1
|
Ikushima A, Ishimura T, Mori KP, Yamada H, Sugioka S, Ishii A, Toda N, Ohno S, Kato Y, Handa T, Yanagita M, Yokoi H. Deletion of p38 MAPK in macrophages ameliorates peritoneal fibrosis and inflammation in peritoneal dialysis. Sci Rep 2024; 14:21220. [PMID: 39261560 PMCID: PMC11391064 DOI: 10.1038/s41598-024-71859-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024] Open
Abstract
One of the most common causes of peritoneal dialysis withdrawal is ultrafiltration failure which is characterized by peritoneal membrane thickening and fibrosis. Although previous studies have demonstrated the inhibitory effect of p38 MAPK inhibitors on peritoneal fibrosis in mice, it was unclear which specific cells contribute to peritoneal fibrosis. To investigate the role of p38 MAPK in peritoneal fibrosis more precisely, we examined the expression of p38 MAPK in human peritoneum and generated systemic inducible p38 MAPK knockout mice and macrophage-specific p38 MAPK knockout mice. Furthermore, the response to lipopolysaccharide (LPS) was assessed in p38 MAPK-knocked down RAW 264.7 cells to further explore the role of p38 MAPK in macrophages. We found that phosphorylated p38 MAPK levels were increased in the thickened peritoneum of both human and mice. Both chlorhexidine gluconate (CG)-treated systemic inducible and macrophage-specific p38 MAPK knockout mice ameliorated peritoneal thickening, mRNA expression related to inflammation and fibrosis, and the number of αSMA- and MAC-2-positive cells in the peritoneum compared to CG control mice. Reduction of p38 MAPK in RAW 264.7 cells suppressed inflammatory mRNA expression induced by LPS. These findings suggest that p38 MAPK in macrophages plays a critical role in peritoneal inflammation and thickening.
Collapse
Affiliation(s)
- Akie Ikushima
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Takuya Ishimura
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Keita P Mori
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
- Department of Nephrology and Dialysis, Medical Research Institute KITANO HOSPITAL, PIIF Tazuke-Kofukai, Osaka, Japan
| | - Hiroyuki Yamada
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
- Department of Primary Care & Emergency Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sayaka Sugioka
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Akira Ishii
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
- Department of Nephrology, Kansai Electric Power Hospital, Osaka, Japan
| | - Naohiro Toda
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
- Department of Nephrology, Kansai Electric Power Hospital, Osaka, Japan
| | - Shoko Ohno
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Yukiko Kato
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
| | - Takaya Handa
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
- Department of Nephrology and Dialysis, Medical Research Institute KITANO HOSPITAL, PIIF Tazuke-Kofukai, Osaka, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Hideki Yokoi
- Department of Nephrology, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, Kyoto, 6068507, Japan.
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan.
| |
Collapse
|
2
|
Pap D, Pajtók C, Veres-Székely A, Szebeni B, Szász C, Bokrossy P, Zrufkó R, Vannay Á, Tulassay T, Szabó AJ. High Salt Promotes Inflammatory and Fibrotic Response in Peritoneal Cells. Int J Mol Sci 2023; 24:13765. [PMID: 37762068 PMCID: PMC10531298 DOI: 10.3390/ijms241813765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Recent studies draw attention to how excessive salt (NaCl) intake induces fibrotic alterations in the peritoneum through sodium accumulation and osmotic events. The aim of our study was to better understand the underlying mechanisms. The effects of additional NaCl were investigated on human primary mesothelial cells (HPMC), human primary peritoneal fibroblasts (HPF), endothelial cells (HUVEC), immune cells (PBMC), as well as ex vivo on peritoneal tissue samples. Our results showed that a high-salt environment and the consequently increased osmolarity increase the production of inflammatory cytokines, profibrotic growth factors, and components of the renin-angiotensin-aldosterone system, including IL1B, IL6, MCP1, TGFB1, PDGFB, CTGF, Renin and Ace both in vitro and ex vivo. We also demonstrated that high salt induces mesenchymal transition by decreasing the expression of epithelial marker CDH1 and increasing the expression of mesenchymal marker ACTA2 and SNAIL1 in HPMCs, HUVECs and peritoneal samples. Furthermore, high salt increased extracellular matrix production in HPFs. We demonstrated that excess Na+ and the consequently increased osmolarity induce a comprehensive profibrotic response in the peritoneal cells, thereby facilitating the development of peritoneal fibrosis.
Collapse
Affiliation(s)
- Domonkos Pap
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1085 Budapest, Hungary
- HUN-REN–SU Pediatrics and Nephrology Research Group, 1052 Budapest, Hungary
| | - Csenge Pajtók
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1085 Budapest, Hungary
| | - Apor Veres-Székely
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1085 Budapest, Hungary
- HUN-REN–SU Pediatrics and Nephrology Research Group, 1052 Budapest, Hungary
| | - Beáta Szebeni
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1085 Budapest, Hungary
- HUN-REN–SU Pediatrics and Nephrology Research Group, 1052 Budapest, Hungary
| | - Csenge Szász
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1085 Budapest, Hungary
| | - Péter Bokrossy
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1085 Budapest, Hungary
| | - Réka Zrufkó
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1085 Budapest, Hungary
| | - Ádám Vannay
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1085 Budapest, Hungary
- HUN-REN–SU Pediatrics and Nephrology Research Group, 1052 Budapest, Hungary
| | - Tivadar Tulassay
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1085 Budapest, Hungary
- HUN-REN–SU Pediatrics and Nephrology Research Group, 1052 Budapest, Hungary
| | - Attila J. Szabó
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1085 Budapest, Hungary
- HUN-REN–SU Pediatrics and Nephrology Research Group, 1052 Budapest, Hungary
| |
Collapse
|
3
|
Zhao H, Zhang HL, Jia L. High glucose dialysate-induced peritoneal fibrosis: Pathophysiology, underlying mechanisms and potential therapeutic strategies. Biomed Pharmacother 2023; 165:115246. [PMID: 37523983 DOI: 10.1016/j.biopha.2023.115246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023] Open
Abstract
Peritoneal dialysis is an efficient renal replacement therapy for patients with end-stage kidney disease. However, continuous exposure of the peritoneal membrane to dialysate frequently leads to peritoneal fibrosis, which alters the function of the peritoneal membrane and results in withdrawal from peritoneal dialysis in patients. Among others, high glucose dialysate is considered as a predisposing factor for peritoneal fibrosis in patients on peritoneal dialysis. Glucose-induced inflammation, metabolism disturbance, activation of the renin-angiotensin-aldosterone system, angiogenesis and noninflammation-induced reactive oxygen species are implicated in the pathogenesis of high glucose dialysate-induced peritoneal fibrosis. Specifically, high glucose causes chronic inflammation and recurrent peritonitis, which could cause migration and polarization of inflammatory cells, as well as release of cytokines and fibrosis. High glucose also interferes with lipid metabolism and glycolysis by activating the sterol-regulatory element-binding protein-2/cleavage-activating protein pathway and increasing hypoxia inducible factor-1α expression, leading to angiogenesis and peritoneal fibrosis. Activation of the renin-angiotensin-aldosterone system and Ras-mitogen activated protein kinase signaling pathway is another contributing factor in high glucose dialysate-induced fibrosis. Ultimately, activation of the transforming growth factor-β1/Smad pathway is involved in mesothelial-mesenchymal transition or epithelial-mesenchymal transition, which leads to the development of fibrosis. Although possible intervention strategies for peritoneal dialysate-induced fibrosis by targeting the transforming growth factor-β1/Smad pathway have occasionally been proposed, lack of laboratory evidence renders clinical decision-making difficult. We therefore aim to revisit the upstream pathways of transforming growth factor-beta1/Smad and propose potential therapeutic targets for high glucose-induced peritoneal fibrosis.
