1
|
Zhao H, Zhang HL, Jia L. High glucose dialysate-induced peritoneal fibrosis: Pathophysiology, underlying mechanisms and potential therapeutic strategies. Biomed Pharmacother 2023; 165:115246. [PMID: 37523983 DOI: 10.1016/j.biopha.2023.115246] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023] Open
Abstract
Peritoneal dialysis is an efficient renal replacement therapy for patients with end-stage kidney disease. However, continuous exposure of the peritoneal membrane to dialysate frequently leads to peritoneal fibrosis, which alters the function of the peritoneal membrane and results in withdrawal from peritoneal dialysis in patients. Among others, high glucose dialysate is considered as a predisposing factor for peritoneal fibrosis in patients on peritoneal dialysis. Glucose-induced inflammation, metabolism disturbance, activation of the renin-angiotensin-aldosterone system, angiogenesis and noninflammation-induced reactive oxygen species are implicated in the pathogenesis of high glucose dialysate-induced peritoneal fibrosis. Specifically, high glucose causes chronic inflammation and recurrent peritonitis, which could cause migration and polarization of inflammatory cells, as well as release of cytokines and fibrosis. High glucose also interferes with lipid metabolism and glycolysis by activating the sterol-regulatory element-binding protein-2/cleavage-activating protein pathway and increasing hypoxia inducible factor-1α expression, leading to angiogenesis and peritoneal fibrosis. Activation of the renin-angiotensin-aldosterone system and Ras-mitogen activated protein kinase signaling pathway is another contributing factor in high glucose dialysate-induced fibrosis. Ultimately, activation of the transforming growth factor-β1/Smad pathway is involved in mesothelial-mesenchymal transition or epithelial-mesenchymal transition, which leads to the development of fibrosis. Although possible intervention strategies for peritoneal dialysate-induced fibrosis by targeting the transforming growth factor-β1/Smad pathway have occasionally been proposed, lack of laboratory evidence renders clinical decision-making difficult. We therefore aim to revisit the upstream pathways of transforming growth factor-beta1/Smad and propose potential therapeutic targets for high glucose-induced peritoneal fibrosis.
Collapse
Affiliation(s)
- Hanxue Zhao
- First Clinical Medical College, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Beijing 100053, China
| | - Hong-Liang Zhang
- Department of Life Sciences, National Natural Science Foundation of China, No. 83 Shuangqing Road, Beijing 100085, China.
| | - Linpei Jia
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Beijing 100053, China.
| |
Collapse
|
2
|
Ensan B, Bathaei P, Nassiri M, Khazaei M, Hassanian SM, Abdollahi A, Ghorbani HR, Aliakbarian M, Ferns GA, Avan A. The Therapeutic Potential of Targeting Key Signaling Pathways as a Novel Approach to Ameliorating Post-Surgical Adhesions. Curr Pharm Des 2022; 28:3592-3617. [PMID: 35466868 DOI: 10.2174/1381612828666220422090238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/24/2022] [Accepted: 02/04/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND Peritoneal adhesions (PA) are a common complication of abdominal operations. A growing body of evidence shows that inhibition of inflammation and fibrosis at sites of peritoneal damaging could prevent the development of intra-abdominal adhesions. METHODS A search of PubMed, Medline, CINAHL and Embase databases was performed using the keywords 'postsurgical adhesion', 'post-operative adhesion', 'peritoneal adhesion', 'surgery-induced adhesion' and 'abdominal adhesion'. Studies detailing the use of pharmacological and non-pharmacological agents for peritoneal adhesion prevention were identified, and their bibliographies were thoroughly reviewed to identify further related articles. RESULTS Several signaling pathways, such as tumor necrosis factor-alpha, tissue plasminogen activator, and type 1 plasminogen activator inhibitor, macrophages, fibroblasts, and mesothelial cells play a key part in the development of plasminogen activator. Several therapeutic approaches based on anti-PA drug barriers and traditional herbal medicines have been developed to prevent and treat adhesion formation. In recent years, the most promising method to prevent PA is treatment using biomaterial-based barriers. CONCLUSION In this review, we provide an overview of the pathophysiology of adhesion formation and various agents targeting different pathways, including chemical agents, herbal agents, physical barriers, and clinical trials concerning this matter.
Collapse
Affiliation(s)
- Behzad Ensan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parsa Bathaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Nassiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Abdollahi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Ghorbani
- Orology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Aliakbarian
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Yu F, Chen J, Wang X, Cai Q, Luo J, Wang L, Chen K, He Y. Establishment of a novel mouse peritoneal dialysis-associated peritoneal injury model. Clin Exp Nephrol 2022; 26:649-658. [PMID: 35353282 DOI: 10.1007/s10157-022-02208-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 03/04/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Peritoneal fibrosis induced by various factors during peritoneal dialysis (PD) can eventually lead to ultrafiltration failure and termination of PD treatment. The existing animal models are caused by a single stimulus, and cannot accurately simulate complex pathogenesis of peritoneal injury and fibrosis. We aim to develop an efficient and realistic mouse model of PD-associated peritoneal injury using daily intraperitoneal injection (I.P.) of human peritonitis PD effluent. METHODS Eight-week-old male C57BL/6 mice were classified into six groups: saline control; 2.5% PD fluid; 2.5% PD fluid + lipopolysaccharide (LPS); 4.25% PD fluid; 4.25% PD fluid + LPS; and peritonitis effluent. Mice received daily I.P. for 6 weeks, and were sacrificed to determine peritoneal structural and functional damage, inflammation, and fibrosis. RESULTS Mice in the peritonitis effluent group had low mortality. The submesothelial thickness in the peritonitis effluent group was significantly greater than that in the 2.5% PD fluid group. The peritonitis effluent group had increased expression of fibrosis markers (α-SMA, Collagen I, etc.), neutrophil granulocytes (MPO), and macrophages (CD68, F4/80) in the peritoneum based on immunohistochemical staining; and significantly higher expression of inflammation markers (IL-1β, IL-6, etc.) and fibrosis markers (TGF-β1, α-SMA, etc.) based on real-time qPCR. Modified peritoneal equilibration tests (PET) demonstrated that I.P. of peritonitis effluent reduced peritoneal ultrafiltration. CONCLUSION Our novel animal model of PD-associated peritoneal injury faithfully simulates the clinical pathophysiological process. This animal model may be useful for study of the pathogenesis of PD-associated peritoneal injury and identification of novel treatments.
