1
|
Seghers F, Tintillier M, Morelle J. Recent advances in the understanding of the peritoneal membrane. Curr Opin Nephrol Hypertens 2025; 34:77-84. [PMID: 39291741 DOI: 10.1097/mnh.0000000000001028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
PURPOSE OF REVIEW The efficiency of peritoneal dialysis (PD) as a life-sustaining replacement therapy for patients with kidney failure relies on the integrity and function of the peritoneal membrane. Here, we review the most recent advances in the understanding of the peritoneal membrane and its role in PD. RECENT FINDINGS A recent update of the ISPD guidelines proposed a revised definition of membrane dysfunction, emphasizing the importance of fluid balance in patients treated with PD and identified three main mechanisms leading to insufficient peritoneal ultrafiltration (UF). The Bio-PD study, the first genomewide association study in PD, demonstrated that 20% of the interindividual variability in the peritoneal solute transfer rate is genetically determined, and identified several loci of potential relevance for peritoneal transport. A candidate-gene approach identified and characterized a common and functional variant in the promoter of the AQP1 gene associated with water transport and clinical outcomes in PD. Innovative strategies to preserve the integrity of the peritoneal membrane and to enhance UF are also discussed, including the use of gliflozins; steady glucose concentration PD; modulation of GLUT proteins; and cytoprotective additives. SUMMARY A comprehensive understanding of the peritoneal membrane and of the mechanisms driving UF may help individualizing PD prescription and improving outcomes in patients treated with PD.
Collapse
Affiliation(s)
- François Seghers
- Division of Nephrology, University Hospitals Namur (CHU UCL Namur), Namur
| | - Michel Tintillier
- Division of Nephrology, University Hospitals Namur (CHU UCL Namur), Namur
| | - Johann Morelle
- Division of Nephrology, University Hospitals Namur (CHU UCL Namur), Namur
- de Duve Institute, UCLouvain, Brussels, Belgium
| |
Collapse
|
2
|
Wang Y, Zhang Y, Ma M, Zhuang X, Lu Y, Miao L, Lu X, Cui Y, Cui W. Mechanisms underlying the involvement of peritoneal macrophages in the pathogenesis and novel therapeutic strategies for dialysis-induced peritoneal fibrosis. Front Immunol 2024; 15:1507265. [PMID: 39749340 PMCID: PMC11693514 DOI: 10.3389/fimmu.2024.1507265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Long-term exposure of the peritoneum to peritoneal dialysate results in pathophysiological changes in the anatomical organization of the peritoneum and progressive development of peritoneal fibrosis. This leads to a decline in peritoneal function and ultrafiltration failure, ultimately necessitating the discontinuation of peritoneal dialysis, severely limiting the potential for long-term maintenance. Additionally, encapsulating peritoneal sclerosis, a serious consequence of peritoneal fibrosis, resulting in patients discontinuing PD and significant mortality. The causes and mechanisms underlying peritoneal fibrosis in patients undergoing peritoneal dialysis remain unknown, with no definitive treatment available. However, abnormal activation of the immune system appears to be involved in altering the structure of the peritoneum and promoting fibrotic changes. Macrophage infiltration and polarization are key contributors to pathological injury within the peritoneum, showing a strong correlation with the epithelial-to-mesenchymal transition of mesothelial cells and driving the process of fibrosis. This article discusses the role and mechanisms underlying macrophage activation-induced peritoneal fibrosis resulting from PD by analyzing relevant literature from the past decade and provides an overview of recent therapeutic approaches targeting macrophages to treat this condition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yingchun Cui
- Department of Nephrology, Second Hospital of Jilin University,
Changchun, China
| | - Wenpeng Cui
- Department of Nephrology, Second Hospital of Jilin University,
Changchun, China
| |
Collapse
|
3
|
Martus G, Siddhuraj P, Erjefält JS, Kádár A, Lindström M, Bergling K, Öberg CM. Transcellular transport of 18F-deoxyglucose via facilitative glucose channels in experimental peritoneal dialysis. Perit Dial Int 2024:8968608241299928. [PMID: 39636030 DOI: 10.1177/08968608241299928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Local and systemic side effects of glucose remain major limitations of peritoneal dialysis (PD). Glucose transport during PD is thought to occur via inter-endothelial pathways, but recent results show that phloretin, a general blocker of facilitative glucose channels (glucose transporters [GLUTs]), markedly reduced glucose diffusion capacity indicating that some glucose may be transferred via facilitative glucose channels (GLUTs). Whether such transport mainly occurs into (absorption), or across (trans-cellular) peritoneal cells is as yet unresolved. METHODS Here we sought to elucidate whether diffusion of radiolabeled 18F-deoxyglucose ([18F]-DG) in the opposite direction (plasma → dialysate) is also affected by GLUT inhibition. During GLUT inhibition, such transport may either be increased or unaltered (favors absorption hypothesis) or decreased (favors transcellular hypothesis). Effects on the transport of solutes other than [18F]-DG (or glucose) during GLUT inhibition indicate effects on paracellular transport (between cells) rather than via GLUTs. RESULTS GLUT inhibition using phloretin markedly reduced [18F]-DG diffusion capacity, improved ultrafiltration (UF) rates and enhanced the sodium dip. No other solutes were significantly affected with the exception of urea and bicarbonate. CONCLUSION The present results indicate that part of glucose is transported via the transcellular route across cells in the peritoneal membrane. Regardless of the channel(s) involved, inhibitors of facilitative GLUTs may be promising agents to improve UF efficacy in patients treated with PD.