Collapse
Affiliation(s)
- Hanxue Zhao
- First Clinical Medical College, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Beijing 100053, China
| | - Hong-Liang Zhang
- Department of Life Sciences, National Natural Science Foundation of China, No. 83 Shuangqing Road, Beijing 100085, China.
| | - Linpei Jia
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Beijing 100053, China.
| |
Collapse
|
4
|
Jubaidi FF, Zainalabidin S, Taib IS, Abdul Hamid Z, Mohamad Anuar NN, Jalil J, Mohd Nor NA, Budin SB. The Role of PKC-MAPK Signalling Pathways in the Development of Hyperglycemia-Induced Cardiovascular Complications. Int J Mol Sci 2022; 23:ijms23158582. [PMID: 35955714 PMCID: PMC9369123 DOI: 10.3390/ijms23158582] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular disease is the most common cause of death among diabetic patients worldwide. Hence, cardiovascular wellbeing in diabetic patients requires utmost importance in disease management. Recent studies have demonstrated that protein kinase C activation plays a vital role in the development of cardiovascular complications via its activation of mitogen-activated protein kinase (MAPK) cascades, also known as PKC-MAPK pathways. In fact, persistent hyperglycaemia in diabetic conditions contribute to preserved PKC activation mediated by excessive production of diacylglycerol (DAG) and oxidative stress. PKC-MAPK pathways are involved in several cellular responses, including enhancing oxidative stress and activating signalling pathways that lead to uncontrolled cardiac and vascular remodelling and their subsequent dysfunction. In this review, we discuss the recent discovery on the role of PKC-MAPK pathways, the mechanisms involved in the development and progression of diabetic cardiovascular complications, and their potential as therapeutic targets for cardiovascular management in diabetic patients.
Collapse
Affiliation(s)
- Fatin Farhana Jubaidi
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
- Correspondence: (F.F.J.); (S.B.B.); Tel.: +603-9289-7645 (S.S.B.)
| | - Satirah Zainalabidin
- Center for Toxicology and Health Risk Research, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.Z.); (N.N.M.A.)
| | - Izatus Shima Taib
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
| | - Zariyantey Abdul Hamid
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
| | - Nur Najmi Mohamad Anuar
- Center for Toxicology and Health Risk Research, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.Z.); (N.N.M.A.)
| | - Juriyati Jalil
- Center for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Nor Anizah Mohd Nor
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
- Faculty of Health Sciences, University College MAIWP International, Kuala Lumpur 68100, Malaysia
| | - Siti Balkis Budin
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
- Correspondence: (F.F.J.); (S.B.B.); Tel.: +603-9289-7645 (S.S.B.)
| |
Collapse
|
5
|
Barone M, Ippoliti M, Mucilli F. SARS-CoV-2 peritoneal positivity and emergency surgery: is there any putative predisposing factor? Updates Surg 2021; 73:1593-1595. [PMID: 33939133 PMCID: PMC8091152 DOI: 10.1007/s13304-021-01066-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 10/29/2022]
Affiliation(s)
- Mirko Barone
- Department of General and Thoracic Surgery, SS. Annunziata Hospital, Chieti, Italy. .,Department of General and Oncological Surgery, SS. Annunziata Hospital, Via dei Vestini n.1, 66100, Chieti, Italy.
| | - Massimo Ippoliti
- Department of General and Thoracic Surgery, SS. Annunziata Hospital, Chieti, Italy
| | - Felice Mucilli
- Department of General and Thoracic Surgery, SS. Annunziata Hospital, Chieti, Italy.,University Department of Medical, Oral and Biotechnological Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
6
|
Liu J, Feng Y, Li N, Shao QY, Zhang QY, Sun C, Xu PF, Jiang CM. Activation of the RAS contributes to peritoneal fibrosis via dysregulation of low-density lipoprotein receptor. Am J Physiol Renal Physiol 2021; 320:F273-F284. [PMID: 33427062 DOI: 10.1152/ajprenal.00149.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 12/01/2020] [Indexed: 12/31/2022] Open
Abstract
Peritoneal dialysis (PD)-related peritoneal fibrosis (PF) is characterized by progressive extracellular matrix (ECM) accumulation in peritoneal mesothelial cells (PMCs) during long-term use of high glucose (HG)-based dialysates. Activation of the renin-angiotensin system (RAS) has been shown to be associated with PF. The aim of this study was to explore the underlying mechanism of the RAS in HG-induced PF. We treated C57BL/6 mice and a human PMC line with HG to induce a PF model and to stimulate ECM accumulation, respectively. RAS activity was blocked using valsartan or angiotensin II (ANGII) type 1 receptor siRNA. The major findings were as follows. First, mice in the HG group exhibited increased collagen deposition and expression of ECM proteins, including α-smooth muscle actin (α-SMA) and collagen type I in the peritoneum. Consistent with the in vivo data, HG upregulated α-SMA expression in human peritoneal mesothelial cells (HPMCs) in a time- and dose-dependent manner. Second, HG stimulation led to RAS activation in HPMCs, and inactivation of RAS decreased the expression of ECM proteins in vivo and in vitro, even during HG stimulation. Finally, RAS-mediated ECM production was associated with lipid accumulation in HPMCs and depended on the dysregulation of the low-density lipoprotein receptor (LDLr) pathway. HG-stimulated HPMCs showed increased coexpression of LDLr and α-SMA, whereas blockade of RAS activity reversed the effect. Furthermore, inhibition of LDLr signaling decreased α-SMA and collagen type I expression in HPMCs when treated with HG and ANG II. In conclusion, increased intracellular RAS activity impaired lipid homeostasis and induced ECM accumulation in HPMCs by disrupting the LDLr pathway, which contributed to PF.