Collapse
Affiliation(s)
- Fang Yu
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, No. 10 Changjiang Road, Chongqing, 400042, China
| | - Jia Chen
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, No. 10 Changjiang Road, Chongqing, 400042, China
| | - Xiaoyue Wang
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, No. 10 Changjiang Road, Chongqing, 400042, China
| | - Qingli Cai
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, No. 10 Changjiang Road, Chongqing, 400042, China
| | - Jia Luo
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, No. 10 Changjiang Road, Chongqing, 400042, China
| | - Liming Wang
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, No. 10 Changjiang Road, Chongqing, 400042, China
| | - Kehong Chen
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, No. 10 Changjiang Road, Chongqing, 400042, China.
| | - Yani He
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, No. 10 Changjiang Road, Chongqing, 400042, China.
| |
Collapse
|
4
|
Liu J, Feng Y, Sun C, Zhu W, Zhang QY, Jin B, Shao QY, Xia YY, Xu PF, Zhang M, Jiang CM. Valsartan ameliorates high glucose-induced peritoneal fibrosis by blocking mTORC1 signaling. Exp Biol Med (Maywood) 2020; 245:983-993. [PMID: 32408765 DOI: 10.1177/1535370220919364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
IMPACT STATEMENT Our study provided new insight into the mechanism underlying the preservation of the peritoneum by valsartan. The results demonstrated that the mice receiving chronic high glucose (HG) peritoneal dialysis solution infusion showed a typical feature of peritoneal fibrosis (PF), as well as higher expression of α-smooth muscle actin (α-SMA) and collagen I. In vitro, HG increased the protein expression of α-SMA and collagen I in a dose-dependent manner, while valsartan significantly ameliorated these pathological changes. Interestingly, there was a parallel decrease in the activity of mammalian target of rapamycin complex 1 (mTORC1) and the protein expression levels of α-SMA and collagen I upon treatment with valsartan in vivo and in vitro. Moreover, the mTOR agonist MHY1485 reversed the downregulation of α-SMA and collagen I in vitro, even in the presence of valsartan. Altogether, our findings reported for the first time that valsartan exerts a protective effect against HG-induced PF by inhibiting the activity of the mTORC1 pathway.
Collapse
Affiliation(s)
- Jing Liu
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing City, Jiangsu Province 210008, China
| | - Yuan Feng
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing City, Jiangsu Province 210008, China
| | - Cheng Sun
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing City, Jiangsu Province 210008, China
| | - Wei Zhu
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing City, Jiangsu Province 210008, China
| | - Qing-Yan Zhang
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing City, Jiangsu Province 210008, China
| | - Bo Jin
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing City, Jiangsu Province 210008, China
| | - Qiu-Yuan Shao
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing City, Jiangsu Province 210008, China
| | - Yang-Yang Xia
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing City, Jiangsu Province 210008, China
| | - Peng-Fei Xu
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing City, Jiangsu Province 210008, China
| | - Miao Zhang
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing City, Jiangsu Province 210008, China
| | - Chun-Ming Jiang
- Institute of Nephrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing City, Jiangsu Province 210008, China
| |
Collapse
|
5
|
Abstract
Peritoneal dialysis (PD) is a successfully used method for renal replacement therapy. However, long-term PD may be associated with peritoneal fibrosis and ultrafiltration failure. The key factors linked to their appearance are repeated episodes of inflammation associated with peritonitis and long-term exposure to bioincompatible PD fluids. Different strategies have been proposed to preserve the peritoneal membrane. This article reviews the functional and structural alterations related to PD and strategies whereby we may prevent them to preserve the peritoneal membrane. The use of new, more biocompatible, PD solutions is promising, although further morphologic studies in patients using these solutions are needed. Blockade of the renin-angiotensin-aldosterone system appears to be efficacious and strongly should be considered. Other agents have been proven in experimental studies, but most of them have not yet been tested appropriately in human beings.
Collapse
Affiliation(s)
- M Auxiliadora Bajo
- Home Dialysis Unit, Nephrology Department, La Paz University Hospital, Madrid, Spain.
| | - Gloria Del Peso
- University Autónoma of Madrid, Hospital La Paz Institute for Health Research, Spanish Renal Research Network, Reina Sofia Institute for Nephrology Research, Madrid, Spain
| | - Isaac Teitelbaum
- Home Dialysis Program, University of Colorado Hospital, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
6
|
Borazan A, Camsari T, Cavdar Z, Sarioglu S, Yilmaz O, Oktay G, Sifil A, Celik A, Cavdar C, Aysal A, Kolatan E. The Effects of Darbepoetin on Peritoneal Fibrosis Induced by Chemical Peritonitis and on Peritoneal Tissue Mmp-2 and Timp-2 Levels in Rats. EUR J INFLAMM 2017. [DOI: 10.1177/1721727x0900700205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
The aim of this study is to assess the influence of darbepoetin on the development of peritoneal fibrosis in rats induced by Chlorhexidine gluconate (0.1%) and ethanol (15%) and to determine the effect on peritoneal tissue levels of MMP–2 and TIMP–2, possible important factors in progression of peritoneal fibrosis. Twenty-four female Wistar albino rats were divided into three groups. The first group (CH group) received 3 ml/200g daily intraperitoneal injections of Chlorhexidine gluconate (0.1%) and ethanol (15%) dissolved in saline to induce chemical peritonitis; group 2 (ESA group) received 3 ml/200g daily injections of Chlorhexidine gluconate (0.1%) and ethanol (15%) dissolved in saline and also darbepoetin 12.5 microgr/ per kilogram/ day subcutaneously on the first and seventh days; group 3 (Control group) received intraperitoneal 0.9% saline (3 ml/200g/d) through the right lower quadrant by 21 gauge needle. The study duration was fourteen days. On the fifteenth day rats were sacrificed, parietal peritoneum samples were obtained from the left anterior abdominal wall. Pathological samples were examined using Hematoxyline & Eosin (HE) stains. The thickness, vasculpathy, and inflammation were determined by light microscopy. MMP-2 and TIMP-2 were studied immunohistochemically by monoclonal antibody staining. The activity of MMP-2 on peritoneal tissue was studied by gelatin zymography and TIMP–2 protein level was analysed by ELISA, biochemically. The decrease in thickness of parietal peritoneum in group ESA was statistically significant when compared to CH group (p<0.05). Inflammation scores, and vascularization score surfaces were not statistically different between these groups (p>0.05). Immunohistochemically, darbepoetin was shown to decrease MMP-2 expression on parietal peritoneum in CH group (p<0.05), but had no effect on TIMP-2 (p>0.05). Biochemically the ratio of active MMP–2 to proMMP–2 was more significantly increased in the ESA group than in the CH group (p<0.001), however, TIMP- 2 levels in both groups were decreased compared to the control group (p<0.05). Darbepoetin histopathologically reduced peritoneal fibrosis induced by Chlorhexidine gluconate. We can suggest that Darbepoetin does not cause peritoneal fibrosis and may prevent peritoneal fibrosis in rats possibly related to an effect on MMP-2 expression. Further research regarding the utility and dosage should be considered.