Collapse
Affiliation(s)
- Giedre Martus
- Nephrology Division, Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| | - Premkumar Siddhuraj
- Unit of Airway Inflammation, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Jonas S Erjefält
- Unit of Airway Inflammation, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
- Department of Allergology and Respiratory Medicine, Skåne University Hospital, Lund University, Lund, Sweden
| | - András Kádár
- Nephrology Division, Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| | - Martin Lindström
- Department of Laboratory Medicine, Lund University, Malmö, Sweden
- Centre for Molecular Pathology, Skåne University Hospital, Malmö, Sweden
| | - Karin Bergling
- Nephrology Division, Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| | - Carl M Öberg
- Nephrology Division, Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Lotfollahzadeh S, Vazirani A, Sellinger IE, Clovie J, Hoekstra I, Patel A, Malloum AB, Yin W, Paul H, Yadati P, Siracus J, Malikova M, Pernar LI, Francis J, Stern L, Chitalia VC. Aryl Hydrocarbon Receptor Pathway Augments Peritoneal Fibrosis in a Murine CKD Model Exposed to Peritoneal Dialysate. KIDNEY360 2024; 5:1238-1250. [PMID: 39235862 PMCID: PMC11441816 DOI: 10.34067/kid.0000000000000516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/11/2024] [Indexed: 09/07/2024]
Abstract
Key Points CKD and high glucose–containing peritoneal dialysate alter peritoneal membrane contributing to peritoneal dialysis failure, with a poorly understood mechanism. CKD milieu activates the aryl hydrocarbon receptor pathway in the subperitoneal vasculature, increasing the peritoneal fibrosis and collagen deposition in humans and mice. An aryl hydrocarbon receptor inhibitor mitigates CKD and peritoneal dialysis–mediated peritoneal fibrosis, collagen deposition, and vasculogenesis in a mouse model. Background CKD is a proinflammatory and profibrotic condition and can independently alter the peritoneal membrane structure. Peritoneal dialysis (PD) results in profound alterations in the peritoneal membrane. The mechanisms contributing to the alterations of the peritoneal membrane structure in CKD milieu, along with PD, are poorly understood. Methods Here, we show that human CKD induces peritoneal membrane thickening, fibrosis, and collagen deposition and activates the aryl hydrocarbon receptor (AHR) pathway in the subperitoneal vasculature. Leveraging a novel model of PD in CKD mice, we confirm these CKD-induced changes in the peritoneal membrane, which are exacerbated on exposure to the peritoneal dialysate. Peritoneal dialysate further augmented the AHR activity in endothelial cells of peritoneal microvasculature in CKD mice. Results Treatment of CKD mice with an AHR inhibitor in peritoneal dialysate for 2 weeks resulted in a seven-fold reduction in AHR expression in the endothelial cells of subperitoneal capillaries, a five-fold decrease in subperitoneal space, and a nine-fold decrease in fibrosis and collagen deposition compared with vehicle-treated CKD mice. AHR inhibition reduced inflammation, subperitoneal neovascular areas, and its downstream target, tissue factor. The AHR inhibitor treatment normalized the peritoneal dialysate-induced proinflammatory and profibrotic cytokines, such as IL-6, monocyte chemoattractant protein-1, and macrophage inflammatory protein 1 levels, in CKD mice. Conclusions This study uncovers the activation of the AHR-cytokine axis in the endothelial cells of subperitoneal vessels in humans and mice with CKD, which is likely to prime the peritoneal membrane to peritoneal dialysate–mediated alterations. This study supports further exploration of AHR as a potential therapeutic target to preserve the structural and functional integrity of the peritoneal membrane in PD.
Collapse
Affiliation(s)
- Saran Lotfollahzadeh
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Aniket Vazirani
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Isaac E. Sellinger
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Janelle Clovie
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Isaac Hoekstra
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Arjun Patel
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Abbas Brahim Malloum
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Wenqing Yin
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Herreet Paul
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Pranav Yadati
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Jeffrey Siracus
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Marina Malikova
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Luise I. Pernar
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Jean Francis
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Lauren Stern
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | - Vipul C. Chitalia
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
7
|
Lai JW, Wang CCN, Chou CY. SGLT-2 inhibitors in chronic peritoneal dialysis patients: a follow-up study. BMC Nephrol 2024; 25:238. [PMID: 39075357 PMCID: PMC11285309 DOI: 10.1186/s12882-024-03683-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: 04/22/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Sodium-glucose transporter-2 inhibitors (SGLT-2i) are recommended for use in patients with type 2 diabetes comorbid atherosclerotic cardiovascular disease, heart failure, or chronic kidney disease. Limited reports are currently available for their use in dialysis patients. In an observational, retrospective follow-up study, we reported the clinical characteristics of chronic peritoneal dialysis (PD) patients on SGLT-2i. METHODS We enrolled 50 diabetic chronic PD patients, and 11 continued SGLT-2i after PD treatment. We reported the patients' ultrafiltration, HbA1c, urinary tract infection episodes, and venous CO2 during follow-up and compared the differences in these factors between patients with and without SGLT-2i. RESULTS The mean age of the patients was 65 ± 15 years, and 16 (32%) patients were female. The age, gender, heart failure, and primary kidney disease were not different between patients with and without SGLT-2i at enrollment. In an average of 31 months follow-up, patients with SGLT-2i had higher ultrafiltration (1322 ± 200 ml/day vs. 985 ± 415 ml/day, p = 0.013), hemoglobin (11.2 ± 1.7 vs. 10.2 ± 1.7 g/dl), white blood cell count (9.2 ± 3.7 vs. 7.4 ± 2.1 109/L), and a lower venous CO2 (p = 0.036). The urine amount, the overall survival, the technical survival, and the chance of UTI were not different between patients with and without SGLT2i. CONCLUSION SGLT-2i may increase ultrafiltration volume and hemoglobin levels in chronic PD patients. SGLT-2i did not increase urinary tract infection but was linked to subclinical metabolic acidosis. WHAT WAS KNOWN The effect of SGLT-2i in chronic PD patients is not clear? THIS STUDY ADDS SGLT-2i is associated with increased ultrafiltration, hemoglobin, white blood cell counts, and a decreased CO2 in PD patient. POTENTIAL IMPACT SGLT-2i may increase ultrafiltration in PD patients.
Collapse
Affiliation(s)
- Jia-Wen Lai
- Division of Nephrology, Asia University Hospital, Taichung, Taiwan
| | - Charles C N Wang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Che-Yi Chou
- Division of Nephrology, Asia University Hospital, Taichung, Taiwan.
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan.
- College of Medicine, China Medical University, Taichung, Taiwan.
- Division of Nephrology and Kidney Institute, China Medical University Hospital, Taichung, Taiwan.
| |
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
|
Zhang R, Xie Q, Lu X, Fan R, Tong N. Research advances in the anti-inflammatory effects of SGLT inhibitors in type 2 diabetes mellitus. Diabetol Metab Syndr 2024; 16:99. [PMID: 38735956 PMCID: PMC11089742 DOI: 10.1186/s13098-024-01325-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 03/28/2024] [Indexed: 05/14/2024] Open
Abstract
Diabetes mellitus is one of the most significant global burden diseases. It is well established that a chronic, systemic, low-grade inflammatory condition is strongly correlated with type 2 diabetes mellitus (T2D) and the development of target-organ damage (TOD). Sodium-glucose cotransporter inhibitors (SGLTis), novel oral drugs for the treatment of diabetes, act mainly by reducing glucose reabsorption in proximal renal tubules and/or the intestine. Several high-quality clinical trials and large observational studies have revealed that SGLTis significantly improve cardiovascular and renal outcomes in T2D patients. Increasing evidence suggests that this is closely related to their anti-inflammatory properties, which are mainly manifested by a reduction in plasma concentrations of inflammatory biomarkers. This review analyses the potential mechanisms behind the anti-inflammatory effects of SGLTis in diabetes and presents recent evidence of their therapeutic efficacy in treating diabetes and related TOD.
Collapse
Affiliation(s)
- Ruining Zhang
- Department of Endocrinology, Center for Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Qingxing Xie
- Department of Endocrinology, Center for Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Xi Lu
- Department of Endocrinology, Center for Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Rongping Fan
- Department of Endocrinology, Center for Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China
| | - Nanwei Tong
- Department of Endocrinology, Center for Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
11
|
Hu J, Teng J, Hui S, Liang L. SGLT-2 inhibitors as novel treatments of multiple organ fibrosis. Heliyon 2024; 10:e29486. [PMID: 38644817 PMCID: PMC11031788 DOI: 10.1016/j.heliyon.2024.e29486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/23/2024] Open
Abstract
Fibrosis, a significant health issue linked to chronic inflammatory diseases, affects various organs and can lead to serious damage and loss of function. Despite the availability of some treatments, their limitations necessitate the development of new therapeutic options. Sodium-glucose cotransporter 2 inhibitors (SGLT2i), known for their glucose-lowering ability, have shown promise in offering protective effects against fibrosis in multiple organs through glucose-independent mechanisms. This review explores the anti-fibrotic potential of SGLT2i across different tissues, providing insights into their underlying mechanisms and highlighting recent research advancements. The evidence positions SGLT2i as a potential future treatments for fibrotic diseases.
Collapse
Affiliation(s)
- Junpei Hu
- Department of Geriatrics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, China
| | - Jianhui Teng
- Department of Geriatrics, Hunan Provincial People's Hospital, China
| | - Shan Hui
- Department of Geriatrics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, China
| | - Lihui Liang
- Department of Geriatrics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, China
| |
Collapse
|
12
|
Song Q, Wang P, Wang H, Pan M, Li X, Yao Z, Wang W, Tang G, Zhou S. Integrative analysis of chromatin accessibility and transcriptome landscapes in the induction of peritoneal fibrosis by high glucose. J Transl Med 2024; 22:243. [PMID: 38443979 PMCID: PMC10916192 DOI: 10.1186/s12967-024-05037-6] [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: 09/12/2023] [Accepted: 02/24/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Peritoneal fibrosis is the prevailing complication induced by prolonged exposure to high glucose in patients undergoing peritoneal dialysis. METHODS To elucidate the molecular mechanisms underlying this process, we conducted an integrated analysis of the transcriptome and chromatin accessibility profiles of human peritoneal mesothelial cells (HMrSV5) during high-glucose treatment. RESULTS Our study identified 2775 differentially expressed genes (DEGs) related to high glucose-triggered pathological changes, including 1164 upregulated and 1611 downregulated genes. Genome-wide DEGs and network analysis revealed enrichment in the epithelial-mesenchymal transition (EMT), inflammatory response, hypoxia, and TGF-beta pathways. The enriched genes included VEGFA, HIF-1α, TGF-β1, EGF, TWIST2, and SNAI2. Using ATAC-seq, we identified 942 hyper (higher ATAC-seq signal in high glucose-treated HMrSV5 cells than in control cells) and 714 hypo (lower ATAC-seq signal in high glucose-treated HMrSV5 cells versus control cells) peaks with differential accessibility in high glucose-treated HMrSV5 cells versus controls. These differentially accessible regions were positively correlated (R = 0.934) with the nearest DEGs. These genes were associated with 566 up- and 398 downregulated genes, including SNAI2, TGF-β1, HIF-1α, FGF2, VEGFA, and VEGFC, which are involved in critical pathways identified by transcriptome analysis. Integrated ATAC-seq and RNA-seq analysis also revealed key transcription factors (TFs), such as HIF-1α, ARNTL, ELF1, SMAD3 and XBP1. Importantly, we demonstrated that HIF-1α is involved in the regulation of several key genes associated with EMT and the TGF-beta pathway. Notably, we predicted and experimentally validated that HIF-1α can exacerbate the expression of TGF-β1 in a high glucose-dependent manner, revealing a novel role of HIF-1α in high glucose-induced pathological changes in human peritoneal mesothelial cells (HPMCs). CONCLUSIONS In summary, our study provides a comprehensive view of the role of transcriptome deregulation and chromosome accessibility alterations in high glucose-induced pathological fibrotic changes in HPMCs. This analysis identified hub genes, signaling pathways, and key transcription factors involved in peritoneal fibrosis and highlighted the novel glucose-dependent regulation of TGF-β1 by HIF-1α. This integrated approach has offered a deeper understanding of the pathogenesis of peritoneal fibrosis and has indicated potential therapeutic targets for intervention.
Collapse
Affiliation(s)
- Qiong Song
- Department of Nephrology, Shaanxi Second People's Hospital, Xi'an, Shaanxi, People's Republic of China
| | - Pengbo Wang
- Department of Nephrology, Shaanxi Second People's Hospital, Xi'an, Shaanxi, People's Republic of China
| | - Huan Wang
- Department of Emergency, Xijing Hospital, The Fourth Military Medical University of People's Liberation Army, Xi'an, Shaanxi, People's Republic of China
| | - Meijing Pan
- Department of Clinical Medicine, Xi'an Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Xiujuan Li
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Zhuan'e Yao
- Department of Nephrology, Shaanxi Second People's Hospital, Xi'an, Shaanxi, People's Republic of China
| | - Wei Wang
- Department of Nephrology, Shaanxi Second People's Hospital, Xi'an, Shaanxi, People's Republic of China
| | - Guangbo Tang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.
| | - Sen Zhou
- Department of Nephrology, The First Hospital of Lanzhou University, Lanzhou, Gansu, People's Republic of China.
| |
Collapse
|
13
|
Morelle J, Lambie M, Öberg CM, Davies S. The Peritoneal Membrane and Its Role in Peritoneal Dialysis. Clin J Am Soc Nephrol 2024; 19:244-253. [PMID: 37616463 PMCID: PMC10861113 DOI: 10.2215/cjn.0000000000000282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023]
Abstract
A healthy and functional peritoneal membrane is key to achieving sufficient ultrafiltration and restoring fluid balance, a major component of high-quality prescription in patients treated with peritoneal dialysis (PD). Variability in membrane function at the start of PD or changes over time on treatment influence dialysis prescription and outcomes, and dysfunction of the peritoneal membrane contributes to fluid overload and associated complications. In this review, we summarize the current knowledge about the structure, function, and pathophysiology of the peritoneal membrane with a focus on clinical implications for patient-centered care. We also discuss the molecular and genetic mechanisms of solute and water transport across the peritoneal membrane, including the role of aquaporin water channels in crystalloid versus colloid osmosis; why and how to assess membrane function using peritoneal equilibration tests; the etiologies of membrane dysfunction and their specific management; and the effect of genetic variation on membrane function and outcomes in patients treated with PD. This review also identifies the gaps in current knowledge and perspectives for future research to improve our understanding of the peritoneal membrane and, ultimately, the care of patients treated with PD.
Collapse
Affiliation(s)
- Johann Morelle
- Division of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium
- UCLouvain School of Medicine, UCLouvain, Brussels, Belgium
| | - Mark Lambie
- Faculty of Medicine and Health Sciences, Keele University, Keele, United Kingdom
| | - Carl M. Öberg
- Division of Nephrology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Simon Davies
- Faculty of Medicine and Health Sciences, Keele University, Keele, United Kingdom
| |
Collapse
|
14
|
Hamdan Z, Abdel-Hafez Y, Enaya A, Sarsour A, Kharraz L, Nazzal Z. Dapagliflozin in peritoneal dialysis patients: a pilot study evaluating peritoneal membrane function. BMC Nephrol 2024; 25:37. [PMID: 38279109 PMCID: PMC10811926 DOI: 10.1186/s12882-023-03429-2] [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: 09/09/2023] [Accepted: 12/06/2023] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND Patients taking SGLT-2 inhibitors may experience delayed peritoneal fibrosis, better ultrafiltration of water and toxins, and higher survival rates. We aimed to evaluate the possible effects of Dapagliflozin in changing the peritoneal solute transfer rate, reducing peritoneal glucose absorption, and, hence, increasing ultrafiltration. METHODOLOGY A pilot pre-post interventional study was used to evaluate 20 patients on continuous ambulatory peritoneal dialysis (CAPD) enrolled in a one-month self-controlled study [Trial#: NCT04923295]. Inclusion criteria included being over 18, and having a Peritoneal Dialysis (PD) vintage of at least six months. All participants were classified as having high or average high transport status based on their Peritoneal Equilibrium Test with a D0/D4 > 0.39. and using at least two exchanges with 2.35% dextrose over the previous three months before enrollment. RESULTS Following the treatment, 13 patients had an increase in median D4/D0 from 0.26 [0.17-0.38] to 0.31 [0.23-0.40], while seven patients had a decline from 0.28 [0.17-0.38] to 0.23 [0.14-0.33]. Additionally, nine patients had a decrease in median D/P from 0.88 [0.67-0.92] to 0.81 [0.54-0.85], while 11 patients had an increase from 0.70 [0.6-0.83] to 0.76 [0.63-0.91]. CONCLUSION According to the findings of this study, Dapagliflozin usage in peritoneal dialysis patients did not result in a reduction in glucose absorption across the peritoneal membrane. Additionally, Dapagliflozin was also associated with a small increase in sodium dip, a decrease in peritoneal VEGF, and a decrease in systemic IL-6 levels all of which were not statistically significant. Further large-scale studies are required to corroborate these conclusions.
Collapse
Affiliation(s)
- Zakaria Hamdan
- Internal Medicine Department, An-Najah National University Hospital, Box 7, Nablus, 707, Palestine.
| | | | - Ahmad Enaya
- Internal Medicine Department, An-Najah National University Hospital, Box 7, Nablus, 707, Palestine
| | - Alaa Sarsour
- Kidney and Dialysis Section, An-Najah National University Hospital, Nablus, Palestine
| | - Lubna Kharraz
- Pathology and Medical Laboratory Sciences, An-Najah National University, Nablus, Palestine
| | - Zaher Nazzal
- Department of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Box 7, Nablus, 707, Palestine.
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
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
|
18
|
Krediet RT, Parikova A. Glucose-induced pseudohypoxia and advanced glycosylation end products explain peritoneal damage in long-term peritoneal dialysis. Perit Dial Int 2024; 44:6-15. [PMID: 37723976 DOI: 10.1177/08968608231196033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023] Open
Abstract
Long-term peritoneal dialysis is associated with the development of peritoneal membrane alterations, both in morphology and function. Impaired ultrafiltration (UF) is the most important functional change, and peritoneal fibrosis is the major morphological alteration. Both are caused by the continuous exposure to dialysis solutions that are different from plasma water with regard to the buffer substance and the extremely high-glucose concentrations. Glucose has been incriminated as the major cause of long-term peritoneal membrane changes, but the precise mechanism has not been identified. We argue that glucose causes the membrane alterations by peritoneal pseudohypoxia and by the formation of advanced glycosylation end products (AGEs). After a summary of UF kinetics including the role of glucose transporters (GLUT), and a discussion on morphologic alterations, relationships between function and morphology and a survey of the pathogenesis of UF failure (UFF), it will be argued that impaired UF is partly caused by a reduction in small pore fluid transport as a consequence of AGE-related vasculopathy and - more importantly - in diminished free water transport due to pseudohypoxia, caused by increased peritoneal cellular expression of GLUT-1. The metabolism of intracellular glucose will be reviewed. This occurs in the glycolysis and in the polyol/sorbitol pathway, the latter is activated in case of a large supply. In both pathways the ratio between the reduced and oxidised form of nicotinamide dinucleotide (NADH/NAD+ ratio) will increase, especially because normal compensatory mechanisms may be impaired, and activate expression of hypoxia-inducible factor-1 (HIF-1). The latter gene activates various profibrotic factors and GLUT-1. Besides replacement of glucose as an osmotic agent, medical treatment/prevention is currently limited to tamoxifen and possibly Renin/angiotensis/aldosteron (RAA) inhibitors.
Collapse
Affiliation(s)
- Raymond T Krediet
- Division of Nephrology, Department of Medicine, Amsterdam UMC, University of Amsterdam, The Netherlands
| | - Alena Parikova
- Department of Nephrology, Transplant Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
19
|
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
|
20
|
Chen X, Hocher CF, Shen L, Krämer BK, Hocher B. Reno- and cardioprotective molecular mechanisms of SGLT2 inhibitors beyond glycemic control: from bedside to bench. Am J Physiol Cell Physiol 2023; 325:C661-C681. [PMID: 37519230 DOI: 10.1152/ajpcell.00177.2023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023]
Abstract
Large placebo-controlled clinical trials have shown that sodium-glucose cotransporter-2 inhibitors (SGLT2i) delay the deterioration of renal function and reduce cardiovascular events in a glucose-independent manner, thereby ultimately reducing mortality in patients with chronic kidney disease (CKD) and/or heart failure. These existing clinical data stimulated preclinical studies aiming to understand the observed clinical effects. In animal models, it was shown that the beneficial effect of SGLT2i on the tubuloglomerular feedback (TGF) improves glomerular pressure and reduces tubular workload by improving renal hemodynamics, which appears to be dependent on salt intake. High salt intake might blunt the SGLT2i effects on the TGF. Beyond the salt-dependent effects of SGLT2i on renal hemodynamics, SGLT2i inhibited several key aspects of macrophage-mediated renal inflammation and fibrosis, including inhibiting the differentiation of monocytes to macrophages, promoting the polarization of macrophages from a proinflammatory M1 phenotype to an anti-inflammatory M2 phenotype, and suppressing the activation of inflammasomes and major proinflammatory factors. As macrophages are also important cells mediating atherosclerosis and myocardial remodeling after injury, the inhibitory effects of SGLT2i on macrophage differentiation and inflammatory responses may also play a role in stabilizing atherosclerotic plaques and ameliorating myocardial inflammation and fibrosis. Recent studies suggest that SGLT2i may also act directly on the Na+/H+ exchanger and Late-INa in cardiomyocytes thus reducing Na+ and Ca2+ overload-mediated myocardial damage. In addition, the renal-cardioprotective mechanisms of SGLT2i include systemic effects on the sympathetic nervous system, blood volume, salt excretion, and energy metabolism.
Collapse
Affiliation(s)
- Xin Chen
- Department of Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Carl-Friedrich Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Klinik für Innere Medizin, Bundeswehrkrankenhaus Berlin, Berlin, Germany
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Changsha, China
- IMD Institut für Medizinische Diagnostik Berlin-Potsdam GbR, Berlin, Germany
| |
Collapse
|
21
|
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
|
22
|
Lai JW, Lin HJ, Chou CY. SGLT-2 inhibitors may increase ultrafiltration in incident peritoneal dialysis patients: a case report. BMC Nephrol 2023; 24:106. [PMID: 37087421 PMCID: PMC10122385 DOI: 10.1186/s12882-023-03164-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/11/2023] [Indexed: 04/24/2023] Open
Abstract
BACKGROUND Adequate fluid removal to achieve euvolemic status can be difficult in patients with incident peritoneal dialysis (PD). Limited treatments such as increased high dextrose PD solutions and icodextrin are currently available. We reported four incident PD patients whose' ultrafiltration volume was increased after sodium-glucose cotransporter-2 inhibitors. CASE PRESENTATION The four reported cases were diabetic kidney disease stage 5 (cases 1-3) and IgA nephritis (case 4) patients whostartedt PD because of acute pulmonary edema (case 1 and 3), nausea vomiting (case 2), and hyperkalemia (case 4). They had an ultrafiltration volume of 700-1000 ml per day but hpersistentted peripheral pitting edema or pulmonary edema. Their ultrafiltration volincreased after dapagliflozin 5 mg daily, and the fluid overload symptoms ere improved. No hypotension, or hypoglycemia was found, and the urine was not increased during dapagliflozin treatment. CONCLUSIONS SGLT-2 inhibitors may increase ultrafiltration in incident PD patients. More studies are needed to support the safety of SGLT-2 inhibitors in PD patients.
Collapse
Affiliation(s)
- Jia-Wen Lai
- Division of Nephrology, Asia University Hospital, Wufeng, Taichung, Taiwan
| | - Hsuan-Jen Lin
- Division of Nephrology, Asia University Hospital, Wufeng, Taichung, Taiwan
- Division of Nephrology, China Medical University Hospital, Taichung, Taiwan
| | - Che-Yi Chou
- Division of Nephrology, Asia University Hospital, Wufeng, Taichung, Taiwan.
- Division of Nephrology, China Medical University Hospital, Taichung, Taiwan.
- Department of Post-Baccalaureate Veterinary Medicine, Asia University, NO 222, Fuxin Rd, Wufeng Dist, Taichung 413, Taiwan.
| |
Collapse
|
23
|
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
|
24
|
Liu J, Jin B, Lu J, Feng Y, Li N, Wan C, Zhang QY, Jiang CM. Angiotensin II type 2 receptor prevents extracellular matrix accumulation in human peritoneal mesothelial cell by ameliorating lipid disorder via LOX-1 suppression. Ren Fail 2022; 44:1687-1697. [PMID: 36226438 PMCID: PMC9578471 DOI: 10.1080/0886022x.2022.2133729] [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] [Indexed: 11/23/2022] Open
Abstract
Evidence suggests that intracellular angiotensin II type 1 receptor (AT1) contributes to peritoneal fibrosis (PF) under high glucose (HG)-based dialysates. It is generally believed that AT2 antagonisticly affects AT1 function. The aim of this study was to explore whether AT2 activation is beneficial for attenuating human peritoneal mesothelial cell (HPMC) injury due to HG. We treated a HPMC line with HG to induce extracellular matrix (ECM) formation. AT2 was increased and blocked using CGP42112A and AT2 siRNA. Lipid deposition was detected, signaling molecules associated with lectin-like oxidized lipoprotein receptor-1 (LOX-1) and ECM proteins were evaluated by real-time PCR and western blot. The results showed that HG led to AT2 inhibition in HPMCs, inhibition of AT2 further aggravated the expression of ECM proteins, including α-smooth muscle actin, fibroblast specific protein-1 and collagen I, while AT2 decreased the expression of ECM proteins, even during HG stimulation. Interestingly, there was a parallel change in lipid accumulation and ECM formation when AT2 was increased or depressed. Moreover, AT2-mediated decreased ECM production was associated with reduced lipid accumulation in HPMCs and depended on the downregulation of LOX-1. Further analysis showed that HG increased oxidized low-density lipoprotein (ox-LDL) deposition in HPMCs concomitant with an enhanced expression of ECM components, whereas blocking LOX-1 reversed ox-LDL deposition even in the presence of HG. This effect was also accompanied by the remission of ECM accumulation. Our results suggested that AT2 prevented ECM formation in HG-stimulated HPMCs by ameliorating lipid via LOX‐1 suppression.
Collapse
Affiliation(s)
- Jing Liu
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Bo Jin
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jian Lu
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yuan Feng
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Nan Li
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Cheng Wan
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qing-Yan Zhang
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chun-Ming Jiang
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
25
|
Bergling K, Martus G, Öberg CM. Phloretin Improves Ultrafiltration and Reduces Glucose Absorption during Peritoneal Dialysis in Rats. J Am Soc Nephrol 2022; 33:1857-1863. [PMID: 35985816 PMCID: PMC9528341 DOI: 10.1681/asn.2022040474] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/14/2022] [Accepted: 07/24/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Harmful glucose exposure and absorption remain major limitations of peritoneal dialysis (PD). We previously showed that inhibition of sodium glucose cotransporter 2 did not affect glucose transport during PD in rats. However, more recently, we found that phlorizin, a dual blocker of sodium glucose cotransporters 1 and 2, reduces glucose diffusion in PD. Therefore, either inhibiting sodium glucose cotransporter 1 or blocking facilitative glucose channels by phlorizin metabolite phloretin would reduce glucose transport in PD. METHODS We tested a selective blocker of sodium glucose cotransporter 1, mizagliflozin, as well as phloretin, a nonselective blocker of facilitative glucose channels, in an anesthetized Sprague-Dawley rat model of PD. RESULTS Intraperitoneal phloretin treatment reduced glucose absorption by >30% and resulted in a >50% higher ultrafiltration rate compared with control animals. Sodium removal and sodium clearances were similarly improved, whereas the amount of ultrafiltration per millimole of sodium removed did not differ. Mizagliflozin did not influence glucose transport or osmotic water transport. CONCLUSIONS Taken together, our results and previous results indicate that blockers of facilitative glucose channels may be a promising target for reducing glucose absorption and improving ultrafiltration efficiency in PD.
Collapse
Affiliation(s)
- Karin Bergling
- Division of Nephrology, Department of Clinical Sciences Lund, Skåne University Hospital, Lund University, Lund, Sweden
| | - Giedre Martus
- Division of Nephrology, Department of Clinical Sciences Lund, Skåne University Hospital, Lund University, Lund, Sweden
| | - Carl M. Öberg
- Division of Nephrology, Department of Clinical Sciences Lund, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
26
|
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
|
27
|
Elrakaybi A, Laubner K, Zhou Q, Hug MJ, Seufert J. Cardiovascular protection by SGLT2 inhibitors - Do anti-inflammatory mechanisms play a role? Mol Metab 2022; 64:101549. [PMID: 35863639 PMCID: PMC9352970 DOI: 10.1016/j.molmet.2022.101549] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Metabolic syndrome and related metabolic disturbances represent a state of low-grade inflammation, which accelerates insulin resistance, type 2 diabetes (T2D) and cardiovascular disease (CVD) progression. Among antidiabetic medications, sodium glucose co-transporter (SGLT) 2 inhibitors are the only agents which showed remarkable reductions in heart failure (HF) hospitalizations and major cardiovascular endpoints (MACE) as well as renal endpoints regardless of diabetes status in large randomized clinical outcome trials (RCTs). Although the exact mechanisms underlying these benefits are yet to be established, growing evidence suggests that modulating inflammation by SGLT2 inhibitors may play a key role. SCOPE OF REVIEW In this manuscript, we summarize the current knowledge on anti-inflammatory effects of SGLT2 inhibitors as one of the mechanisms potentially mediating their cardiovascular (CV) benefits. We introduce the different metabolic and systemic actions mediated by these agents which could mitigate inflammation, and further present the signalling pathways potentially responsible for their proposed direct anti-inflammatory effects. We also discuss controversies surrounding some of these mechanisms. MAJOR CONCLUSIONS SGLT2 inhibitors are promising anti-inflammatory agents by acting either indirectly via improving metabolism and reducing stress conditions or via direct modulation of inflammatory signalling pathways. These effects were achieved, to a great extent, in a glucose-independent manner which established their clinical use in HF patients with and without diabetes.
Collapse
Affiliation(s)
- Asmaa Elrakaybi
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Department of Clinical Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Katharina Laubner
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Qian Zhou
- Department of Cardiology and Angiology I, Heart Centre, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Department of Cardiology, University Hospital Basel, 4031 Basel, Switzerland
| | - Martin J Hug
- Pharmacy, Medical Centre - University of Freiburg, 79106 Freiburg, Germany
| | - Jochen Seufert
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.
| |
Collapse
|
28
|
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: 6] [Impact Index Per Article: 2.0] [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
|
29
|
Stepanova N, Snisar L, Burdeyna O. Peritoneal dialysis and peritoneal fibrosis: molecular mechanisms, risk factors and prospects for prevention. UKRAINIAN JOURNAL OF NEPHROLOGY AND DIALYSIS 2022:81-90. [DOI: 10.31450/ukrjnd.4(76).2022.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Peritoneal dialysis (PD) leads to structural and functional changes in the peritoneal membrane, the endpoint of which is peritoneal fibrosis. Peritoneal fibrosis is diagnosed in 50% and 80% of PD patients within 1 and 2 years of treatment initiation, respectively. A key role in the development of peritoneal fibrosis is played by mesothelial-mesenchymal transformation, a complex biological process of transition from mesothelium to mesenchyme. This review summarizes the current knowledge on the changes in peritoneal function and morphology, the molecular mechanisms of peritoneal fibrosis development, and its clinical consequences during PD. Special attention is given to established and potential risk factors for peritoneal fibrosis, and existing prevention strategies are considered.
Collapse
|
30
|
Zarogiannis SG, Schmitt CP. Molecular Mechanisms of Peritoneal Membrane Pathophysiology. Biomolecules 2022; 12:biom12060757. [PMID: 35740882 PMCID: PMC9220859 DOI: 10.3390/biom12060757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 02/01/2023] Open
Abstract
The peritoneal membrane is the largest internal membrane of the human body, having a surface area that approximates the surface area of the skin [...]
Collapse
Affiliation(s)
- Sotirios G. Zarogiannis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece
- Correspondence: ; Tel.: +30 2410 685558
| | - Claus Peter Schmitt
- Pediatric Nephology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, 69210 Heidelberg, Germany;
| |
Collapse
|
31
|
Bartosova M, Zarogiannis SG, Schmitt CP. How peritoneal dialysis transforms the peritoneum and vasculature in children with chronic kidney disease-what can we learn for future treatment? Mol Cell Pediatr 2022; 9:9. [PMID: 35513740 PMCID: PMC9072612 DOI: 10.1186/s40348-022-00141-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/09/2022] [Indexed: 01/04/2023] Open
Abstract
Children with chronic kidney disease (CKD) suffer from inflammation and reactive metabolite-induced stress, which massively accelerates tissue and vascular aging. Peritoneal dialysis (PD) is the preferred dialysis mode in children, but currently used PD fluids contain far supraphysiological glucose concentrations for fluid and toxin removal and glucose degradation products (GDP). While the peritoneal membrane of children with CKD G5 exhibits only minor alterations, PD fluids trigger numerous molecular cascades resulting in major peritoneal membrane inflammation, hypervascularization, and fibrosis, with distinct molecular and morphological patterns depending on the GDP content of the PD fluid used. PD further aggravates systemic vascular disease. The systemic vascular aging process is particularly pronounced when PD fluids with high GDP concentrations are used. GDP induce endothelial junction disintegration, apoptosis, fibrosis, and intima thickening. This review gives an overview on the molecular mechanisms of peritoneal and vascular transformation and strategies to improve peritoneal and vascular health in patients on PD.
Collapse
Affiliation(s)
- Maria Bartosova
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Sotirios G Zarogiannis
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany.,Department of Physiology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|
32
|
Borkum M, Jamal A, Suneet Singh R, Levin A. The rationale for the need to study sodium-glucose co-transport 2 inhibitor usage in peritoneal dialysis patients. ARCH ESP UROL 2022; 43:139-144. [PMID: 35491897 DOI: 10.1177/08968608221096556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The wave of kidney and heart outcome trials, showing multiple potential benefits for sodium-glucose co-transport 2 (SGLT2) inhibitors, have excluded patients with an estimated glomerular filtration rate below 25 ml/min/1.73 m2. However, dialysis patients are at the highest risk of cardiovascular disease and would benefit most from effective cardioprotective therapies. There is emerging evidence from experimental studies and post hoc analyses of randomised clinical trials that SGLT2 inhibitors are well tolerated and may also be effective in preventing cardiovascular and mortality outcomes in patients with severe chronic kidney disease, including patients receiving dialysis. As such, extending the usage of SGLT2 inhibitors to dialysis patients could provide a major advancement in their care. Peritoneal dialysis (PD) patients have an additional unmet need for effective pharmacotherapy to preserve their residual kidney function (RKF), with its associated mortality benefits, and for treatment options that help reduce the risk of transfer to haemodialysis. Experimental data suggest that SGLT2 inhibitors, via various mechanisms, may preserve RKF and protect the peritoneal membrane. There is sound physiological rationale and an urgent clinical need to execute robust randomised control trials to study the use of SGLT2 inhibitors in PD patients to answer important questions of relevance to patients and healthcare systems.
Collapse
Affiliation(s)
- Megan Borkum
- Division of Nephrology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Abeed Jamal
- Division of Nephrology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rajinder Suneet Singh
- Division of Nephrology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Adeera Levin
- Division of Nephrology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
33
|
Krediet RT. Aging of the Peritoneal Dialysis Membrane. Front Physiol 2022; 13:885802. [PMID: 35574465 PMCID: PMC9096116 DOI: 10.3389/fphys.2022.885802] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/11/2022] [Indexed: 12/02/2022] Open
Abstract
Long-term peritoneal dialysis as currently performed, causes structural and functional alterations of the peritoneal dialysis membrane. This decay is brought about by the continuous exposure to commercially available glucose-based dialysis solutions. This review summarizes our knowledge on the peritoneum in the initial phase of PD, during the first 2 years and the alterations in function and morphology in long-term PD patients. The pseudohypoxia hypothesis is discussed and how this glucose-induced condition can be used to explain all peritoneal alterations in long-term PD patients. Special attention is paid to the upregulation of hypoxia inducing factor-1 and the subsequent stimulation of the genes coding for glucose transporter-1 (GLUT-1) and the growth factors transforming growth factor-β (TGFβ), vascular endothelial growth factor (VEGF), plasminogen growth factor activator inhibitor-1 (PAI-1) and connective tissue growth factor (CTGF). It is argued that increased pseudohypoxia-induced expression of GLUT-1 in interstitial fibroblasts is the key factor in a vicious circle that augments ultrafiltration failure. The practical use of the protein transcripts of the upregulated growth factors in peritoneal dialysis effluent is considered. The available and developing options for prevention and treatment are examined. It is concluded that low glucose degradation products/neutral pH, bicarbonate buffered solutions with a combination of various osmotic agents all in low concentration, are currently the best achievable options, while other accompanying measures like the use of RAAS inhibitors and tamoxifen may be valuable. Emerging developments include the addition of alanyl glutamine to the dialysis solution and perhaps the use of nicotinamide mononucleotide, available as nutritional supplement.
Collapse
|
34
|
Jia M, Qiu H, Lin L, Zhang S, Li D, Jin D. Inhibition of PI3K/AKT/mTOR Signalling Pathway Activates Autophagy and Suppresses Peritoneal Fibrosis in the Process of Peritoneal Dialysis. Front Physiol 2022; 13:778479. [PMID: 35309056 PMCID: PMC8931542 DOI: 10.3389/fphys.2022.778479] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/17/2022] [Indexed: 12/21/2022] Open
Abstract
Peritoneal dialysis (PD) is an important part of replacement therapy for kidney failure. However, long-term PD treatment can cause peritoneal fibrosis. Autophagy may be involved in the pathological mechanism of peritoneal fibrosis (PF). Although autophagy is currently known to be involved in course of PF, its specific effects still lack in-depth research. In this experiment, a high-glucose (HG)-induced peritoneal fibrosis rat model was successfully established via intraperitoneal injection of HG peritoneal dialysate, and the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 and the mechanistic target of rapamycin (mTOR) inhibitor rapamycin were used to treat peritoneal fibrosis rats. In addition, in vitro studies of high glucose-induced peritoneal fibrosis were performed using rat peritoneal mesothelial cells (PMCs). In vivo and in vitro experiments showed that LY294002 and rapamycin effectively inhibited the process of PF induced by high glucose. In addition, LY294002 and rapamycin were found to alleviate fibrosis by eliminating intracellular reactive oxygen species (ROS) levels, promoting the expression of the epithelial mesenchymal transdifferentiation proteins zonula occludens-1 (ZO-1) and E-cadherin, and inhibiting the expression of p-PI3K, PI3K, p-mTOR, mTOR, the fibroblast-specific proteins ferroptosis suppressor protein 1 (FSP1), and alpha-smooth muscle actin (α-SMA). Moreover, LY294002 and rapamycin promoted expression of autophagy-related proteins LC3-II/I, p62, and beclin-1. The current data indicated that inhibition of PI3K/AKT/mTOR signalling pathway activated autophagy and suppressed PF in the process of PD. Therefore, intervention in this signalling pathway may become a research goal for the prevention and treatment of PF, which has important clinical significance.
Collapse
|
35
|
Krediet RT. Acquired Decline in Ultrafiltration in Peritoneal Dialysis: The Role of Glucose. J Am Soc Nephrol 2021; 32:2408-2415. [PMID: 34321252 PMCID: PMC8722789 DOI: 10.1681/asn.2021010080] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/26/2021] [Indexed: 02/04/2023] Open
Abstract
Ultrafiltration is essential in peritoneal dialysis (PD) for maintenance of euvolemia, making ultrafiltration insufficiency-preferably called ultrafiltration failure-an important complication. The mechanisms of ultrafiltration and ultrafiltration failure are more complex than generally assumed, especially after long-term treatment. Initially, ultrafiltration failure is mainly explained by a large number of perfused peritoneal microvessels, leading to a rapid decline of the crystalloid osmotic gradient, thereby decreasing aquaporin-mediated free water transport. The contribution of peritoneal interstitial tissue to ultrafiltration failure is limited during the first few years of PD, but becomes more important in long-term PD due to the development of interstitial fibrosis, which mainly consists of myofibroblasts. A dual hypothesis has been developed to explain why the continuous exposure of peritoneal tissues to the extremely high dialysate glucose concentrations causes progressive ultrafiltration decline. First, glucose absorption causes an increase of the intracellular NADH/NAD+ ratio, also called pseudohypoxia. Intracellular hypoxia stimulates myofibroblasts to produce profibrotic and angiogenetic factors, and the glucose transporter GLUT-1. Second, the increased GLUT-1 expression by myofibroblasts increases glucose uptake in these cells, leading to a reduction of the osmotic gradient for ultrafiltration. Reduction of peritoneal glucose exposure to prevent this vicious circle is essential for high-quality, long-term PD.
Collapse
Affiliation(s)
- Raymond T. Krediet
- Division of Nephrology, Department of Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands,Correspondence: Prof. Raymond T. Krediet, Division of Nephrology, Department of Medicine, Amsterdam University Medical Centre, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|