Collapse
Affiliation(s)
- Jing Liu
- Institute of Nephrology, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Yuan Feng
- Institute of Nephrology, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Nan Li
- Institute of Nephrology, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Qiu-Yuan Shao
- Institute of Nephrology, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Qing-Yan Zhang
- Institute of Nephrology, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Cheng Sun
- Institute of Nephrology, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Peng-Fei Xu
- Institute of Nephrology, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Chun-Ming Jiang
- Institute of Nephrology, Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| |
Collapse
|
7
|
Abstract
Peritoneal fibrosis (PF) is invariably observed in patients undergoing long-term peritoneal dialysis (PD). The condition is thought to occur in response to a variety of insults, including bioincompatible dialysates (acidic solution, high glucose, glucose degradation products, or a combination), peritonitis, uremia, and chronic inflammation. Recently, the pathophysiologic mechanisms that contribute to the fibrosing process have been intensively studied. Transforming growth factor-β has been shown to be a key mediator of PF. Loss of the mesothelial cell layer has been identified in several studies and shown to correlate with submesothelial thickening and vasculopathy. An association has also been identified between increased submesothelial thickness in the peritoneal membrane and increased solute transport, suggesting a relationship between PF and loss of ultrafiltration capacity. Thus, to maintain long-term PD and improve quality of life for patients, it is important to develop interventions for prevention and treatment of PF. Several strategies for peritoneal fibrosis intervention have been reported, including developing biocompatible dialysate, targeting mediators responsible for inflammation and fibrosis, and reconstituting the peritoneum using mesothelial or bone marrow–derived cells. Recent experimental trials in animal models and clinical studies are presented in this review.
Collapse
Affiliation(s)
- Kayo Kaneko
- Division of Nephrology, Department of Internal Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Chieko Hamada
- Division of Nephrology, Department of Internal Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Yasuhiko Tomino
- Division of Nephrology, Department of Internal Medicine, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Increased miR-7641 Levels in Peritoneal Hyalinizing Vasculopathy in Long-Term Peritoneal Dialysis Patients. Int J Mol Sci 2020; 21:ijms21165824. [PMID: 32823722 PMCID: PMC7461593 DOI: 10.3390/ijms21165824] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
Peritoneal hyalinizing vasculopathy (PHV) represents the cornerstone of long-term peritoneal dialysis (PD), and especially characterizes patients associated with encapsulating peritoneal sclerosis. However, the mechanisms of PHV development remain unknown. A cross sectional study was performed in 100 non-selected peritoneal biopsies of PD patients. Clinical data were collected and lesions were evaluated by immunohistochemistry. In selected biopsies a microRNA (miRNA)-sequencing analysis was performed. Only fifteen patients (15%) showed PHV at different degrees. PHV prevalence was significantly lower among patients using PD fluids containing low glucose degradation products (GDP) (5.9% vs. 24.5%), angiotensin converting enzyme inhibitors (ACEIs) (7.5% vs. 23.4%), statins (6.5% vs. 22.6%) or presenting residual renal function, suggesting the existence of several PHV protective factors. Peritoneal biopsies from PHV samples showed loss of endothelial markers and induction of mesenchymal proteins, associated with collagen IV accumulation and wide reduplication of the basement membrane. Moreover, co-expression of endothelial and mesenchymal markers, as well as TGF-β1/Smad3 signaling activation were found in PHV biopsies. These findings suggest that an endothelial-to-mesenchymal transition (EndMT) process was taking place. Additionally, significantly higher levels of miR-7641 were observed in severe PHV compared to non-PHV peritoneal biopsies. Peritoneal damage by GDPs induce miRNA deregulation and an EndMT process in submesothelial vessels, which could contribute to collagen IV accumulation and PHV.
Collapse
|
9
|
Wang Y, Shi Y, Tao M, Zhuang S, Liu N. Peritoneal fibrosis and epigenetic modulation. Perit Dial Int 2020; 41:168-178. [PMID: 32662737 DOI: 10.1177/0896860820938239] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Peritoneal dialysis (PD) is an effective treatment for patients with end-stage renal disease. However, peritoneal fibrosis (PF) is a common complication that ultimately leads to ultrafiltration failure and discontinuation of PD after long-term PD therapy. There is currently no effective therapy to prevent or delay this pathologic process. Recent studies have reported epigenetic modifications involved in PF, and accumulating evidence suggests that epigenetic therapies may have the potential to prevent and treat PF clinically. The major epigenetic modifications in PF include DNA methylation, histone modification, and noncoding RNAs. The mechanisms of epigenetic regulation in PF are complex, predominantly involving modification of signaling molecules, transcriptional factors, and genes. This review will describe the mechanisms of epigenetic modulation in PF and discuss the possibility of targeting them to prevent and treat this complication.
Collapse
Affiliation(s)
- Yi Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, China
| | - Yingfeng Shi
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, China
| | - Min Tao
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, China.,Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, USA
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, China
| |
Collapse
|
10
|
Menikdiwela KR, Ramalingam L, Abbas MM, Bensmail H, Scoggin S, Kalupahana NS, Palat A, Gunaratne P, Moustaid-Moussa N. Role of microRNA 690 in Mediating Angiotensin II Effects on Inflammation and Endoplasmic Reticulum Stress. Cells 2020; 9:cells9061327. [PMID: 32466437 PMCID: PMC7348980 DOI: 10.3390/cells9061327] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/15/2020] [Accepted: 05/22/2020] [Indexed: 12/27/2022] Open
Abstract
Overactivation of the renin–angiotensin system (RAS) during obesity disrupts adipocyte metabolic homeostasis and induces endoplasmic reticulum (ER) stress and inflammation; however, underlying mechanisms are not well known. We propose that overexpression of angiotensinogen (Agt), the precursor protein of RAS in adipose tissue or treatment of adipocytes with Angiotensin II (Ang II), RAS bioactive hormone, alters specific microRNAs (miRNA), that target ER stress and inflammation leading to adipocyte dysfunction. Epididymal white adipose tissue (WAT) from B6 wild type (Wt) and transgenic male mice overexpressing Agt (Agt-Tg) in adipose tissue and adipocytes treated with Ang II were used. Small RNA sequencing and microarray in WAT identified differentially expressed miRNAs and genes, out of which miR-690 and mitogen-activated protein kinase kinase 3 (MAP2K3) were validated as significantly up- and down-regulated, respectively, in Agt-Tg, and in Ang II-treated adipocytes compared to respective controls. Additionally, the direct regulatory role of miR-690 on MAP2K3 was confirmed using mimic, inhibitors and dual-luciferase reporter assay. Downstream protein targets of MAP2K3 which include p38, NF-κB, IL-6 and CHOP were all reduced. These results indicate a critical post-transcriptional role for miR-690 in inflammation and ER stress. In conclusion, miR-690 plays a protective function and could be a useful target to reduce obesity.
Collapse
Affiliation(s)
- Kalhara R. Menikdiwela
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.R.M.); (L.R.); (S.S.); (N.S.K.)
| | - Latha Ramalingam
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.R.M.); (L.R.); (S.S.); (N.S.K.)
| | - Mostafa M. Abbas
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha 34110, Qatar; (M.M.A.); (H.B.)
- Department of Imaging Science and Innovation, Geisinger Health System, Danville, PA 17822, USA
| | - Halima Bensmail
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha 34110, Qatar; (M.M.A.); (H.B.)
| | - Shane Scoggin
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.R.M.); (L.R.); (S.S.); (N.S.K.)
| | - Nishan S. Kalupahana
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.R.M.); (L.R.); (S.S.); (N.S.K.)
- Department of Physiology, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Asha Palat
- Biology and Biochemistry, University of Houston, Houston, TX 77204, USA; (A.P.); (P.G.)
| | - Preethi Gunaratne
- Biology and Biochemistry, University of Houston, Houston, TX 77204, USA; (A.P.); (P.G.)
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.R.M.); (L.R.); (S.S.); (N.S.K.)
- Correspondence: ; Tel.: +806-834-7946
| |
Collapse
|
11
|
Shimizu M, Ishibashi Y, Taki F, Shimizu H, Hirahara I, Kaname S, Fujita T. EndothelinB Receptor Blocker Inhibits High Glucose-Induced Synthesis of Fibronectin in Human Peritoneal Mesothelial Cells. Perit Dial Int 2020. [DOI: 10.1177/089686080602600318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background Long-term peritoneal dialysis using glucose-based dialysates is associated with peritoneal fibrosis. The object of this study was to investigate the hypothesis that endothelin (ET)-1, which is known to play an important role in various fibrotic diseases, may also be involved in peritoneal fibrosis using human peritoneal mesothelial cells (HPMC). Methods HPMC were cultured with 4% d- or l-glucose, or loaded with 10 nmol/L ET-1. In some experiments, the ETA receptor antagonist BQ-123, the ETB receptor antagonist BQ-788, and antioxidants 4-hydroxy-2,2,6,6-tetramethylpiperidine 1-oxyl (TEMPOL) and diphenyleneiodium chloride (DPI) were used. mRNA expression of ET-1, ETA receptor, ETB receptor, and fibronectin (FN) was analyzed by real-time polymerase chain reaction (real-time PCR). The protein levels for FN and ET-1 were measured by ELISA. CM-H2DCFDA-sensitive reactive oxygen species (ROS) were evaluated by flow cytometry. Results d-Glucose significantly induced mRNA expression of ET-1 and the ETB receptor but not the ETA receptor. FN production under high glucose conditions was inhibited by BQ-788. ET-1 directly stimulated HPMC to increase mRNA expression of FN and CM-H2DCFDA-sensitive ROS production. BQ-788, TEMPOL, and DPI inhibited mRNA expression of FN induced by ET-1. Conclusion The present study suggests that high-glucose-induced FN synthesis is mediated by the ET-1/ETB receptor pathway and, therefore, an ETB receptor antagonist may be useful in preventing FN production in HPMC.
Collapse
Affiliation(s)
- Miyuki Shimizu
- Division of Total Renal Care Medicine, Department of Nephrology and Endocrinology, University of Tokyo School of Medicine, Tokyo
- Terumo Corporation R&D Center, Kanagawa, Japan
| | - Yoshitaka Ishibashi
- Division of Total Renal Care Medicine, Department of Nephrology and Endocrinology, University of Tokyo School of Medicine, Tokyo
| | - Fumika Taki
- Division of Total Renal Care Medicine, Department of Nephrology and Endocrinology, University of Tokyo School of Medicine, Tokyo
| | - Hideki Shimizu
- Division of Total Renal Care Medicine, Department of Nephrology and Endocrinology, University of Tokyo School of Medicine, Tokyo
| | - Ichiro Hirahara
- Division of Total Renal Care Medicine, Department of Nephrology and Endocrinology, University of Tokyo School of Medicine, Tokyo
- Terumo Corporation R&D Center, Kanagawa, Japan
| | - Shinya Kaname
- Division of Total Renal Care Medicine, Department of Nephrology and Endocrinology, University of Tokyo School of Medicine, Tokyo
| | - Toshiro Fujita
- Division of Total Renal Care Medicine, Department of Nephrology and Endocrinology, University of Tokyo School of Medicine, Tokyo
| |
Collapse
|
12
|
Sun L, Yu M, Zhou T, Zhang S, He G, Wang G, Gang X. Current advances in the study of diabetic cardiomyopathy: From clinicopathological features to molecular therapeutics (Review). Mol Med Rep 2019; 20:2051-2062. [PMID: 31322242 DOI: 10.3892/mmr.2019.10473] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 05/29/2019] [Indexed: 11/06/2022] Open
Abstract
The incidence of diabetes mellitus has become a major public health concern due to lifestyle alterations. Moreover, the complications associated with diabetes mellitus deeply influence the quality of life of patients. Diabetic cardiomyopathy (DC) is a type of diabetes mellitus complication characterized by functional and structural damage in the myocardium but not accompanied by coronary arterial disease. Currently, diagnosing and preventing DC is still a challenge for physicians due to its atypical symptoms. For this reason, it is necessary to summarize the current knowledge on DC, especially in regards to the underlying molecular mechanisms toward the goal of developing useful diagnostic approaches and effective drugs based on these mechanisms. There exist several review articles which have focused on these points, but there still remains a lot to learn from published studies. In this review, the features, diagnosis and molecular mechanisms of DC are reviewed. Furthermore, potential therapeutic and prophylactic drugs are discussed.
Collapse
Affiliation(s)
- Lin Sun
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ming Yu
- Department of Cardiology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Tong Zhou
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Siwen Zhang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guangyu He
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
13
|
Tamura R, Doi S, Nakashima A, Sasaki K, Maeda K, Ueno T, Masaki T. Inhibition of the H3K4 methyltransferase SET7/9 ameliorates peritoneal fibrosis. PLoS One 2018; 13:e0196844. [PMID: 29723250 PMCID: PMC5933785 DOI: 10.1371/journal.pone.0196844] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 04/20/2018] [Indexed: 11/18/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1) is a major mediator of peritoneal fibrosis and reportedly affects expression of the H3K4 methyltransferase, SET7/9. SET7/9-induced H3K4 mono-methylation (H3K4me1) critically activates transcription of fibrosis-related genes. In this study, we examined the effect of SET7/9 inhibition on peritoneal fibrosis in mice and in human peritoneal mesothelial cells (HPMCs). We also examined SET7/9 expression in nonadherent cells isolated from the effluent of peritoneal dialysis (PD) patients. Murine peritoneal fibrosis was induced by intraperitoneal injection of methylglyoxal (MGO) into male C57/BL6 mice over 21 days. Sinefungin, a SET7/9 inhibitor, was administered subcutaneously just before MGO injection (10 mg/kg). SET7/9 expression was elevated in both MGO-injected mice and nonadherent cells isolated from the effluent of PD patients. SET7/9 expression was positively correlated with dialysate/plasma ratio of creatinine in PD patients. Sinefungin was shown immunohistochemically to suppress expression of mesenchymal cells and collagen deposition, accompanied by decreased H3K4me1 levels. Peritoneal equilibration tests showed that sinefungin attenuated the urea nitrogen transport rate from plasma and the glucose absorption rate from the dialysate. In vitro, sinefungin suppressed TGF-β1-induced expression of fibrotic markers and inhibited H3K4me1. These findings suggest that inhibiting the H3K4 methyltransferase SET7/9 ameliorates peritoneal fibrosis.
Collapse
Affiliation(s)
- Ryo Tamura
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Shigehiro Doi
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
- * E-mail:
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kensuke Sasaki
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kazuya Maeda
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Toshinori Ueno
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
14
|
Acidic organelles mediate TGF-β1-induced cellular fibrosis via (pro)renin receptor and vacuolar ATPase trafficking in human peritoneal mesothelial cells. Sci Rep 2018; 8:2648. [PMID: 29422602 PMCID: PMC5805675 DOI: 10.1038/s41598-018-20940-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/26/2018] [Indexed: 01/30/2023] Open
Abstract
TGF-β1, which can cause renal tubular injury through a vacuolar-type H+-ATPase (V-ATPase)-mediated pathway, is induced by the glucose degradation product methylglyoxal to yield peritoneal injury and fibrosis. The present study investigated the roles of V-ATPase and its accessory protein, the (pro)renin receptor, in peritoneal fibrosis during peritoneal dialysis. Rats daily administered 20 mM methylglyoxal intraperitoneally developed significant peritoneal fibrosis after 7 days with increased expression of TGF-β and V-ATPase, which was reduced by the inhibition of V-ATPase with co-administration of 100 mM bafilomycin A1. The (pro)renin receptor and V-ATPase were expressed in acidic organelles and cell membranes of human peritoneal mesothelial cells. TGF-β1 upregulated the expression of collagens, α-SMA, and EDA-fibronectin, together with ERK1/2 phosphorylation, which was reduced by inhibition of V-ATPase, (pro)renin receptor, or the MAPK pathway. Fibronectin and the soluble (pro)renin receptor were excreted from cells by acidic organelle trafficking in response to TGF-β1; this excretion was also suppressed by inhibition of V-ATPase. Soluble (pro)renin receptor concentrations in effluents of patients undergoing peritoneal dialysis were associated with the dialysate-to-plasma ratio of creatinine. Together, these results demonstrate a novel fibrosis mechanism through the (pro)renin receptor and V-ATPase in the acidic organelles of peritoneal mesothelial cells.
Collapse
|
15
|
Liappas G, González-Mateo G, Aguirre AR, Abensur H, Albar-Vizcaino P, Parra EG, Sandoval P, Ramírez LG, Del Peso G, Acedo JM, Bajo MA, Selgas R, Sánchez Tomero JA, López-Cabrera M, Aguilera A. Nebivolol, a β1-adrenergic blocker, protects from peritoneal membrane damage induced during peritoneal dialysis. Oncotarget 2017; 7:30133-46. [PMID: 27102153 PMCID: PMC5058669 DOI: 10.18632/oncotarget.8780] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 03/31/2016] [Indexed: 01/26/2023] Open
Abstract
Peritoneal dialysis (PD) is a form of renal replacement treatment, which employs the peritoneal membrane (PM) to eliminate toxins that cannot be removed by the kidney. The procedure itself, however, contributes to the loss of the PM ultrafiltration capacity (UFC), leading consequently to the technique malfunction. β-blockers have been considered deleterious for PM due to their association with loss of UFC and induction of fibrosis. Herein we analyzed the effects of Nebivolol, a new generation of β1-blocker, on PM alterations induced by PD fluids (PDF). In vitro: We found that mesothelial cells (MCs) express β1-adrenergic receptor. MCs were treated with TGF-β to induce mesothelial-to-mesenchymal transition (MMT) and co-treated with Nebivolol. Nebivolol reversed the TGF-β effects, decreasing extracellular matrix synthesis, and improved the fibrinolytic capacity, decreasing plasminogen activator inhibitor-1 (PAI-1) and increasing tissue-type plasminogen activator (tPA) supernatant levels. Moreover, Nebivolol partially inhibited MMT and decreased vascular endothelial growth factor (VEGF) and IL-6 levels in supernatants. In vivo: Twenty-one C57BL/6 mice were divided into 3 groups. Control group carried a catheter without PDF infusion. Study group received intraperitoneally PDF and oral Nebivolol during 30 days. PDF group received PDF alone. Nebivolol maintained the UFC and reduced PM thickness, MMT and angiogenesis promoted by PDF. It also improved the fibrinolytic capacity in PD effluents decreasing PAI-1 and IL-8 and increased tPA levels. Conclusion: Nebivolol protects PM from PDF-induced damage, promoting anti-fibrotic, anti-angiogenic, anti-inflammatory and pro-fibrinolytic effects.
Collapse
Affiliation(s)
- Georgios Liappas
- Immunology and Cellular Biology Department, Molecular Biology Centre Severo Ochoa, Madrid, Spain
| | - Guadalupe González-Mateo
- Immunology and Cellular Biology Department, Molecular Biology Centre Severo Ochoa, Madrid, Spain
| | - Anna Rita Aguirre
- Nephrology Department, University of Sao Paulo, School of Medicine, Sao Paulo, Brazil
| | - Hugo Abensur
- Nephrology Department, University of Sao Paulo, School of Medicine, Sao Paulo, Brazil
| | - Patricia Albar-Vizcaino
- Molecular Biology Unit and Nephrology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - Emilio González Parra
- Nephrology Department, Fundación Jiménez-Díaz, Instituto de Investigación Sanitaria, Madrid, Spain
| | - Pilar Sandoval
- Immunology and Cellular Biology Department, Molecular Biology Centre Severo Ochoa, Madrid, Spain
| | - Laura García Ramírez
- Molecular Biology Unit and Nephrology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - Gloria Del Peso
- Nephrology Department, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | | | - María A Bajo
- Nephrology Department, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Rafael Selgas
- Nephrology Department, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - José A Sánchez Tomero
- Molecular Biology Unit and Nephrology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - Manuel López-Cabrera
- Immunology and Cellular Biology Department, Molecular Biology Centre Severo Ochoa, Madrid, Spain
| | - Abelardo Aguilera
- Molecular Biology Unit and Nephrology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| |
Collapse
|
16
|
Ito Y, Kinashi H, Katsuno T, Suzuki Y, Mizuno M. Peritonitis-induced peritoneal injury models for research in peritoneal dialysis review of infectious and non-infectious models. RENAL REPLACEMENT THERAPY 2017. [DOI: 10.1186/s41100-017-0100-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
17
|
Inhibition of H3K9 methyltransferase G9a ameliorates methylglyoxal-induced peritoneal fibrosis. PLoS One 2017; 12:e0173706. [PMID: 28278257 PMCID: PMC5344517 DOI: 10.1371/journal.pone.0173706] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 02/24/2017] [Indexed: 01/28/2023] Open
Abstract
Activity of H3K9 histone methyltransferase G9a is reportedly induced by transforming growth factor-β1 (TGF-β1) and plays an important role in the progression of cancer and fibrosis. In this study, we investigated whether inhibition of G9a-mediated H3K9 methylation attenuates peritoneal fibrosis in mice and human peritoneal mesothelial cells (HPMCs). Nonadherent cells of peritoneal dialysis (PD) patients were isolated from PD effluent to examine expression of G9a. Peritoneal fibrosis was induced by peritoneal injection of methylglyoxal (MGO) in male C57/B6 mice for 3 weeks. BIX01294, a G9a inhibitor, was administered by subcutaneous injection. Effects of BIX01294 on MGO-induced pathological and functional changes in mice were evaluated by immunohistochemistry and a peritoneal equilibration test. HPMCs were isolated from human omentum, and the inhibitory effect of BIX01294 on TGF-β1-induced fibrotic changes was investigated in the HPMCs by western blotting. G9a was upregulated in nonadherent cells of human PD effluent, the peritoneum of MGO-injected mice, and TGF-β1-stimulated HPMCs. BIX01294 significantly reduced the submesothelial zone thickness and cell density in MGO-injected mice. Immunohistochemical staining revealed that BIX01294 treatment decreased not only mono-methylation of H3K9 (H3K9me1), but also the number of mesenchymal cells, accumulation of collagen, and infiltration of monocytes. In addition to the pathological changes, BIX01294 reduced the level of TGF-β1 in peritoneal fluid and improved peritoneal functions. Furthermore, BIX01294 inhibited TGF-β1-induced fibrotic changes along with suppression of H3K9me1 in HPMCs. Therefore, inhibition of H3K9 methyltransferase G9a suppresses peritoneal fibrosis through a reduction of H3K9me1.
Collapse
|
18
|
Kawaoka K, Doi S, Nakashima A, Yamada K, Ueno T, Doi T, Masaki T. Valproic acid attenuates renal fibrosis through the induction of autophagy. Clin Exp Nephrol 2016; 21:771-780. [PMID: 27928635 DOI: 10.1007/s10157-016-1365-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 11/27/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Renal fibrosis is a common pathological feature of the progression of chronic kidney disease. Although valproic acid (VPA) has been recently shown to induce autophagy, the effect of VPA-induced autophagy on renal fibrosis remains unknown. We, therefore, investigated whether VPA-induced autophagy suppresses renal fibrosis in a mouse model of unilateral ureteral obstruction (UUO). METHODS Male C57BL/6 mice were divided into five groups (n = 8 per group): (1) sham group; (2) vehicle group; (3) VPA-treated group; (4) 3-methyladenine (3-MA; autophagy inhibitor)-treated group; and (5) VPA plus 3-MA-treated group. Mice underwent UUO and the kidneys were studied after 5 days. We also investigated the effect of VPA-induced autophagy on α-smooth muscle actin (α-SMA) in transforming growth factor (TGF)-β1-stimulated rat kidney fibroblasts and epithelial cells. RESULTS VPA attenuated renal fibrosis and induced autophagy in UUO mice, while 3-MA increased renal fibrosis and suppressed autophagy. In addition, the anti-fibrotic effect of VPA was diminished by 3-MA in UUO mice. In rat kidney fibroblasts and epithelial cells, VPA suppressed TGF-β1-stimulated α-SMA expression and induced autophagy. In contrast, 3-MA enhanced α-SMA expression while inhibiting autophagy. Furthermore, the combined use of VPA and 3-MA treatments increased the expression of α-SMA compared with VPA treatment alone in TGF-β1-stimulated rat kidney fibroblasts and epithelial cells, which was accompanied by the inhibition of autophagy. CONCLUSION These findings suggest that VPA may be a candidate drug for the treatment of renal fibrosis through the induction of autophagy.
Collapse
Affiliation(s)
- Koichiro Kawaoka
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku, Hiroshima, 734-8551, Japan
| | - Shigehiro Doi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku, Hiroshima, 734-8551, Japan. .,Department of Blood Purification, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku, Hiroshima, 734-8551, Japan.
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku, Hiroshima, 734-8551, Japan
| | - Kyoko Yamada
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku, Hiroshima, 734-8551, Japan
| | - Toshinori Ueno
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku, Hiroshima, 734-8551, Japan
| | - Toshiki Doi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku, Hiroshima, 734-8551, Japan
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
19
|
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: 12] [Impact Index Per Article: 1.3] [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
|
20
|
Hong Z, Hong Z, Wu D, Nie H. Specific MAPK inhibitors prevent hyperglycemia-induced renal diseases in type 1 diabetic mouse model. Mol Cell Biochem 2016; 419:1-9. [DOI: 10.1007/s11010-016-2722-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 05/13/2016] [Indexed: 12/13/2022]
|
21
|
Osma-Garcia IC, Punzón C, Fresno M, Díaz-Muñoz MD. Dose-dependent effects of prostaglandin E2 in macrophage adhesion and migration. Eur J Immunol 2015; 46:677-88. [PMID: 26631603 DOI: 10.1002/eji.201545629] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 10/08/2015] [Accepted: 11/26/2015] [Indexed: 12/15/2022]
Abstract
Macrophage migration to the focus of infection is a hallmark of the innate immune response. Macrophage spreading, adhesion, and migration through the extracellular matrix require dynamic remodeling of the actin cytoskeleton associated to integrin clustering in podosomes and focal adhesions. Here, we show that prostaglandin E2 (PGE2 ), the main prostaglandin produced by macrophages during inflammation, promote the distinctive dose-dependent formation of podosomes or focal adhesions in macrophages. Low concentrations of PGE2 increased p110γ PI3K expression, phosphorylation of actin-related protein 2, and formation of podosomes, which enhanced macrophage migration in response to chemokines. However, high doses of PGE2 increased phosphorylation of paxillin and focal adhesion kinase, the expression of serine/threonine protein kinase 1, and promoted focal adhesion formation and macrophage adhesion, reducing macrophage chemotaxis. In summary, we describe the dual role of PGE2 as a promoter of macrophage chemotaxis and adhesion, proposing a new model of macrophage migration to the inflammatory focus in the presence of a gradient of PGE2 .
Collapse
Affiliation(s)
- Inés C Osma-Garcia
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmen Punzón
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Fresno
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel D Díaz-Muñoz
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
22
|
Palominos MM, Dünner NH, Wabitsch M, Rojas CV. Angiotensin II directly impairs adipogenic differentiation of human preadipose cells. Mol Cell Biochem 2015; 408:115-22. [PMID: 26112903 DOI: 10.1007/s11010-015-2487-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 06/18/2015] [Indexed: 01/20/2023]
Abstract
Angiotensin II reduces adipogenic differentiation of preadipose cells present in the stroma-vascular fraction of human adipose tissue, which also includes several cell types. Because of the ability of non-adipose lineage cells in the stroma-vascular fraction to respond to angiotensin II, it is not possible to unequivocally ascribe the anti-adipogenic response to a direct effect of this hormone on preadipose cells. Therefore, we used the human Simpson-Golabi-Behmel syndrome (SGBS) preadipocyte cell strain to investigate the consequences of angiotensin II treatment on adipogenic differentiation under serum-free conditions, by assessing expression of typical adipocyte markers perilipin and fatty acid-binding protein 4 (FABP4), at the transcript and protein level. Reverse transcription-polymerase chain reaction showed that perilipin and FABP4 transcripts were, respectively, reduced to 0.33 ± 0.07 (P < 0.05) and 0.41 ± 0.19-fold (P < 0.05) in SGBS cells induced to adipogenic differentiation in the presence of angiotensin II. Western Blot analysis corroborated reduction of the corresponding proteins to 0.23 ± 0.21 (P < 0.01) and 0.46 ± 0.30-fold (P < 0.01) the respective controls without angiotensin II. Angiotensin II also impaired morphological changes associated with early adipogenesis. Hence, we demonstrated that angiotensin II is able to directly reduce adipogenic differentiation of SGBS preadipose cells.
Collapse
Affiliation(s)
- Marisol M Palominos
- Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Clasificador 7 Correo 7, Santiago, Chile
| | - Natalia H Dünner
- Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Clasificador 7 Correo 7, Santiago, Chile
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Cecilia V Rojas
- Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Clasificador 7 Correo 7, Santiago, Chile. .,Institute of Nutrition and Food Technology, Universidad de Chile, Casilla, 138-11, Santiago, Chile.
| |
Collapse
|
23
|
Mutsaers SE, Birnie K, Lansley S, Herrick SE, Lim CB, Prêle CM. Mesothelial cells in tissue repair and fibrosis. Front Pharmacol 2015; 6:113. [PMID: 26106328 PMCID: PMC4460327 DOI: 10.3389/fphar.2015.00113] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/12/2015] [Indexed: 12/21/2022] Open
Abstract
Mesothelial cells are fundamental to the maintenance of serosal integrity and homeostasis and play a critical role in normal serosal repair following injury. However, when normal repair mechanisms breakdown, mesothelial cells take on a profibrotic role, secreting inflammatory, and profibrotic mediators, differentiating and migrating into the injured tissues where they contribute to fibrogenesis. The development of new molecular and cell tracking techniques has made it possible to examine the origin of fibrotic cells within damaged tissues and to elucidate the roles they play in inflammation and fibrosis. In addition to secreting proinflammatory mediators and contributing to both coagulation and fibrinolysis, mesothelial cells undergo mesothelial-to-mesenchymal transition, a process analogous to epithelial-to-mesenchymal transition, and become fibrogenic cells. Fibrogenic mesothelial cells have now been identified in tissues where they have not previously been thought to occur, such as within the parenchyma of the fibrotic lung. These findings show a direct role for mesothelial cells in fibrogenesis and open therapeutic strategies to prevent or reverse the fibrotic process.
Collapse
Affiliation(s)
- Steven E Mutsaers
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia and Harry Perkins Institute of Medical Research , Nedlands, WA, Australia ; Institute for Respiratory Health, Centre for Asthma, Allergy and Respiratory Research, School of Medicine and Pharmacology, University of Western Australia , Nedlands, WA, Australia
| | - Kimberly Birnie
- Institute for Respiratory Health, Centre for Asthma, Allergy and Respiratory Research, School of Medicine and Pharmacology, University of Western Australia , Nedlands, WA, Australia
| | - Sally Lansley
- Institute for Respiratory Health, Centre for Asthma, Allergy and Respiratory Research, School of Medicine and Pharmacology, University of Western Australia , Nedlands, WA, Australia
| | - Sarah E Herrick
- Institute of Inflammation and Repair, Faculty of Medical and Human Sciences and Manchester Academic Health Science Centre, University of Manchester , Manchester, UK
| | - Chuan-Bian Lim
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia and Harry Perkins Institute of Medical Research , Nedlands, WA, Australia ; Institute for Respiratory Health, Centre for Asthma, Allergy and Respiratory Research, School of Medicine and Pharmacology, University of Western Australia , Nedlands, WA, Australia
| | - Cecilia M Prêle
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia and Harry Perkins Institute of Medical Research , Nedlands, WA, Australia ; Institute for Respiratory Health, Centre for Asthma, Allergy and Respiratory Research, School of Medicine and Pharmacology, University of Western Australia , Nedlands, WA, Australia
| |
Collapse
|
24
|
Ullian ME, Luttrell LM, Lee MH, Morinelli TA. Stimulation of Cyclooxygenase 2 Expression in Rat Peritoneal Mesothelial Cells. NEPHRON. EXPERIMENTAL NEPHROLOGY 2014; 128:000368673. [PMID: 25531215 DOI: 10.1159/000368673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 09/25/2014] [Indexed: 11/19/2022]
Abstract
Objective: Since peritoneal dialysis causes peritoneal fibrosis, we examined how glucose (osmotic factor), mannitol (osmotic control), and angiotensin II (AngII) regulate proinflammatory cyclooxygenase 2 (COX-2) in primary rat peritoneal mesothelial cells. Materials and Methods: For this study, we used the following material (n = 4-8 cell lines): cells, passages 1-2; 125I-AngII receptor surface binding (AT1R antagonist losartan, AT2R antagonist PD123319; both 10 µM); intracellular calcium probe calcium-5; COX-2 immunoblotting (β-actin normalized); real-time PCR of COX-2 gene PTGS2, and NF-κB inhibitor Ro-1069920 (5 µM). Results: AngII surface receptors were predominantly AT1R (minimally AT2R). AngII and glucose increased COX-2 protein expression concentration dependently; mannitol also increased COX-2 expression. Maximal COX-2 protein expression was observed after 6 h (AngII) and 24 h (glucose, mannitol). The time course of increases in PTGS2 mRNA levels reflected that of COX-2 protein expression. At optimal exposure conditions (time/concentration), glucose was 5-fold more efficacious in stimulating COX-2 protein expression than AngII or mannitol. Losartan fully inhibited COX-2 protein responses to AngII and mannitol, but minimally inhibited responses to glucose. Ro-1069920 fully inhibited COX-2 protein responses to each effector. Conclusion: AngII, glucose, and osmotic stress (mannitol) activate COX-2; NF-κB may be an ideal site for COX-2 blockade, and COX-2 activation by osmotic stress requires AT1R, but activation by glucose is more robust and mechanistically complex. © 2014 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Michael E Ullian
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, S.C., USA
| | | | | | | |
Collapse
|
25
|
Chang TI, Kang HY, Kim KS, Lee SH, Nam BY, Paeng J, Kim S, Park JT, Yoo TH, Kang SW, Han SH. The effect of statin on epithelial-mesenchymal transition in peritoneal mesothelial cells. PLoS One 2014; 9:e109628. [PMID: 25275561 PMCID: PMC4183618 DOI: 10.1371/journal.pone.0109628] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 09/03/2014] [Indexed: 01/05/2023] Open
Abstract
Background Statins have recently been highlighted for their pleiotropic actions distinct from cholesterol-lowering effects. Despite this interest, it is currently unknown whether statin therapy inhibits peritoneal dialysis (PD)-related epithelial-mesenchymal transition (EMT). Methods In vitro, human peritoneal mesothelial cells (HPMCs) were exposed to 5.6 mM glucose (NG) or 100 mM glucose (HG) with or without simvastatin (1 µM). In vivo, PD catheters were inserted into 32 Sprague-Dawley rats, and saline (C, n = 16) or 4.25% peritoneal dialysis fluid (PDF) (PD, n = 16) was infused for 4 weeks. Eight rats from each group were treated with 5 mg/kg/day of simvastatin intraperitoneally. Changes in the protein expression of EMT markers such as E-cadherin, α-SMA, Snail, and fibronectin in HPMCs and the peritoneum were evaluated by Western blot analysis and immunofluorescence or immunohistochemical staining. We also explored whether activation of the mevalonate pathway and its downstream small GTPases were involved in dialysis-related peritoneal EMT and could be inhibited by statin treatment. Results Compared to NG cells, E-cadherin expression was significantly decreased, while α-SMA, Snail, and fibronectin expression were significantly increased in HPMCs exposed to HG, and these changes were abrogated by simvastatin (p<0.05). In addition, the cobblestone-like appearance of normal HPMCs was converted into a fibroblast-like morphology after HG treatment, which was reversed by simvastatin. These EMT-like changes were also observed in HPMCs treated with geranyl-geranyl pyrophosphate (5 µM). HG significantly increased the protein expression of RhoA and Rac1 in the membrane fractions, and these increases were ameliorated by simvastatin (p<0.05). In PD rats, E-cadherin in the peritoneum was significantly decreased, whereas α-SMA, Snail, and fibronectin expression were significantly increased (p<0.05) compared to C rats. The thickness of the mesothelial layer in the peritoneum were also significantly greater in PD rats than in C rats (p<0.05). These changes of the peritoneum in PD rats were significantly attenuated by simvastatin. Conclusion This study demonstrated that PD-related EMT was mediated via the mevalonate pathway, and statin treatment inhibited the EMT changes in HG-treated HPMCs and PDF-stimulated PD rats. These findings suggest that statins may be a promising therapeutic strategy for preservation of peritoneal membrane integrity in long-term PD patients.
Collapse
Affiliation(s)
- Tae Ik Chang
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
- Department of Internal Medicine, NHIS Medical Center, Ilsan Hospital, Goyang-shi, Gyeonggi-do, Korea
| | - Hye-Young Kang
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Kyung Sik Kim
- Department of Surgery, College of Medicine, Brain Korea 21 Project for Medical Sciences, Yonsei University, Seoul, Korea
| | - Sun Ha Lee
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Bo Young Nam
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Jisun Paeng
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Seonghun Kim
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Jung Tak Park
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Tae-Hyun Yoo
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Shin-Wook Kang
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Seung Hyeok Han
- Department of Internal Medicine, College of Medicine, Brain Korea 21 Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
- * E-mail:
| |
Collapse
|
26
|
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: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 01/10/2014] [Indexed: 11/11/2022]
|
27
|
Pan Y, Huang Y, Wang Z, Fang Q, Sun Y, Tong C, Peng K, Wang Y, Miao L, Cai L, Zhao Y, Liang G. Inhibition of MAPK-mediated ACE expression by compound C66 prevents STZ-induced diabetic nephropathy. J Cell Mol Med 2013; 18:231-41. [PMID: 24330074 PMCID: PMC3930410 DOI: 10.1111/jcmm.12175] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 10/02/2013] [Indexed: 01/09/2023] Open
Abstract
A range of in vitro, experimental and clinical intervention studies have implicated an important role for hyperglycaemia-induced activation of the renin-angiotensin system (RAS) in the development and progression of diabetic nephropathy (DN). Blockade of RAS by angiotensin converting enzyme (ACE) inhibitors is an effective strategy in treating diabetic kidney diseases. However, few studies demonstrate the mechanism by which hyperglycaemia up-regulates the expression of ACE gene. Our previous studies have identified a novel curcumin analogue, (2E,6E)-2,6-bis(2-(trifluoromethyl)benzylidene)cyclohexanone (C66), which could inhibit the high glucose (HG)-induced phosphorylation of mitogen-activated protein kinases in mouse macrophages. In this study, we found that the renal protection of C66 in diabetic mice was associated with mitogen-activated protein kinase (MAPK) inactivation and ACE/angiotensin II (Ang II) down-regulation. Generally, MAPKs have been considered as a downstream signalling of Ang II and a mediator for Ang II-induced pathophysiological actions. However, using C66 and specific inhibitors as small molecule probes, in vitro experiments demonstrate that the MAPK signalling pathway regulates ACE expression under HG stimulation, which contributes to renal Ang II activation and the development of DN. This study indicates that C66 is a potential candidate of DN therapeutic agents, and more importantly, that reduction in ACE expression by MAPKs inhibition seems to be an alternative strategy for the treatment of DN.
Collapse
Affiliation(s)
- Yong Pan
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Peritoneal fibrosis and the putative role of decorin. Int J Organ Transplant Med 2013. [DOI: 10.1016/j.hkjn.2013.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
29
|
Inflammation, neoangiogenesis and fibrosis in peritoneal dialysis. Clin Chim Acta 2013; 421:46-50. [DOI: 10.1016/j.cca.2013.02.027] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 02/14/2013] [Accepted: 02/19/2013] [Indexed: 11/22/2022]
|
30
|
|
31
|
Morales MG, Vazquez Y, Acuña MJ, Rivera JC, Simon F, Salas JD, