in advanced stages. This review describes the potential role of SGLT2 inhibitors as a breakthrough therapeutic option in PD, emphasizing their ability to address unmet clinical needs and improve patient outcomes. The multiple effects of SGLT2 inhibitors in CKD, including metabolic modulation, antihypertensive, diuretic, anemia-reducing, antioxidant, and antiinflammatory properties, are reviewed in the context of PD challenges. Additionally, the potentially protective influence of SGLT2 inhibitors on the integrity of the peritoneal membrane and the transport of solutes and water in the peritoneum are emphasized. Despite these encouraging results, the paper highlights the potential risks associated with SGLT2 inhibitors in PD and emphasizes the need for cautious and thorough investigation of dosing, long-term safety considerations, and patient-specific factors through comprehensive clinical trials. Looking forward, the review argues for well-designed studies to evaluate the expanded safety profile of SGLT2 inhibitors in PD, with particular attention paid to peritoneal membrane integrity and overall patient outcomes.
Collapse
|
13
|
Pitaraki E, Jagirdar RM, Rouka E, Bartosova M, Sinis SI, Gourgoulianis KI, Eleftheriadis T, Stefanidis I, Liakopoulos V, Hatzoglou C, Schmitt CP, Zarogiannis SG. 2-Deoxy-glucose ameliorates the peritoneal mesothelial and endothelial barrier function perturbation occurring due to Peritoneal Dialysis fluids exposure. Biochem Biophys Res Commun 2024; 693:149376. [PMID: 38104523 DOI: 10.1016/j.bbrc.2023.149376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Peritoneal dialysis (PD) and prolonged exposure to PD fluids (PDF) induce peritoneal membrane (PM) fibrosis and hypervascularity, leading to functional PM degeneration. 2-deoxy-glucose (2-DG) has shown potential as PM antifibrotic by inhibiting hyper-glycolysis induced mesothelial-to-mesenchymal transition (MMT). We investigated whether administration of 2-DG with several PDF affects the permeability of mesothelial and endothelial barrier of the PM. The antifibrotic effect of 2-DG was confirmed by the gel contraction assay with embedded mesothelial (MeT-5A) or endothelial (EA.hy926) cells cultured in Dianeal® 2.5 % (CPDF), BicaVera® 2.3 % (BPDF), Balance® 2.3 % (LPDF) with/without 2-DG addition (0.2 mM), and qPCR for αSMA, CDH2 genes. Moreover, 2-DG effect was tested on the permeability of monolayers of mesothelial and endothelial cells by monitoring the transmembrane resistance (RTM), FITC-dextran (10, 70 kDa) diffusion and mRNA expression levels of CLDN-1 to -5, ZO1, SGLT1, and SGLT2 genes. Contractility of MeT-5A cells in CPDF/2-DG was decreased, accompanied by αSMA (0.17 ± 0.03) and CDH2 (2.92 ± 0.29) gene expression fold changes. Changes in αSMA, CDH2 were found in EA.hy926 cells, though αSMA also decreased under LPDF/2-DG incubation (0.42 ± 0.02). Overall, 2-DG mitigated the PDF-induced alterations in mesothelial and endothelial barrier function as shown by RTM, dextran transport and expression levels of the CLDN-1 to -5, ZO1, and SGLT2. Thus, supplementation of PDF with 2-DG not only reduces MMT but also improves functional permeability characteristics of the PM mesothelial and endothelial barrier.
Collapse
Affiliation(s)
- Eleanna Pitaraki
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Rajesh M Jagirdar
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Erasmia Rouka
- Department of Nursing, School of Health Sciences, University of Thessaly, GAIOPOLIS, 41500, Larissa, Greece
| | - Maria Bartosova
- Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, 69120, Heidelberg, Germany
| | - Sotirios I Sinis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece; Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Konstantinos I Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Theodoros Eleftheriadis
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Ioannis Stefanidis
- Department of Nephrology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Vassilios Liakopoulos
- 2(nd) Department of Nephrology, AHEPA Hospital, Aristotle University of Thessaloniki, 54636, Thessaloniki, Greece
| | - Chrissi Hatzoglou
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece
| | - Claus Peter Schmitt
- Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, 69120, Heidelberg, Germany
| | - Sotirios G Zarogiannis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500, Larissa, Greece.
| |
Collapse
|
14
|
Elsayed M, Moustafa YM, Mehanna ET, Elrayess RA, El-Sayed NM, Hazem RM. Empagliflozin protects against isoprenaline-induced fibrosis in rat heart through modulation of TGF-β/SMAD pathway. Life Sci 2024; 337:122354. [PMID: 38110076 DOI: 10.1016/j.lfs.2023.122354] [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: 09/18/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023]
Abstract
AIM Cardiac fibrosis is characterized by excessive accumulation of fibrous tissue, particularly collagens, in the myocardium. Accumulated fibrous tissue renders myocardium stiffer and reduces its contractility. Empagliflozin is an oral hypoglycemic agent with extra-diabetic functional profile toward maintaining cardiac functions. The present study aimed to examine protective effect of empagliflozin against an in-vivo model of cardiac fibrosis induced by isoprenaline and targeting TGF-β/SMAD signaling as a possible pathway responsible for such effect. MAIN METHODS Sixty animals were divided into six groups; the first was normal, and the second was treated with isoprenaline only (5 mg/kg/day I.P.) as a control. The third received pirfenidone (500 mg/kg/day P.O.), and the remaining groups received graded doses (5, 10, 20 mg/kg respectively) of empagliflozin for 14 days before fibrosis induction by isoprenaline (5 mg/kg/day) for 30 days. KEY FINDINGS Isoprenaline increased cardiac enzymes, and cardiac tissues revealed elevated concentrations of transforming growth factor β (TGF-β1), monocyte chemoattractant protein 1 (MCP-1), tumor necrosis factor α (TNF-α), and c-jun N-terminal kinase (JNK) proteins. Expression of nuclear factor kappa B (NF-κB), alpha smooth muscle actin (α-SMA), collagens, suppressor of mothers against decapentaplegic (SMADs), connective tissue growth factor (CTGF), and fibronectin was upregulated. Empagliflozin improved the histological picture of heart tissue in comparison to fibrosis developed in controls, and protected against fibrosis through significant modulation of all mentioned parameters' concentrations and expressions. SIGNIFICANCE Empagliflozin demonstrated a promising protective approach against biological model of cardiac fibrosis through an anti-fibrotic effect through targeting TGF-β signaling pathways.