Collapse
Affiliation(s)
- A. Borazan
- Department of Nephrology, Faculty of Medicine, Mustafa Kemal University, Hatay
| | - T. Camsari
- Department of Nephrology, Research Center, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Z. Cavdar
- Department of Biochemistry, Research Center, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - S. Sarioglu
- Department of Pathology, Research Center, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - O. Yilmaz
- Department of Animal Research Center, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - G. Oktay
- Department of Biochemistry, Research Center, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - A. Sifil
- Department of Nephrology, Research Center, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - A. Celik
- Department of Nephrology, Research Center, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - C. Cavdar
- Department of Nephrology, Research Center, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - A. Aysal
- Department of Pathology, Research Center, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - E. Kolatan
- Department of Animal Research Center, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW The purpose of this study was to summarize recent findings about cardiovascular benefits and safety of aldosterone blockade in patients with end-stage renal disease (ESRD). RECENT FINDINGS It is now well recognized that aldosterone's deleterious cardiovascular impact is not limited to its pressor effect arising from an increase in sodium reabsorption in the kidneys. Aldosterone has also been shown to increase blood pressure by a direct activation of the sympathetic nervous system, to cause endothelial and vascular smooth muscle cell dysfunction, myocardial remodeling and fibrosis, and to have pro-arrhythmogenic actions in the heart. These unconventional extra-renal effects of aldosterone make its blockade feasible and potentially beneficial for patients with ESRD. Accumulating data support the idea that aldosterone antagonism leads to a better blood pressure control, reduction in left ventricular (LV) mass, improved LV function, and reduced all-cause and cardiovascular mortality in ESRD patients. Reassuringly, rates of major adverse events, especially, significant hyperkalemia-the most feared adverse consequence-were low with careful patient selection and monitoring.
Collapse
Affiliation(s)
- Radmila Lyubarova
- Division of Cardiology, Department of Medicine, Albany Medical College, Albany, NY, USA
| | - Elvira O Gosmanova
- Nephrology Section, Stratton VA Medical Center, 113 Holland Ave, Albany, NY, 12208, USA.
- Division of Nephrology and Hypertension, Department of Medicine, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
8
|
Morinelli TA, Luttrell LM, Strungs EG, Ullian ME. Angiotensin II receptors and peritoneal dialysis-induced peritoneal fibrosis. Int J Biochem Cell Biol 2016; 77:240-50. [PMID: 27167177 PMCID: PMC5038354 DOI: 10.1016/j.biocel.2016.04.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 12/22/2022]
Abstract
The vasoactive hormone angiotensin II initiates its major hemodynamic effects through interaction with AT1 receptors, a member of the class of G protein-coupled receptors. Acting through its AT1R, angiotensin II regulates blood pressure and renal salt and water balance. Recent evidence points to additional pathological influences of activation of AT1R, in particular inflammation, fibrosis and atherosclerosis. The transcription factor nuclear factor κB, a key mediator in inflammation and atherosclerosis, can be activated by angiotensin II through a mechanism that may involve arrestin-dependent AT1 receptor internalization. Peritoneal dialysis is a therapeutic modality for treating patients with end-stage kidney disease. The effectiveness of peritoneal dialysis at removing waste from the circulation is compromised over time as a consequence of peritoneal dialysis-induced peritoneal fibrosis. The non-physiological dialysis solution used in peritoneal dialysis, i.e. highly concentrated, hyperosmotic glucose, acidic pH as well as large volumes infused into the peritoneal cavity, contributes to the development of fibrosis. Numerous trials have been conducted altering certain components of the peritoneal dialysis fluid in hopes of preventing or delaying the fibrotic response with limited success. We hypothesize that structural activation of AT1R by hyperosmotic peritoneal dialysis fluid activates the internalization process and subsequent signaling through the transcription factor nuclear factor κB, resulting in the generation of pro-fibrotic/pro-inflammatory mediators producing peritoneal fibrosis.
Collapse
Affiliation(s)
- Thomas A Morinelli
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, United States.
| | - Louis M Luttrell
- Division of Endocrinology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, United States; Research Service of the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, United States
| | - Erik G Strungs
- Division of Endocrinology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Michael E Ullian
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, United States; Research Service of the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, United States
| |
Collapse
|
9
|
Fülöp T, Zsom L, Rodríguez B, Afshan S, Davidson JV, Szarvas T, Dixit MP, Tapolyai MB, Rosivall L. Clinical Utility of Potassium-Sparing Diuretics to Maintain Normal Serum Potassium in Peritoneal Dialysis Patients. Perit Dial Int 2016; 37:63-69. [PMID: 27282853 DOI: 10.3747/pdi.2016.00022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 04/01/2016] [Indexed: 11/15/2022] Open
Abstract
♦ BACKGROUND: Hypokalemia is a vexing problem in end-stage renal disease patients on peritoneal dialysis (PD), and oral potassium supplements (OPS) have limited palatability. Potassium-sparing diuretics (KSD) (spironolactone, amiloride) may be effective in these patients. ♦ METHODS: We performed a cross-sectional review of 75 current or past (vintage > 6 months) PD patients with regard to serum potassium (K+), OPS, and KSD utilization. We reviewed charts for multiple clinical and laboratory variables, including dialysis adequacy, residual renal function, nutritional status and co-existing medical therapy. ♦ RESULTS: The cohort was middle-aged with a mean age of 49.2 years (standard deviation [SD] = 14.7) and overweight with a body mass index of 29.5 (6.7) kg/m2. Of all the participants, 57.3% were female, 73.3% African-American, and 48% diabetic with an overall PD vintage of 28.2 (24.3) months at the time of enrollment. Weekly Kt/V was 2.12 (0.43), creatinine clearance was 73.5 (33.6) L/week/1.73 m2 with total daily exchange volume of 10.8 (2.7) L. Residual urine output (RUO) measured at 440 (494) mL (anuric 30.6%). Three-month averaged serum K+ measured at 4 (0.5) mmol/L with 36% of the participants receiving K+ supplements (median: 20 [0;20] mmol/day) and 41.3% KSD (spironolactone dose: 25 - 200 mg/day; amiloride dose: 5 - 10 mg/day). Serum K+ correlated positively with weekly Kt/V (r = 0.239; p = 0.039), PD vintage (r = 0.272; p = 0.018) but not with PD modality, daily exchange volume, RUO, or KSD use. However, KSD use was associated with decreased use of OPS (r = -0.646; p < 0.0001). ♦ CONCLUSIONS: Potassium-sparing diuretics were effective in this cohort of PD patients and decreased the need for OPS utilization.
Collapse
Affiliation(s)
- Tibor Fülöp
- Department of Medicine, Division of Nephrology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Lajos Zsom
- Department of Surgery, Division of Transplantation, University of Debrecen, Debrecen, Hungary
| | - Betzaida Rodríguez
- Department of Medicine, Division of Hospital Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sabahat Afshan
- Department of Pediatrics, Division of Pediatric Nephrology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jamie V Davidson
- Department of Medicine, Division of Nephrology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Tibor Szarvas
- Department of Mathematics, Louisiana State University Shreveport, LA, USA
| | - Mehul P Dixit
- Department of Pediatrics, Division of Pediatric Nephrology, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - László Rosivall
- Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
10
|
Guo T, Lok KY, Yu C, Li Z. Lung fibrosis: drug screening and disease biomarker identification with a lung slice culture model and subtracted cDNA Library. Altern Lab Anim 2016; 42:235-43. [PMID: 25290944 DOI: 10.1177/026119291404200405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pulmonary fibrosis is a progressive and irreversible disorder with no appropriate cure. A practical and effective experimental model that recapitulates the disease will greatly benefit the research community and, ultimately, patients. In this study, we tested the lung slice culture (LSC) system for its potential use in drug screening and disease biomarker identification. Fibrosis was induced by treating rat lung slices with 1ng/ml TGF-β1 and 2.5μM CdCl2, quantified by measuring the content of hydroxyproline, and confirmed by detecting the expression of collagen type III alpha 1 (Col3α1) and connective tissue growth factor (CTGF) genes. The anti-fibrotic effects of pirfenidone, spironolactone and eplerenone were assessed by their capability to reduce hydroxyproline content. A subtractive hybridisation technique was used to create two cDNA libraries (subtracted and unsubtracted) from lung slices. The housekeeping gene glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was employed to assess the subtraction efficiency of the subtracted cDNA library. Clones from the two libraries were sequenced and the genes were identified by performing a BLAST search on the NCBI GenBank database. Furthermore, the relevance of the genes to fibrosis formation was verified. The results presented here show that fibrosis was effectively induced in cultured lung slices, which exhibited significantly elevated levels of hydroxyproline and Col3α1/CTGF gene expression. Several inhibitors have demonstrated their anti-fibrotic effects by significantly reducing hydroxyproline content. The subtracted cDNA library, which was enriched for differentially expressed genes, was used to successfully identify genes associated with fibrosis. Collectively, the results indicate that our LSC system is an effective model for the screening of drug candidates and for disease biomarker identification.
Collapse
Affiliation(s)
- Tong Guo
- Goodman Institute of Investment Management, John Molson School of Business, Concordia University, Montreal, Quebec, Canada
| | | | | | - Zhuo Li
- Bio S&T, Montreal, Quebec, Canada
| |
Collapse
|
11
|
Yu M, Shin HS, Lee HK, Ryu DR, Kim SJ, Choi KB, Kang DH. Effect of aldosterone on epithelial-to-mesenchymal transition of human peritoneal mesothelial cells. Kidney Res Clin Pract 2015; 34:83-92. [PMID: 26484027 PMCID: PMC4570652 DOI: 10.1016/j.krcp.2015.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/07/2015] [Accepted: 03/27/2015] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Peritoneal fibrosis is one of the major causes of technical failure in patients on peritoneal dialysis. Epithelial-to-mesenchymal transition (EMT) of the peritoneum is an early and reversible mechanism of peritoneal fibrosis. Human peritoneal mesothelial cells (HPMCs) have their own renin-angiotensin-aldosterone system (RAAS), however, it has not been investigated whether aldosterone, an end-product of the RAAS, induces EMT in HPMCs, and which mechanisms are responsible for aldosterone-induced EMT. METHODS EMT of HPMCs was evaluated by comparing the expression of epithelial cell marker, E-cadherin, and mesenchymal cell marker, α-smooth muscle actin after stimulation with aldosterone (1-100nM) or spironolactone. Activation of extracellular signal-regulated kinase (ERK)1/2 and p38 mitogen-activated protein kinase (MAPK) and generation of reactive oxygen species (ROS) were assessed by western blotting and 2',7'-dichlorofluororescein diacetate staining, respectively. The effects of MAPK inhibitors or antioxidants (N-acetyl cysteine, apocynin, and rotenone) on aldosterone-induced EMT were evaluated. RESULTS Aldosterone induced EMT in cultured HPMCs, and spironolactone blocked aldosterone-induced EMT. Aldosterone induced activation of both ERK1/2 and p38 MAPK from 1 hour. Either PD98059, an inhibitor of ERK1/2, or SB20358, an inhibitor of p38 MAPK, attenuated aldosterone-induced EMT. Aldosterone induced ROS in HPMCs from 5 minutes, and antioxidant treatment ameliorated aldosterone-induced EMT. N-acetyl cysteine and apocynin alleviated activation of ERK and p38 MAPK. CONCLUSION Aldosterone induced EMT in HPMCs by acting through the mineralocorticoid receptor. Aldosterone-induced generation of ROS followed by activation of ERK, and p38 MAPK served as one of the mechanisms of aldosterone-induced EMT of HPMCs.
Collapse
Affiliation(s)
- Mina Yu
- Department of Internal Medicine, Seonam Hospital, Seoul, Korea
| | - Hyun-Soo Shin
- Department of Internal Medicine, Ewha Womans University School of Medicine, Ewha Medical Research Center, Seoul, Korea
| | - Hyeon Kook Lee
- Department of Surgery, Ewha Womans University School of Medicine, Seoul, Korea
| | - Dong-Ryeol Ryu
- Department of Internal Medicine, Ewha Womans University School of Medicine, Ewha Medical Research Center, Seoul, Korea
| | - Seung-Jung Kim
- Department of Internal Medicine, Ewha Womans University School of Medicine, Ewha Medical Research Center, Seoul, Korea
| | - Kyu-Bok Choi
- Department of Internal Medicine, Ewha Womans University School of Medicine, Ewha Medical Research Center, Seoul, Korea
| | - Duk-Hee Kang
- Department of Internal Medicine, Ewha Womans University School of Medicine, Ewha Medical Research Center, Seoul, Korea
| |
Collapse
|
12
|
Intestinal organoids: a model of intestinal fibrosis for evaluating anti-fibrotic drugs. Exp Mol Pathol 2015; 98:346-51. [PMID: 25828392 DOI: 10.1016/j.yexmp.2015.03.033] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 03/27/2015] [Indexed: 01/16/2023]
Abstract
BACKGROUND & AIMS Intestinal fibrosis is a critical complication of Crohn's disease (CD). Current in vitro models of intestinal fibrosis cannot model the complex intestinal architecture, while in vivo rodent models do not fully recapitulate human disease and have limited utility for large-scale screening. Here, we exploit recent advances in stem cell derived human intestinal organoids (HIOs) as a new human model of fibrosis in CD. METHODS Human pluripotent stem cells were differentiated into HIOs. We identified myofibroblasts, the key effector cells of fibrosis, by immunofluorescence staining for alpha-smooth muscle actin (αSMA), vimentin, and desmin. We examined the fibrogenic response of HIOs by treatment with transforming growth factor beta (TGFβ) in the presence or absence of the anti-fibrotic drug spironolactone. Fibrotic response was assayed by expression of fibrogenic genes (COL1A1 (collagen, type I, alpha 1), ACTA2 (alpha smooth muscle actin), FN1 (fibronectin 1), MYLK (myosin light chain kinase), and MKL1 (megakaryoblastic leukemia (translocation) 1)) and proteins (αSMA). RESULTS Immunofluorescent staining of organoids identified a population of myofibroblasts within the HIO mesenchyme. TGFβ stimulation of HIOs produced a dose-dependent pro-fibrotic response. Spironolactone treatment blocked the fibrogenic response of HIOs to TGFβ. CONCLUSIONS HIOs contain myofibroblasts and respond to a pro-fibrotic stimulus in a manner that is consistent with isolated human myofibroblasts. HIOs are a promising model system that might bridge the gap between current in vitro and in vivo models of intestinal fibrosis in IBD.
Collapse
|
13
|
Wang J, Liu S, Li H, Sun J, Zhang S, Xu X, Liu Y, Wang Y, Miao L. A review of rodent models of peritoneal dialysis and its complications. Int Urol Nephrol 2014; 47:209-15. [PMID: 25425436 DOI: 10.1007/s11255-014-0829-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 08/26/2014] [Indexed: 12/14/2022]
Abstract
This article reviews the available rodent models of peritoneal dialysis (PD) that have been developed over the past 20 years and the complications associated with their use. Although there are several methods used in different studies, the focus of this article is not to review or provide detailed summaries of these methods. Rather, this article reviews the most common methods of establishing a dialysis model in rodents, the assays used to observe function of the peritoneum in dialysis, and how these models are adapted to study peritonitis and peritoneal fibrosis. We compared the advantages and disadvantages of different methods, which should be helpful in studies of PD and may provide valuable data for further clinical studies.
Collapse
Affiliation(s)
- Ji Wang
- Department of Nephrology, Second Hospital of Jilin University, Ziqiang Street 218, Nanguan District, Changchun, China
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Zhang L, Hao JB, Ren LS, Ding JL, Hao LR. The aldosterone receptor antagonist spironolactone prevents peritoneal inflammation and fibrosis. J Transl Med 2014; 94:839-50. [PMID: 24862968 DOI: 10.1038/labinvest.2014.69] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 03/31/2014] [Accepted: 04/21/2014] [Indexed: 11/09/2022] Open
Abstract
Peritoneal fibrosis is a complication of patients with long-term continuous ambulatory peritoneal dialysis (CAPD). Reports have indicated that angiotensin (Ang) II may correlate with the development of peritoneal fibrosis. However, it is unknown whether aldosterone also has a role in the development of peritoneal inflammation and fibrosis. The aim of the present study was to clarify the role of aldosterone in peritoneal inflammation and fibrosis. A rat model of peritoneal inflammation and fibrosis was established by daily intraperitoneal injection of dialysates and lipopolysaccharide in a 4-day interval over a period of 7 days. The animals were randomly assigned to five groups as follows: control (C); peritoneal dialysis (PD); peritoneal dialysis-spironolactone (PD-S); peritoneal dialysis-cilazapril (PD-C); and peritoneal dialysis-spironolactone-cilazapril (PD-SC). After 30 days, the TGF-β1 concentration in dialysates from all treatment groups was determined by ELISA. The histopathology of the parietal peritoneum was examined, and the expression of MCP-1, c-Jun, fibronectin (FN) and TGF-β1 in the abdominal membrane was determined by immunohistochemistry. Mineralocorticoid receptor (MR), 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2) and CYP11B2 (aldosterone synthase) were analyzed by real time-PCR. Collagen deposition was significantly higher in PD compared with the other groups. The expression of MR, 11β-HSD2 and CYP11B2 was significantly higher in PD compared with the other groups. Spironolactone and/or cilazapril treatment partially ablated the increase in monocyte chemoattractant protein (MCP)-1, p-c-Jun, transforming growth factor (TGF)-β1, FN, MR, 11β-HSD2 and CYP11B2. Furthermore, treatment with spironolactone and/or cilazapril also reduced the infiltration of CD-4- and ED-1-positive cells in rat peritoneal tissues after peritoneal fibrosis. Exogenous aldosterone may have a key role in the development of peritoneal inflammation and fibrosis. Spironolactone decreased peritoneal inflammation and fibrosis, which was associated with reduced secretion from peritoneal macrophages, inactivation of the c-Jun N-terminal kinase (JNK) pathway and subsequent downregulation of the expression of TGF-β1.
Collapse
Affiliation(s)
- Lei Zhang
- The Second Ward of the Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jian-Bing Hao
- The Second Ward of the Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lian-Sheng Ren
- The Second Ward of the Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiu-Li Ding
- Hospital of Hei Long Jiang Province, Harbin, China
| | - Li-Rong Hao
- The Second Ward of the Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
15
|
Vazquez-Rangel A, Soto V, Escalona M, Toledo RG, Castillo EA, Polanco Flores NA, Falcon-Chavez I, Madero M. Spironolactone to Prevent Peritoneal Fibrosis in Peritoneal Dialysis Patients: A Randomized Controlled Trial. Am J Kidney Dis 2014; 63:1072-4. [DOI: 10.1053/j.ajkd.2014.01.426] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 01/10/2014] [Indexed: 11/11/2022]
|
16
|
Alonso JDM, Alves ALG, Watanabe MJ, Rodrigues CA, Hussni CA. Peritoneal response to abdominal surgery: the role of equine abdominal adhesions and current prophylactic strategies. Vet Med Int 2014; 2014:279730. [PMID: 24587939 PMCID: PMC3918701 DOI: 10.1155/2014/279730] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/22/2013] [Indexed: 12/27/2022] Open
Abstract
Intra-abdominal adhesions constitute a significant clinical and surgical problem that can lead to complications such as pain and bowel occlusion or subocclusion. These adhesions are frustrating and potentially fatal, representing a major postoperative complication in abdominal surgery. It is estimated that 32% of horses undergoing laparotomy will present clinical symptoms due to adhesions, but the true prevalence is not known because a large proportion of animals with postoperative recurrent colics are medically treated or submitted to euthanasia without necropsy. Adhesions are highly cellular, vascularized, dynamic structures that are influenced by complex signaling mechanisms. Understanding their pathogenesis could assist in applying better therapeutic strategies and in developing more effective antiadhesion products. Currently, there are no definitive strategies that prevent adhesion formation, and it is difficult to interpret the results of existing studies due to nonstandardization of an induction model and evaluation of their severity. The best clinical results have been obtained from using minimally traumatic surgical techniques, anti-inflammatory agents, antimicrobials, anticoagulants, and mechanical separation of serosal surfaces by viscous intraperitoneal solutions or physical barriers. This paper aims to review adhesion formation pathogenesis, guide the understanding of major products and drugs used to inhibit adhesion formation, and address their effectiveness in the equine species.
Collapse
Affiliation(s)
- Juliana de Moura Alonso
- School of Veterinary Medicine and Animal Science, UNESP, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Ana Liz Garcia Alves
- School of Veterinary Medicine and Animal Science, UNESP, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Marcos Jun Watanabe
- School of Veterinary Medicine and Animal Science, UNESP, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Celso Antonio Rodrigues
- School of Veterinary Medicine and Animal Science, UNESP, University Estadual Paulista, Botucatu, São Paulo, Brazil
| | - Carlos Alberto Hussni
- School of Veterinary Medicine and Animal Science, UNESP, University Estadual Paulista, Botucatu, São Paulo, Brazil
| |
Collapse
|
17
|
Dinarvand P, Farhadian S, Seyedjafari E, Shafiee A, Jalali A, Sanaei-rad P, Dinarvand B, Soleimani M. Novel approach to reduce postsurgical adhesions to a minimum: Administration of losartan plus atorvastatin intraperitoneally. J Surg Res 2013; 181:91-8. [DOI: 10.1016/j.jss.2012.05.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 05/02/2012] [Accepted: 05/09/2012] [Indexed: 12/08/2022]
|
18
|
Johnson LA, Govani SM, Joyce JC, Waljee AK, Gillespie BW, Higgins PDR. Spironolactone and colitis: increased mortality in rodents and in humans. Inflamm Bowel Dis 2012; 18:1315-24. [PMID: 22081497 PMCID: PMC3288762 DOI: 10.1002/ibd.21929] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 09/28/2011] [Indexed: 12/30/2022]
Abstract
BACKGROUND Crohn's disease causes intestinal inflammation leading to intestinal fibrosis. Spironolactone is an antifibrotic medication commonly used in heart failure to reduce mortality. We examined whether spironolactone is antifibrotic in the context of intestinal inflammation. METHODS In vitro, spironolactone repressed fibrogenesis in transforming growth factor beta (TGF-β)-stimulated human colonic myofibroblasts. However, spironolactone therapy significantly increased mortality in two rodent models of inflammation-induced intestinal fibrosis, suggesting spironolactone could be harmful during intestinal inflammation. Since inflammatory bowel disease (IBD) patients rarely receive spironolactone therapy, we examined whether spironolactone use was associated with mortality in a common cause of inflammatory colitis, Clostridium difficile infection (CDI). RESULTS Spironolactone use during CDI infection was associated with increased mortality in a retrospective cohort of 4008 inpatients (15.9% vs. 9.1%, n = 390 deaths, P < 0.0001). In patients without liver disease, the adjusted odds ratio (OR) for inpatient mortality associated with 80 mg spironolactone was 1.99 (95% confidence interval [CI]: 1.51-2.63) In contrast to the main effect of spironolactone mortality, multivariate modeling revealed a protective interaction between liver disease and spironolactone dose. The adjusted OR for mortality after CDI was 1.96 (95% CI: 1.50-2.55) for patients without liver disease on spironolactone vs. 1.28 (95% CI: 0.82-2.00) for patients with liver disease on spironolactone when compared to a reference group without liver disease or spironolactone use. CONCLUSIONS We propose that discontinuation of spironolactone in patients without liver disease during CDI could reduce hospital mortality by 2-fold, potentially reducing mortality from CDI by 35,000 patients annually across Europe and the U.S.
Collapse
Affiliation(s)
- Laura A. Johnson
- University of Michigan, Department of Internal Medicine, Ann Arbor, MI
| | - Shail M. Govani
- University of Michigan, Department of Internal Medicine, Ann Arbor, MI
| | - Joel C. Joyce
- Medical College of Wisconsin, Department of Dermatology, Milwaukee, WI
| | - Akbar K. Waljee
- University of Michigan, Department of Internal Medicine, Ann Arbor, MI
| | - Brenda W. Gillespie
- University of Michigan, Department of Biostatistics, School of Public Health, Ann Arbor, MI
| | | |
Collapse
|
19
|
Kim DK, Lee SM, Son YK, Kim SE, Kim KH, An WS. Factors Influencing Survival According to Elapsed Time in Peritoneal Dialysis Patients. Ren Fail 2012; 34:559-65. [DOI: 10.3109/0886022x.2012.664761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
20
|
Kato H, Mizuno T, Mizuno M, Sawai A, Suzuki Y, Kinashi H, Nagura F, Maruyama S, Noda Y, Yamada K, Matsuo S, Ito Y. Atrial natriuretic peptide ameliorates peritoneal fibrosis in rat peritonitis model. Nephrol Dial Transplant 2011; 27:526-36. [DOI: 10.1093/ndt/gfr302] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
21
|
Yeniçerioglu Y, Üzelce Ö, Akar H, Kolatan E, Yilmaz O, Yenisey Ç, Sarioglu S, Meteoglu I. Effects of atorvastatin on development of peritoneal fibrosis in rats on peritoneal dialysis. Ren Fail 2010; 32:1095-102. [DOI: 10.3109/0886022x.2010.508859] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
22
|
No need for an "expiry date" in chronic peritoneal dialysis to prevent encapsulating peritoneal sclerosis. Int Urol Nephrol 2010; 41:903-7. [PMID: 19856126 DOI: 10.1007/s11255-009-9661-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
23
|
Xie JY, Chen N, Ren H, Wang WM. Angiotensin II-mediated activation of fibrotic pathways through ERK1/2 in rat peritoneal mesothelial cells. Ren Fail 2010; 32:871-9. [DOI: 10.3109/0886022x.2010.494807] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
24
|
The renin-angiotensin-aldosterone system in peritoneal dialysis: is what is good for the kidney also good for the peritoneum? Kidney Int 2010; 78:23-8. [PMID: 20336052 DOI: 10.1038/ki.2010.90] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Morphological changes of the peritoneal membrane that occur over time among patients on peritoneal dialysis include fibrosis and neoangiogenesis. While the pathophysiologic mechanisms underlying these changes are not fully understood, the activation of the renin-angiotensin-aldosterone system (RAAS) may have an important role. Components of the RAAS are constitutively expressed within peritoneal mesothelial cells, and are upregulated in the presence of acute inflammation and chronic exposure to peritoneal dialysate. The high glucose concentration, low pH, and the presence of glucose degradation products in peritoneal dialysis solutions have all been implicated in modulation of peritoneal RAAS. Furthermore, activation of the RAAS, as well as the downstream production of transforming growth factor-beta, contributes to epithelial-to-mesenchymal transformation of mesothelial cells, resulting in progressive fibrosis of the peritoneal membrane. This process also leads to increased vascular endothelial growth factor production, which promotes peritoneal neoangiogenesis. Functionally, these changes translate into reduced ultrafiltration capacity of the peritoneal membrane, which is an important cause of technique failure among patients on long-term peritoneal dialysis. This brief review will describe our current state of knowledge about the role of peritoneal RAAS in peritoneal membrane damage and potential strategies to protect the membrane.
Collapse
|
25
|
Ucar E, Borazan A, Semerci E, Binici DN, Yaldiz M, Aysal A, Altug E, Kuvandik C, Huzmeli C, Yetim T, Canda S. The Effects of Interferon α2b on Chemically-Induced Peritoneal Fibrosis and on Peritoneal Tissue MMP-2 and TIMP-2 Levels in Rats. J Int Med Res 2010; 38:187-94. [DOI: 10.1177/147323001003800121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
This study investigated the effect of interferon α2b on chlorhexidine gluconate (CH)-induced peritoneal fibrosis (PF) in rats and assessed peritoneal tissue levels of metalloproteinase (MMP)-2 and tissue inhibitors of metalloproteinases (TIMP)-2. Wistar albino rats ( n = 8 per group) were treated as follows: control group, 3 ml/day of 0.9% saline intra-peritoneally for 28 days; CH group, 0.1% CH (200 g [3 ml]/day) in 15% ethanol and 0.9% saline intra-peritoneally for 28 days; CH + interferon (IFN) group, CH (as above) plus pegylated IFN-α2b 1.5 μg/kg per week subcutaneously on days 0, 7, 14, 21 and 28; IFN group, pegylated IFN-α2b (as above). Parietal peritoneum samples were obtained from the left anterior abdominal wall after 35 days. Parietal thickness, degree of vascular proliferation and inflammation, and MMP-2 and TIMP-2 levels were determined. The mean peritoneal thicknesses of the control, CH, CH + IFN and IFN groups were 7.02 ± 3.89, 156.86 ± 29.13, 59.88 ± 22.1, 9.27 ± 2.03 μm, respectively. Pegylated IFN-α2b decreased CH-induced expression of MMP-2 in the parietal peritoneum, but had no effect on TIMP-2 levels. Further studies are needed to determine the optimal dosage and duration for pegylated IFN-α2b treatment.
Collapse
Affiliation(s)
- E Ucar
- Department of Nephrology, Erzurum Education and Research Hospital, Erzurum, Turkey
| | - A Borazan
- Department of Nephrology, Erzurum Education and Research Hospital, Erzurum, Turkey
| | - E Semerci
- Department of General Surgery, Erzurum Education and Research Hospital, Erzurum, Turkey
| | - DN Binici
- Department of Internal Medicine, Erzurum Education and Research Hospital, Erzurum, Turkey
| | - M Yaldiz
- Department of Pathology, Mustafa Kemal University, Hatay, Turkey
| | - A Aysal
- Department of Pathology, Dokuz Eylul University, Izmir, Turkey
| | - E Altug
- Department of General Surgery, Faculty of Veterinary Medicine, Mustafa Kemal University, Hatay, Turkey
| | - C Kuvandik
- Department of Infectious Disease, Kırıkhan Government Hospital, Hatay, Turkey
| | - C Huzmeli
- Department of Nephrology, Erzurum Education and Research Hospital, Erzurum, Turkey
| | - T Yetim
- Department of Nephrology, Erzurum Education and Research Hospital, Erzurum, Turkey
| | - S Canda
- Department of Pathology, Mustafa Kemal University, Hatay, Turkey
| |
Collapse
|
26
|
Yildirim A, Ozkan OV, Aslan A, Koseoglu Z, Borazan A. The effects of low-dose erythropoiesis-stimulating agents on peritoneal fibrosis induced by chemical peritonitis and on peritoneal tissue MMP-2 and TIMP-2 Levels in rats. Ren Fail 2010; 31:567-72. [PMID: 19839852 DOI: 10.1080/08860220903033716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
AIM The objective of the present study was to investigate the effect of low-dose erytropoesis-stimulating agents (ESA) on the development of peritoneal fibrosis in chlorhexidine gluconate-induced peritoneal sclerosing rats and to assess the peritoneal tissue levels of MMP-2 and TIMP-2, which may be regarded as factors in the development of peritoneal fibrosis. SUBJECTS AND METHODS Twenty-four Wistar albino rats were divided into three groups. The control group received 0.9% saline (3 ml/d) intraperitoneally, the CH group received 3 ml daily injections of 0.1% chlorhexidine gluconate (CH) intraperitoneally, and the CH+ESA group received 3 ml daily injections of 0.1% CH intraperitoneally and epoetin beta (3 x 20 IU/kg/week) subcutaneously. On the twenth-first day, rats were sacrificed, and parietal peritoneum samples were obtained from the left anterior abdominal wall. Pathological samples were examined using Hematoxyline & Eosin (HE) stains. The thickness, vascular proliferation, and inflammation were determined by light microscopy. MMP-2 and TIMP-2 were studied immunohistochemically by monoclonal antibody staining. RESULTS Inflammation, vascular proliferation, and fibrotic area percentages were not statistically significant between groups. Histopathologically control, CH, CH+ESA groups peritoneal thickness were 8.02 +/- 2.89, 146.74 +/- 26.1, and 48.12 +/- 16.8 micrometers, respectively. The decrease in thickness of parietal peritoneum in CH+ESA group was statistically significant when compared to CH. Immunohistochemically, interferon was shown to decrease MMP-2 expression on parietal peritoneum than group CH, but has no effect on TIMP-2. DISCUSSION Low-dose ESA histopatologically reduces peritoneal fibrosis induced by chlorhexidine gluconate. However, from dosage and duration points of view, we need extended clinical and experimental studies.
Collapse
Affiliation(s)
- Ayse Yildirim
- Mustafa Kemal University Faculty of Medicine, Department of Histology and Embryology, Hatay, Turkey.
| | | | | | | | | |
Collapse
|
27
|
Schilte MN, Celie JW, ter Wee PM, Beelen RH, van den Born J. Factors Contributing to Peritoneal Tissue Remodeling in Peritoneal Dialysis. Perit Dial Int 2009. [DOI: 10.1177/089686080902900604] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Peritoneal dialysis (PD) is associated with functional and structural changes of the peritoneal membrane. In this review we describe factors contributing to peritoneal tissue remodeling, including uremia, peritonitis, volume loading, the presence of a catheter, and the PD fluid itself. These factors initiate recruitment and activation of peritoneal cells such as macrophages and mast cells, as well as activation of peritoneal cells, including mesothelial cells, fibroblasts, and endothelial cells. We provide an overview of cytokines, growth factors, and other mediators involved in PD-associated changes. Activation of downstream pathways of cellular modulators can induce peritoneal tissue remodeling, leading to ultrafiltration loss. Identification of molecular pathways, cells, and cytokines involved in the development of angiogenesis, fibrosis, and membrane failure may lead to innovative therapeutic strategies that can protect the peritoneal membrane from the consequences of long-term PD.
Collapse
Affiliation(s)
- Margot N. Schilte
- Departments of Molecular Cell Biology and Immunology VU University Medical Center, Amsterdam, The Netherlands
| | - Johanna W.A.M Celie
- Departments of Molecular Cell Biology and Immunology VU University Medical Center, Amsterdam, The Netherlands
| | - Piet M. ter Wee
- Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - Robert H.J. Beelen
- Departments of Molecular Cell Biology and Immunology VU University Medical Center, Amsterdam, The Netherlands
| | - Jacob van den Born
- Departments of Molecular Cell Biology and Immunology VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Angiotensin II upregulates Toll-like receptor 4 and enhances lipopolysaccharide-induced CD40 expression in rat peritoneal mesothelial cells. Inflamm Res 2009; 58:473-82. [PMID: 19271152 DOI: 10.1007/s00011-009-0012-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2008] [Revised: 12/23/2008] [Accepted: 01/11/2009] [Indexed: 10/21/2022] Open
Abstract
OBJECTIVE Activation of Toll-like receptor 4 (TLR4) in peritoneal mesothelial cells by endotoxin contributes to peritoneal inflammation and fibrosis. Here we investigated TLR4 expression induced by angiotensin II (Ang II) and functional consequences of nuclear factor-kappaB (NF-kappaB) activation and CD40 expression in rat peritoneal mesothelial cells (RPMCs). METHODS TLR4, CD40, tumor necrosis factor-alpha (TNF-alpha), and interleukin-6 (IL-6) were determined by reverse transcription polymerase chain reaction (RT-PCR) and TLR4, IkappaBalpha, phospho-IkappaBalpha, NF-kappaB p65, and phospho-NF-kappaB p65 were analyzed by Western blot. The intracellular distribution of NF-kappaB p65 was detected by immunofluorescence. RESULTS Treatment of RPMCs with Ang II resulted in an increase in the expression of TLR4 mRNA and protein levels. Preincubation of RPMCs with Ang II significantly increased lipopolysaccharide (LPS)-induced phospho-IkappaBalpha and phospho-NF-kappaB p65 protein (P < 0.05 vs. LPS alone) and CD40, TNF-alpha, and IL-6 mRNA levels (P < 0.05 vs. LPS alone). A significantly increased nuclear staining of NF-kappaB p65 in cells treated with Ang II plus LPS was also observed. CONCLUSIONS Ang II upregulates the expression of TLR4 by RPMCs, resulting in enhanced NF-kappaB signaling and induction of CD40, TNF-alpha, and IL-6 expression. Locally produced Ang II in the peritoneum may have an amplified role in LPS-induced peritoneal inflammation.
Collapse
|
29
|
Bozkurt D, Cetin P, Sipahi S, Hur E, Nar H, Ertilav M, Sezak M, Duman S. The Effects of Renin–angiotensin System Inhibition on Regression of Encapsulating Peritoneal Sclerosis. Perit Dial Int 2008. [DOI: 10.1177/089686080802805s08] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background Encapsulating peritoneal sclerosis (EPS) is a clinical syndrome associated with symptoms of ileus and irreversible sclerosis of both visceral and parietal peritoneum. Peritoneal dialysis (PD) patients rarely develop EPS, a severe life-threatening condition of unknown pathogenesis. Angiotensin II is known to promote fibrosis and inflammation in various tissues. Renin–angiotensin system (RAS) blockade provides advantages in the course of diseases such as hypertension, chronic kidney disease, and proteinuria. We have also previously shown that RAS blockade has beneficial effects on hypertonic (3.86%) PD solution-induced peritoneal alterations. Because it shares the same characteristics as other fibrotic processes, peritoneal fibrosis can benefit from RAS blockade. Objective To determine the advantages of RAS blockade in regression of EPS. Methods We divided 56 nonuremic albino Wistar rats into 6 groups: control group ( n = 10), daily intraperitoneal (IP) injection of 2 mL isotonic saline for 3 weeks; CG group ( n = 10), daily IP injection of 2 mL/200 g chlorhexidine gluconate (CG) for 3 weeks; resting group ( n = 10), daily IP injection of CG (0 – 3 weeks) plus peritoneal rest (4 – 6 weeks). After 3 weeks of being injected with CG (0 – 3 weeks), a fourth group ( n = 9) was treated with 100 mg/L enalapril (ENA group); a fifth group ( n = 10) was treated with 80 mg/L valsartan (VAL group), and a sixth group ( n = 7) was treated with 100 mg/L enalapril + 80 mg/L valsartan (ENA+VAL group) in drinking water for an additional 3 weeks (4 – 6 weeks). At the end, a 1-hour peritoneal equilibration test was performed with 25 mL 3.86% PD solution. Dialysate-to-plasma ratio of urea (D/P urea), dialysate WBC count, ultrafiltration volume (UF), and morphological changes of parietal peritoneum were examined. Results Exposure to CG for 3 weeks resulted in alterations in peritoneal transport (increased D/P urea, decreased UF volume; p < 0.05) and morphology (increased inflammation, neovascularization, fibrosis, and peritoneal thickness; p < 0.05). Peritoneal rest had some beneficial effect only on UF failure and dialysate cell count ( p < 0.05). However, RAS blockade was more effective than peritoneal rest with respect to UF volume, vascularity ( p < 0.05), and peritoneal thickness ( p > 0.05). Dual blockade of RAS had no additional beneficial effects. Conclusion We suggest that RAS blockade either with angiotensin-converting enzyme inhibitors or angiotensin receptor blockers may be a more effective option than resting in the management of EPS.
Collapse
Affiliation(s)
- Devrim Bozkurt
- Departments of Nephrology, Ege University, Izmir, Turkey
| | - Pinar Cetin
- Departments of Nephrology, Ege University, Izmir, Turkey
| | - Savas Sipahi
- Departments of Nephrology, Ege University, Izmir, Turkey
| | - Ender Hur
- Departments of Nephrology, Ege University, Izmir, Turkey
| | - Hasim Nar
- Departments of Nephrology, Ege University, Izmir, Turkey
| | | | | | - Soner Duman
- Departments of Nephrology, Ege University, Izmir, Turkey
| |
Collapse
|