Collapse
Affiliation(s)
- Mohammed Elsayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Yasser M Moustafa
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University in Cairo, Cairo 11829, Egypt
| | - Eman T Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; Department of Biochemistry, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt.
| | - Ranwa A Elrayess
- Department of Zoology, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Norhan M El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Reem M Hazem
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
15
|
Vorobiov M, Rogachev B, Riff R, Chaimowitz C, Neulander EZ, Basok A, Shnaider A, Douvdevani A, Haviv YS. Blockade of sodium-glucose co-transporters improves peritoneal ultrafiltration in uraemic rodent models. Perit Dial Int 2024; 44:48-55. [PMID: 37131323 DOI: 10.1177/08968608231165865] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND The most used PD fluids contain glucose as a primary osmotic agent. Glucose peritoneal absorption during dwell decreases the osmotic gradient of peritoneal fluids and causes undesirable metabolic consequences. Inhibitors of sodium-glucose co-transporter (SGLT) type 2 are wildly used for the treatment of diabetes, heart and kidney failure. Previous attempts to use SGLT2 blockers in experimental peritoneal dialysis yielded contrasting results. We studied whether peritoneal SGLTs blockade may improve ultrafiltration (UF) via partial inhibition of glucose uptake from dialysis fluids. METHODS Kidney failure was induced in mice and rats by bilateral ureteral ligation, and dwell was performed by injection of glucose-containing dialysis fluids. The effect of SGLT inhibitors on glucose absorption during fluid dwell and UF was measured in vivo. RESULTS Diffusion of glucose from dialysis fluid into the blood appeared to be sodium-dependent, and blockade of SGLTs by phlorizin and sotagliflozin attenuated blood glucose increment thereby decreasing fluid absorption. Specific SGLT2 inhibitors failed to reduce glucose and fluid absorption from the peritoneal cavity in a rodent kidney failure model. CONCLUSIONS Our study suggests that peritoneal non-type 2 SGLTs facilitate glucose diffusion from dialysis solutions, and we propose that limiting glucose reabsorption by specific SGLT inhibitors may emerge as a novel strategy in PD treatment to enhance UF and mitigate the deleterious effects of hyperglycaemia.
Collapse
Affiliation(s)
- Marina Vorobiov
- Department of Nephrology, Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Boris Rogachev
- Department of Nephrology, Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Reut Riff
- Department of Clinical Biochemistry and Pharmacology, Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Cidio Chaimowitz
- Department of Nephrology, Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Endre Z Neulander
- Department of Urology, Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anna Basok
- Department of Nephrology, Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alla Shnaider
- Department of Nephrology, Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Amos Douvdevani
- Department of Nephrology, Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Clinical Biochemistry and Pharmacology, Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yosef-Shmuel Haviv
- Department of Nephrology, Soroka University Medical Center and Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
16
|
Shakour N, Karami S, Iranshahi M, Butler AE, Sahebkar A. Antifibrotic effects of sodium-glucose cotransporter-2 inhibitors: A comprehensive review. Diabetes Metab Syndr 2024; 18:102934. [PMID: 38154403 DOI: 10.1016/j.dsx.2023.102934] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/25/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND AND AIMS Scar tissue accumulation in organs is the underlying cause of many fibrotic diseases. Due to the extensive array of organs affected, the long-term nature of fibrotic processes and the large number of people who suffer from the negative impact of these diseases, they constitute a serious health problem for modern medicine and a huge economic burden on society. Sodium-glucose cotransporter-2 inhibitors (SGLT2is) are a relatively new class of anti-diabetic pharmaceuticals that offer additional benefits over and above their glucose-lowering properties; these medications modulate a variety of diseases, including fibrosis. Herein, we have collated and analyzed all available research on SGLT2is and their effects on organ fibrosis, together with providing a proposed explanation as to the underlying mechanisms. METHODS PubMed, ScienceDirect, Google Scholar and Scopus were searched spanning the period from 2012 until April 2023 to find relevant articles describing the antifibrotic effects of SGLT2is. RESULTS The majority of reports have shown that SGLT2is are protective against lung, liver, heart and kidney fibrosis as well as arterial stiffness. According to the results of clinical trials and animal studies, many SGLT2 inhibitors are promising candidates for the treatment of fibrosis. Recent studies have demonstrated that SGLT2is affect an array of cellular processes, including hypoxia, inflammation, oxidative stress, the renin-angiotensin system and metabolic activities, all of which have been linked to fibrosis. CONCLUSION Extensive evidence indicates that SGLT2is are promising treatments for fibrosis, demonstrating protective effects in various organs and influencing key cellular processes linked to fibrosis.
Collapse
Affiliation(s)
- Neda Shakour
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shima Karami
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Adliya, Bahrain
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
17
|
Ghafouri‐Fard S, Askari A, Shoorei H, Seify M, Koohestanidehaghi Y, Hussen BM, Taheri M, Samsami M. Antioxidant therapy against TGF-β/SMAD pathway involved in organ fibrosis. J Cell Mol Med 2024; 28:e18052. [PMID: 38041559 PMCID: PMC10826439 DOI: 10.1111/jcmm.18052] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 12/03/2023] Open
Abstract
Fibrosis refers to excessive build-up of scar tissue and extracellular matrix components in different organs. In recent years, it has been revealed that different cytokines and chemokines, especially Transforming growth factor beta (TGF-β) is involved in the pathogenesis of fibrosis. It has been shown that TGF-β is upregulated in fibrotic tissues, and contributes to fibrosis by mediating pathways that are related to matrix preservation and fibroblasts differentiation. There is no doubt that antioxidants protect against different inflammatory conditions by reversing the effects of nitrogen, oxygen and sulfur-based reactive elements. Oxidative stress has a direct impact on chronic inflammation, and as results, prolonged inflammation ultimately results in fibrosis. Different types of antioxidants, in the forms of vitamins, natural compounds or synthetic ones, have been proven to be beneficial in the protection against fibrotic conditions both in vitro and in vivo. In this study, we reviewed the role of different compounds with antioxidant activity in induction or inhibition of TGF-β/SMAD signalling pathway, with regard to different fibrotic conditions such as gastro-intestinal fibrosis, cardiac fibrosis, pulmonary fibrosis, skin fibrosis, renal fibrosis and also some rare cases of fibrosis, both in animal models and cell lines.
Collapse
Affiliation(s)
- Soudeh Ghafouri‐Fard
- Department of Medical Genetics, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Arian Askari
- Phytochemistry Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Hamed Shoorei
- Cellular and Molecular Research CenterBirjand University of Medical SciencesBirjandIran
- Clinical Research Development Unit of Tabriz Valiasr HospitalTabriz University of Medical SciencesTabrizIran
| | - Mohammad Seify
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences InstituteShahid Sadoughi University of Medical SciencesYazdIran
| | - Yeganeh Koohestanidehaghi
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences InstituteShahid Sadoughi University of Medical SciencesYazdIran
| | - Bashdar Mahmud Hussen
- Department of Clinical Analysis, College of PharmacyHawler Medical UniversityErbilIraq
| | - Mohammad Taheri
- Institute of Human GeneticsJena University HospitalJenaGermany
- Urology and Nephrology Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Majid Samsami
- Cancer Research Center, Loghman Hakim HospitalShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
18
|
Ye Q, Zha K. SGLT2i‑treated heart failure patients with a reduced ejection fraction: A meta‑analysis. Exp Ther Med 2023; 26:548. [PMID: 37928504 PMCID: PMC10623217 DOI: 10.3892/etm.2023.12248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 09/15/2023] [Indexed: 11/07/2023] Open
Abstract
The aim of this study was to investigate the effects of SGLT2 inhibitors (SGLT2i) on patients with heart failure (HF) and reduced ejection fraction, with or without diabetes. A systematic review of randomized controlled trials (RCTs) was conducted, comparing SGLT2i to a placebo for HF patients. Relevant studies from PubMed, Web of Science, and EMBASE were searched from inception to July 2021, without any language restrictions. The pooled effect was estimated using the odds ratio (OR) and 95% confidence interval (CI). Depending on the heterogeneity test results, either random effects or fixed effects models were selected to estimate the pooled effects. Sensitivity analysis was conducted by gradually removing each study to evaluate the results' stability. A total of 5 RCT studies were included in the analysis. The fixed-effects model demonstrated that the patients in the SGLT2i group had a lower risk of hospitalization for HF/cardiovascular death (OR=0.72; 95% CI, 0.67-0.78), P<0.0001; I2=0.0%, P=0.966), cardiovascular death (OR=0.84, 95% CI (0.77, 0.93), P<0.0001; I2=0.0%, P=0.633), hospitalization for HF (OR=0.69, 95% CI (0.63, 0.75), P<0.0001; I2=0.0%, P=0.933), and all-cause mortality (OR=0.79, 95% CI (0.71, 0.89), P<0.0001; I2=3.3%, P=0.376) compared to the placebo group. Sensitivity analysis showed that the pooled effect value remained stable within the corresponding range, even after each study was gradually removed. In conclusion, SGLT2i can reduce the risk of HF hospitalization, cardiovascular death, and all-cause mortality in patients with HF and a reduced ejection fraction, regardless of the presence or absence of diabetes.
Collapse
Affiliation(s)
- Qiang Ye
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Kelan Zha
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
19
|
Neutel CHG, Wesley CD, Van Praet M, Civati C, Roth L, De Meyer GRY, Martinet W, Guns PJ. Empagliflozin decreases ageing-associated arterial stiffening and vascular fibrosis under normoglycemic conditions. Vascul Pharmacol 2023; 152:107212. [PMID: 37619798 DOI: 10.1016/j.vph.2023.107212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Arterial stiffness is a hallmark of vascular ageing and results in increased blood flow pulsatility to the periphery, damaging end-organs such as the heart, kidneys and brain. Treating or "reversing" arterial stiffness has therefore become a central target in the field of vascular ageing. SGLT2 inhibitors, initially developed in the context of type 2 diabetes mellitus, have become a cornerstone of heart failure treatment. Additionally, effects on the vasculature have been reported. Here, we demonstrate that treatment with the SGLT2 inhibitor empagliflozin (7 weeks, 15 mg/kg/day) decreased ageing-induced arterial stiffness of the aorta in old mice with normal blood glucose levels. However, no universal mechanism was identified. While empagliflozin reduced the ageing-associated increase in collagen type I in the medial layer of the abdominal infrarenal aorta and decreased medial TGF-β deposition, this was not observed in the thoracic descending aorta. Moreover, empagliflozin was not able to prevent elastin fragmentation. In conclusion, empagliflozin decreased arterial stiffness in aged mice, indicating that SGLT2 inhibition could be a valuable strategy in mitigating vascular ageing. Further research is warranted to unravel the underlying, possibly region-specific, mechanisms.
Collapse
Affiliation(s)
- Cédric H G Neutel
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium.
| | - Callan D Wesley
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Melissa Van Praet
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Celine Civati
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| |
Collapse
|
20
|
Xie M, Xia B, Xiao L, Yang D, Li Z, Wang H, Wang X, Zhang X, Peng Q. Astragaloside IV ameliorates peritoneal fibrosis by promoting PGC-1α to reduce apoptosis in vitro and in vivo. J Cell Mol Med 2023; 27:2945-2955. [PMID: 37494130 PMCID: PMC10538260 DOI: 10.1111/jcmm.17871] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 06/24/2023] [Accepted: 07/16/2023] [Indexed: 07/28/2023] Open
Abstract
Prolonged exposure of the peritoneum to high glucose dialysate leads to the development of peritoneal fibrosis (PF), and apoptosis of peritoneal mesothelial cells (PMCs) is a major cause of PF. The aim of this study is to investigate whether Astragaloside IV could protect PMCs from apoptosis and alleviate PF. PMCs and rats PF models were induced by high glucose peritoneal fluid. We examined the pathology of rat peritoneal tissue by HE staining, the thickness of rat peritoneal tissue by Masson's staining, the number of mitochondria and oxidative stress levels in peritoneal tissue by JC-1 and DHE fluorescence staining, and mitochondria-related proteins and apoptosis-related proteins such as PGC-1α, NRF1, TFAM, Caspase3, Bcl2 smad2 were measured. We used hoechst staining and flow cytometry to assess the apoptotic rate of PMCs in the PF model, and further validated the observed changes in the expressions of PGC-1α, NRF1, TFAM, Caspase3, Bcl2 smad2 in PMCs. We further incubated PMCs with MG-132 (proteasome inhibitor) and Cyclohexylamine (protein synthesis inhibitor). The results demonstrated that Astragaloside IV increased the expression of PGC-1α by reducing the ubiquitination of PGC-1α. It was further found that the protective effects of Astragaloside IV on PMCs were blocked when PGC-1α was inhibited. In conclusion, Astragaloside IV effectively alleviated PF both in vitro and in vivo, possibly by promoting PGC-1α to enhance mitochondrial synthesis to reduce apoptotic effects.
Collapse
Affiliation(s)
- Mingxia Xie
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, People's Republic of China
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Bohou Xia
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, People's Republic of China
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Lan Xiao
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, People's Republic of China
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Dun Yang
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, People's Republic of China
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Zhenghong Li
- Departments of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Hanqing Wang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, People's Republic of China
- College of Pharmacy, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Xiaoye Wang
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Xi Zhang
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, People's Republic of China
| | - Qinghua Peng
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, People's Republic of China
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People's Republic of China
| |
Collapse
|
21
|
Li J, Liu Y, Liu J. A review of research progress on mechanisms of peritoneal fibrosis related to peritoneal dialysis. Front Physiol 2023; 14:1220450. [PMID: 37817984 PMCID: PMC10560738 DOI: 10.3389/fphys.2023.1220450] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/13/2023] [Indexed: 10/12/2023] Open
Abstract
Peritoneal dialysis (PD) is an effective alternative treatment for patients with end-stage renal disease (ESRD) and is increasingly being adopted and promoted worldwide. However, as the duration of peritoneal dialysis extends, it can expose problems with dialysis inadequacy and ultrafiltration failure. The exact mechanism and aetiology of ultrafiltration failure have been of great concern, with triggers such as biological incompatibility of peritoneal dialysis solutions, uraemia toxins, and recurrent intraperitoneal inflammation initiating multiple pathways that regulate the release of various cytokines, promote the transcription of fibrosis-related genes, and deposit extracellular matrix. As a result, peritoneal fibrosis occurs. Exploring the pathogenic factors and molecular mechanisms can help us prevent peritoneal fibrosis and prolong the duration of Peritoneal dialysis.
Collapse
Affiliation(s)
- Jin’e Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yinghong Liu
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
22
|
Zhao H, Zhang HL, Jia L. High glucose dialysate-induced peritoneal fibrosis: Pathophysiology, underlying mechanisms and potential therapeutic strategies. Biomed Pharmacother 2023; 165:115246. [PMID: 37523983 DOI: 10.1016/j.biopha.2023.115246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023] Open
Abstract
Peritoneal dialysis is an efficient renal replacement therapy for patients with end-stage kidney disease. However, continuous exposure of the peritoneal membrane to dialysate frequently leads to peritoneal fibrosis, which alters the function of the peritoneal membrane and results in withdrawal from peritoneal dialysis in patients. Among others, high glucose dialysate is considered as a predisposing factor for peritoneal fibrosis in patients on peritoneal dialysis. Glucose-induced inflammation, metabolism disturbance, activation of the renin-angiotensin-aldosterone system, angiogenesis and noninflammation-induced reactive oxygen species are implicated in the pathogenesis of high glucose dialysate-induced peritoneal fibrosis. Specifically, high glucose causes chronic inflammation and recurrent peritonitis, which could cause migration and polarization of inflammatory cells, as well as release of cytokines and fibrosis. High glucose also interferes with lipid metabolism and glycolysis by activating the sterol-regulatory element-binding protein-2/cleavage-activating protein pathway and increasing hypoxia inducible factor-1α expression, leading to angiogenesis and peritoneal fibrosis. Activation of the renin-angiotensin-aldosterone system and Ras-mitogen activated protein kinase signaling pathway is another contributing factor in high glucose dialysate-induced fibrosis. Ultimately, activation of the transforming growth factor-β1/Smad pathway is involved in mesothelial-mesenchymal transition or epithelial-mesenchymal transition, which leads to the development of fibrosis. Although possible intervention strategies for peritoneal dialysate-induced fibrosis by targeting the transforming growth factor-β1/Smad pathway have occasionally been proposed, lack of laboratory evidence renders clinical decision-making difficult. We therefore aim to revisit the upstream pathways of transforming growth factor-beta1/Smad and propose potential therapeutic targets for high glucose-induced peritoneal fibrosis.
Collapse
Affiliation(s)
- Hanxue Zhao
- First Clinical Medical College, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Beijing 100053, China
| | - Hong-Liang Zhang
- Department of Life Sciences, National Natural Science Foundation of China, No. 83 Shuangqing Road, Beijing 100085, China.
| | - Linpei Jia
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Beijing 100053, China.
| |
Collapse
|
23
|
Mo M, Zeng Y, Zeng Y, Li S, He X, Chen X, Luo Q, Liu M, Luo C, Dou X, Peng F, Long H. N-methylpiperazine-diepoxyovatodiolide ameliorates peritoneal fibrosis via suppressing TGF-β/Smad and JAK/STAT signaling pathway. Chem Biol Interact 2023; 382:110589. [PMID: 37268199 DOI: 10.1016/j.cbi.2023.110589] [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: 12/28/2022] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/04/2023]
Abstract
Peritoneal fibrosis (PF) is the main cause of peritoneal ultrafiltration failure in patients undergoing long-term peritoneal dialysis (PD). Epithelial-mesenchymal transition (EMT) is the key pathogenesis of PF. However, currently, no specific treatments are available to suppress PF. N-methylpiperazine-diepoxyovatodiolide (NMPDOva) is a newly synthesized compound that involves a chemical modification of ovatodiolide. In this study, we aimed to explore the antifibrotic effects of NMPDOva in PD-related PF and underlying mechanisms. A mouse model of PD-related PF was established via daily intraperitoneal injection of 4.25% glucose PD fluid. In vitro studies were performed using the transforming growth factor-beta1 (TGF-β1)-stimulated HMrSV5 cell line. Pathological changes were observed, and fibrotic markers were significantly elevated in the peritoneal membrane in mice model of PD-related PF. However, NMPDOva treatment significantly alleviated PD-related PF by decreasing the extracellular matrix accumulation. NMPDOva treatment decreased the expression of fibronectin, collagen Ⅰ, and alpha-smooth muscle actin (α-SMA) in mice with PD-related PF. Moreover, NMPDOva could alleviate TGF-β1-induced EMT in HMrSV5 cells, inhibited phosphorylation and nuclear translocation of Smad2/3, and increased the expression of Smad7. Meanwhile, NMPDOva inhibited phosphorylation of JAK2 and STAT3. Collectively, these results indicated that NMPDOva prevents PD-related PF by inhibiting the TGF-β1/Smad and JAK/STAT signaling pathway. Therefore, because of these antifibrotic effects, NMPDOva may be a promising therapeutic agent for PD-related PF.
Collapse
Affiliation(s)
- Min Mo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, 528308, China
| | - Yao Zeng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yiqun Zeng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Shuting Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaoyang He
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaowen Chen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Qimei Luo
- Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, 528308, China
| | - Mi Liu
- Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, 528308, China
| | - Congwei Luo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xianrui Dou
- Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, 528308, China.
| | - Fenfen Peng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Haibo Long
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
24
|
Lu J, Gao J, Sun J, Wang H, Sun H, Huang Q, Zhang Y, Zhong S. Apolipoprotein A-I attenuates peritoneal fibrosis associated with peritoneal dialysis by inhibiting oxidative stress and inflammation. Front Pharmacol 2023; 14:1106339. [PMID: 37576813 PMCID: PMC10422021 DOI: 10.3389/fphar.2023.1106339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Apolipoprotein A-I (apoA-I), 90% of which is present in high-density lipoprotein (HDL), is the main constituent of HDL, has anti-inflammatory and anti-oxidant properties, and has received extensive attention in anti-atherosclerosis. Yet little is known about apoA-I 's role in peritoneal dialysis. In this study, by analyzing PD patients (n = 81), we found that decreased apoA/HDL-C ratio is significantly associated with rapid decline in peritoneal function. Further studies were performed in animal experiments to determine the ascendancy of apolipoprotein A-I mimetic peptide (D-4F) on peritoneum, we found that D-4F administration reduced peritoneal fibrosis and peritoneal endothelial mesenchymal transformation (EMT) induced by high glucose peritoneal dialysate, such as N-cadherin, Fibronectin, Vimentin, and α-smooth muscle actin (α-SMA) expression decreased. In mechanism, D-4F can significantly inhibit Smad2/3 phosphorylation, which is the major pathway leading to fibrosis. Furthermore, D-4F treatment inhibited NADPH oxidase and thiobarbituric acid reactive substances (TBARS) expression, increased the activity of certain enzymes, such as superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). Finally, treatment with D-4F inhibits the expression of interleukins-6(IL-6), Interleukin-1β(IL-1β), and tumor necrosis factor-α(TNF-α). Taken together, based on the above research evidence, apoA-I and its peptide mimic may regulate the oxidative stress, TGF- β1/Smads signaling pathway and inflammatory response to reduce peritoneal fibrosis due to peritoneal dialysis.
Collapse
Affiliation(s)
- Jing Lu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jie Gao
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jing Sun
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Haiping Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Huijuan Sun
- Jinzhou First People’s Hospital, Dalian, China
| | - Qian Huang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yao Zhang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shuo Zhong
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
25
|
Wang J, Lv X, A-Ni-Wan ASJ, Tian SS, Wang JM, Liu HY, Fan XG, Zhou SJ, Yu P. Canagliflozin alleviates high glucose-induced peritoneal fibrosis via HIF-1α inhibition. Front Pharmacol 2023; 14:1152611. [PMID: 37251320 PMCID: PMC10213900 DOI: 10.3389/fphar.2023.1152611] [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: 01/28/2023] [Accepted: 04/26/2023] [Indexed: 05/31/2023] Open
Abstract
The cardioprotective effects of sodium-glucose cotransporter type 2 (SGLT2) inhibitors have been demonstrated in many studies. However, their benefits for end-stage kidney disease patients, particularly those on peritoneal dialysis, remain unclear. SGLT2 inhibition has shown peritoneal protective effects in some studies, but the mechanisms are still unknown. Herein, we investigated the peritoneal protective mechanisms of Canagliflozin in vitro by simulating hypoxia with CoCl2 in human peritoneal mesothelial cells (HPMCs) and rats by intraperitoneal injection of 4.25% peritoneal dialysate simulating chronic high glucose exposure. CoCl2 hypoxic intervention significantly increased HIF-1α abundance in HPMCs, activated TGF-β/p-Smad3 signaling, and promoted the production of fibrotic proteins (Fibronectin, COL1A2, and α-SMA). Meanwhile, Canagliflozin significantly improved the hypoxia of HPMCs, decreased HIF-1α abundance, inhibited TGF-β/p-Smad3 signaling, and decreased the expression of fibrotic proteins. Five-week intraperitoneal injection of 4.25% peritoneal dialysate remarkably increased peritoneal HIF-1α/TGF-β/p-Smad3 signaling and promoted peritoneal fibrosis and peritoneal thickening. At the same time, Canagliflozin significantly inhibited the HIF-1α/TGF-β/p-Smad3 signaling, prevented peritoneal fibrosis and peritoneal thickening, and improved peritoneal transportation and ultrafiltration. High glucose peritoneal dialysate increased the expression of peritoneal GLUT1, GLUT3 and SGLT2, all of which were inhibited by Canagliflozin. In conclusion, we showed that Canagliflozin could improve peritoneal fibrosis and function by ameliorating peritoneal hypoxia and inhibiting the HIF-1α/TGF-β/p-Smad3 signaling pathway, providing theoretical support for the clinical use of SGLT2 inhibitors in patients on peritoneal dialysis.
Collapse
Affiliation(s)
- Jian Wang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China
- Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xin Lv
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - A-Shan-Jiang A-Ni-Wan
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China
| | - Sha-Sha Tian
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China
| | - Jun-Mei Wang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China
| | - Hong-Yan Liu
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China
| | - Xiao-Guang Fan
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China
- Department of Nephrology, Henan Provincial People’s Hospital, Department of Nephrology of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China
| | - Sai-Jun Zhou
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China
| | - Pei Yu
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China
| |
Collapse
|
26
|
Abstract
Sodium-glucose cotransporter-2 inhibitors (SGLT2 inhibitors) were originally developed as antidiabetic agents, with cardiovascular (CV) outcome trials demonstrating improved CV outcomes in patients with type 2 diabetes mellitus (T2D). Secondary analyses of CV outcome trials and later dedicated kidney outcome trials consistently reported improved kidney-related outcomes independent of T2D status and across a range of kidney function and albuminuria. Importantly, SGLT2 inhibitors are generally safe and well tolerated, with clinical trials and real-world analyses demonstrating a decrease in the risk of acute kidney injury. The kidney protective effects of SGLT2 inhibitors generally extend across different members of the class, possibly on the basis of hemodynamic, metabolic, anti-inflammatory, and antifibrotic mechanisms. In this review, we summarize the effects of SGLT2 inhibitors on kidney outcomes in diverse patient populations.
Collapse
Affiliation(s)
- Atit Dharia
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada; , , , .,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Abid Khan
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada; , , , .,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Vikas S Sridhar
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada; , , , .,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - David Z I Cherney
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada; , , , .,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Shi P, Zhan Z, Ye X, Lu Y, Song K, Sheng F, Shen H, Yin P. The antioxidative effects of empagliflozin on high glucose‑induced epithelial-mesenchymal transition in peritoneal mesothelial cells via the Nrf2/HO-1 signaling. Ren Fail 2022; 44:1528-1542. [PMID: 36098217 PMCID: PMC9481091 DOI: 10.1080/0886022x.2022.2118066] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
High glucose (HG)-induced epithelial-mesenchymal transition (EMT) and oxidative stress play an important role in peritoneal fibrosis, which could be regulated by the nuclear factor erythroid-2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway. This study aimed to investigate whether empagliflozin could inhibit HG-induced EMT and oxidative stress via activating the Nrf2/HO-1 signaling pathway. We used HG-based peritoneal dialysis (PD) solution in rats and HG in human peritoneal mesothelial cells (HPMCs) to induce EMT in vivo and in vitro respectively. The peritoneal structure and function were evaluated by hematoxylin and eosin, Masson's trichrome staining, and the peritoneal equilibrium test. Oxidative stress was measured by assay kits. EMT was analyzed using immunohistochemistry and western blot. The PD rats showed decreased ultrafiltration capacity and increased levels of oxidative stress. Histopathological analysis revealed markedly peritoneal thickening, excessive collagen deposition, increased expression of α-SMA, Collagen-I, and Fibronectin, and decreased expression of E‑cadherin. Empagliflozin significantly ameliorated the aforementioned changes. The protein expression levels of nuclear Nrf2 (N-Nrf2) and HO-1 increased in PD rats, which were further promoted by treatment with empagliflozin. In in vitro experiments, the EMT of HPMCs was induced with 60 mM glucose for 24 h and inhibited by empagliflozin. Empagliflozin suppressed oxidative stress and promoted the protein expression of N-Nrf2 and HO-1 in HG‑stimulated HPMCs, which was reversed by the Nrf2 inhibitor. In conclusion, empagliflozin exerted a protective effect against HG-induced EMT and suppressed oxidative stress in PMCs by activating the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Ping Shi
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhoubing Zhan
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaojie Ye
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ying Lu
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Kai Song
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng Sheng
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Huaying Shen
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China, Huaying Shen Department of Nephrology, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Jinchang, Suzhou, 215000, Jiangsu, China
| | - Peiran Yin
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China,CONTACT Peiran Yin
| |
Collapse
|
28
|
Sridhar VS, Bargman JM. The Sweet Science of Glucose Transport. J Am Soc Nephrol 2022; 33:1803-1804. [PMID: 36630519 PMCID: PMC9528328 DOI: 10.1681/asn.2022070841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Vikas S. Sridhar
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Joanne M. Bargman
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
29
|
SGLT2 inhibitors attenuate nephrin loss and enhance TGF-β 1 secretion in type 2 diabetes patients with albuminuria: a randomized clinical trial. Sci Rep 2022; 12:15695. [PMID: 36127497 PMCID: PMC9489863 DOI: 10.1038/s41598-022-19988-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/07/2022] [Indexed: 11/15/2022] Open
Abstract
To evaluate the effect of SGLT2 inhibitor (SGLT2i) on albuminuria, nephrin (NPH) and transforming-growth-factor-beta1 (TGF-β1) levels in urine and low-grade inflammation in type 2 diabetes (T2D) patients. A randomized, blank-controlled clinical trial included 68 T2D patients and 10 controls. Based on the urinary albumin-to-creatinine ratio (UACR), 68 diabetic patients were stratified into three levels, UACR < 30 mg/g, UACR ≧ 30 mg/g to ≦ 300 mg/g and UACR ˃ 300 mg/g, who were randomized (1:1:1) to receive SGLT2i treatment for 12 weeks. The concentrations of NPH and TGF-β1 in urine were measured as indications of podocyte injury and renal fibrosis. Low-grade inflammation was assessed by the levels of IL-6, TNFα and hsCRP. After 12 weeks of SGLT2i treatment, the levels of UACR and NPH decreased, UTGF-β1 increased in the T2D with microalbuminuria and macroalbuminuria groups, NPH (1.12 [0.59, 1.29] vs. 0.71 [0.41, 1.07] µg/ml, P = 0.022) and (1.29 [0.99, 1.96] vs. 0.93 [0.57, 1.31] µg/ml, P = 0.002), UTGF-β1 (4.88 ± 1.31 vs. 7.27 ± 1.21 pg/ml, P < 0.001) and (4.30 ± 1.34 vs. 6.78 ± 2.59 pg/ml, P < 0.001), respectively. The changes in NPH were positively correlated with the UACR and negatively correlated with UTGF-β1 in T2D with albuminuria. SGLT2i alleviate nephrin loss and enhance TGF-β1 excretion in urine in T2DM with albuminuria. The anti-albuminuric effect of SGLT2i could be attributed to mitigating podocyte apoptosis and attenuating renal fibrosis. Trial registration This clinical trial was registered on 15/10/2019, in ClinicalTrials.gov, and the registry number is NCT04127084.
Collapse
|
30
|
Li J, Zhou L, Gong H. New insights and advances of sodium-glucose cotransporter 2 inhibitors in heart failure. Front Cardiovasc Med 2022; 9:903902. [PMID: 36186974 PMCID: PMC9520058 DOI: 10.3389/fcvm.2022.903902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2is) are newly emerging insulin-independent anti-hyperglycemic agents that work independently of β-cells. Quite a few large-scale clinical trials have proven the cardiovascular protective function of SGLT2is in both diabetic and non-diabetic patients. By searching all relevant terms related to our topics over the previous 3 years, including all the names of agents and their brands in PubMed, here we review the mechanisms underlying the improvement of heart failure. We also discuss the interaction of various mechanisms proposed by diverse works of literature, including corresponding and opposing viewpoints to support each subtopic. The regulation of diuresis, sodium excretion, weight loss, better blood pressure control, stimulation of hematocrit and erythropoietin, metabolism remodeling, protection from structural dysregulation, and other potential mechanisms of SGLT2i contributing to heart failure improvement have all been discussed in this manuscript. Although some remain debatable or even contradictory, those newly emerging agents hold great promise for the future in cardiology-related therapies, and more research needs to be conducted to confirm their functionality, particularly in metabolism, Na+-H+ exchange protein, and myeloid angiogenic cells.
Collapse
Affiliation(s)
- Juexing Li
- Department of Cardiology, Jinshan Hospital of Fudan University, Shanghai, China
- Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Zhou
- Department of Cardiology, Jinshan Hospital of Fudan University, Shanghai, China
- Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Gong
- Department of Cardiology, Jinshan Hospital of Fudan University, Shanghai, China
- Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Hui Gong
| |
Collapse
|
31
|
Jing Y, Yang R, Chen W, Ye Q. Anti-Arrhythmic Effects of Sodium-Glucose Co-Transporter 2 Inhibitors. Front Pharmacol 2022; 13:898718. [PMID: 35814223 PMCID: PMC9263384 DOI: 10.3389/fphar.2022.898718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/04/2022] [Indexed: 12/11/2022] Open
Abstract
Arrhythmias are clinically prevalent with a high mortality rate. They impose a huge economic burden, thereby substantially affecting the quality of life. Sodium-glucose co-transporter 2 inhibitor (SGLT2i) is a new type of hypoglycemic drug, which can regulate blood glucose level safely and effectively. Additionally, it reduces the occurrence and progression of heart failure and cardiovascular events significantly. Recently, studies have found that SGLT2i can alleviate the occurrence and progression of cardiac arrhythmias; however, the exact mechanism remains unclear. In this review, we aimed to discuss and summarize new literature on different modes in which SGLT2i ameliorates the occurrence and development of cardiac arrhythmias.
Collapse
|
32
|
Li ZH, Xu R, Shi J, Yu MS, Zhong Y, He WM, Cao LP, Sheng MX. Astragalus Total Saponins Ameliorate Peritoneal Fibrosis by Promoting Mitochondrial Synthesis and Inhibiting Apoptosis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:261-274. [PMID: 34983328 DOI: 10.1142/s0192415x22500094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Peritoneal fibrosis (PF) is a disease caused by prolonged exposure of the peritoneum to high levels of dialysis fluid. Astragalus total saponins (ATS) is a phytochemical naturally occurring in Radix Astragali that has anti-inflammatory and anti-oxidant properties. In this study, we constructed an in vivo model of PF using 4.25% glucose-containing administered intraperitoneally to rats and incubated peritoneal mesothelial cells (PMCs) with 4.25% glucose-containing peritoneal dialysis fluid to construct an in vitro model of PF. Furthermore, siRNA of PGC-1[Formula: see text] was used to inhibit the expression of PGC-1[Formula: see text] to further investigate the mechanism of the protective effect of ATS on PF. In both in vivo and in vitro models, ATS treatment showed a protective effect against PF, with ATS reducing the thickness of peritoneal tissues in PF rats, increasing the viability of PMCs, increasing the mitochondrial membrane potential and reducing apoptosis ratio. ATS treatment also reduced the expressions of peritoneal fibrosis markers (Smad2, p-Smad2 and [Formula: see text]-SMA) and apoptosis markers (Caspase3, cleaved-Caspase3 and Bax) and restored the expressions of mitochondrial synthesis proteins (PGC-1[Formula: see text], NRF1 and TFAM) in ATS-treated peritoneal tissues or PMCs. Furthermore, in the presence of PGC-1[Formula: see text] inhibition, the protective effect of ATS on PF was blocked. In conclusion, ATS treatment may be an effective therapeutic agent to inhibit high glucose-induced in peritoneal fibrosis through PGC-1[Formula: see text]-mediated apoptosis.
Collapse
Affiliation(s)
- Zheng-Hong Li
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P. R. China
| | - Rong Xu
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P. R. China
| | - Jun Shi
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P. R. China
| | - Man-Shu Yu
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P. R. China
| | - Yu Zhong
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P. R. China
| | - Wei-Ming He
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P. R. China
| | - Li-Ping Cao
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P. R. China
| | - Mei-Xiao Sheng
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P. R. China
| |
Collapse
|
33
|
A Role for SGLT-2 Inhibitors in Treating Non-diabetic Chronic Kidney Disease. Drugs 2021; 81:1491-1511. [PMID: 34363606 DOI: 10.1007/s40265-021-01573-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
In recent years, inhibitors of the sodium-glucose co-transporter 2 (SGLT2 inhibitors) have been shown to have significant protective effects on the kidney and the cardiovascular system in patients with diabetes. This effect is also manifested in chronic kidney disease (CKD) patients and is minimally due to improved glycaemic control. Starting from these positive findings, SGLT2 inhibitors have also been tested in patients with non-diabetic CKD or heart failure with reduced ejection fraction. Recently, the DAPA-CKD trial showed a significantly lower risk of CKD progression or death from renal or cardiovascular causes in a mixed population of patients with diabetic and non-diabetic CKD receiving dapagliflozin in comparison with placebo. In patients with heart failure and reduced ejection fraction, two trials (EMPEROR-Reduced and DAPA-HF) also found a significantly lower risk of reaching the secondary renal endpoint in those treated with an SGLT2 inhibitor in comparison with placebo. This also applied to patients with CKD. Apart from their direct mechanism of action, SGLT2 inhibitors have additional effects that could be of particular interest for patients with non-diabetic CKD. Among these, SGLT2 inhibitors reduce blood pressure and serum acid uric levels and can increase hemoglobin levels. Some safety issues should be further explored in the CKD population. SGLT2 inhibitors can minimally increase potassium levels, but this has not been shown by the CREDENCE trial. They also increase magnesium and phosphate reabsorption. These effects could become more significant in patients with advanced CKD and will need monitoring when these agents are used more extensively in the CKD population. Conversely, they do not seem to increase the risk of acute kidney injury.
Collapse
|
34
|
Wu H, Wang D, Shi H, Liu N, Wang C, Tian J, Wang X, Zhang Z. PM 2.5 and water-soluble components induce airway fibrosis through TGF-β1/Smad3 signaling pathway in asthmatic rats. Mol Immunol 2021; 137:1-10. [PMID: 34175710 DOI: 10.1016/j.molimm.2021.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 01/12/2023]
Abstract
Epidemiological studies have suggested that fine particulate matter (PM2.5) and asthma have been independently associated with pulmonary fibrosis but rarely studied together. Furthermore, it is unknown whether airway fibrosis in asthma is more attributable to water-soluble ions of PM2.5. Our current study was to explore the potential mechanism of PM2.5 and water-soluble components on airway fibrosis in ovalbumin (OVA)-sensitized asthmatic rats. Rats were intratracheally instilled with PM2.5 and water-soluble components every 3 days for 4 times or 8 times. Histopathological examination demonstrated that lung inflammatory and airway fibrosis were induced after PM2.5 and water-soluble components exposure. Meanwhile, PM2.5, in particular water-soluble extracts, increased expression of collagen 1 (COL-1), connective tissue growth factor (CTGF), interleukin-6 (IL-6), transforming growth factor-β1 (TGF-β1), Smad family member 3 (Smad3), and p-Smad3, whereas decreased secretion of heme oxygenase-1 (HO-1). However, pretreating asthmatic rats with SB432542, the inhibitor of TGF-β1, and SIS3 HCl, the antagonist of Smad3, both reversed the activation of airway fibrosis induced by water-soluble extracts. Therefore, TGF-β1/Smad3 signaling pathway may be responsible for the pathological process of airway fibrosis in asthmatic rats following PM2.5 and water-soluble components exposure.
Collapse
Affiliation(s)
- Hongyan Wu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Dan Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Hao Shi
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Nannan Liu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Caihong Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Jiayu Tian
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Xin Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China
| | - Zhihong Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, China.
| |
Collapse
|
35
|
Pan D, Xu L, Chen P, Jiang H, Shi D, Guo M. Empagliflozin in Patients With Heart Failure: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front Cardiovasc Med 2021; 8:683281. [PMID: 34239906 PMCID: PMC8257947 DOI: 10.3389/fcvm.2021.683281] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 04/30/2021] [Indexed: 12/29/2022] Open
Abstract
Purpose: The purpose of the study is to evaluate the effect of empagliflozin in patients with heart failure (HF). Method: We performed a systematic search of PubMed, EMBASE, and the Cochrane Library database through January 20, 2021. Randomized controlled trials (RCTs) were included that compared empagliflozin and placebo in patients with HF. Dichotomous variables were expressed as risk ratios (RRs) with 95% confidence intervals (CIs). Continuous variables were calculated and expressed as mean differences (MD) and standard deviation (SD). Meta-analysis was conducted using a random-effects model on outcomes with high heterogeneity. Results: Seven studies were included in our meta-analysis (n = 5,150). Significant differences were observed in a composite of cardiovascular death or hospitalization for worsening heart failure [RR: 0.77 (95% CI 0.68-0.87); I 2 = 18%; P < 0.0001), hospitalization for worsening heart failure [RR: 0.71 (95% CI 0.61-0.82); I 2 = 0%; P < 0.00001], changes in Kansas City Cardiomyopathy Questionnaire (KCCQ) score [MD: 1.70 (95% CI 1.67-1.73); I 2 = 0%; P < 0.00001], and changes in body weight [MD: -1.43 (95% CI -2.15 to -0.72); I 2 = 84%; P < 0.0001) from baseline. However, empagliflozin did not show a better change in the 6-min walk test (6MWT) [MD: 34.06 (95% CI -29.75-97.88); I 2 = 97%; P = 0.30] or NT-proBNP [MD: -98.36 (95% CI, -225.83-29.11); I 2 = 68%; P = 0.13] from baseline. Conclusion: The findings suggest that empagliflozin was effective in reducing a composite of cardiovascular death or hospitalization for worsening heart failure. Further well-designed RCTs are needed to evaluate the long-term effect of empagliflozin in patients with HF. PROSPERO: CRD42021231712.
Collapse
Affiliation(s)
- Deng Pan
- Graduate School of Beijing University of Traditional Chinese Medicine, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lin Xu
- Gynecological Department of Traditional Chinese Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Pengfei Chen
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Graduate School of China Academy of Chinese Medical Sciences, Beijing, China
| | | | - Dazhuo Shi
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ming Guo
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|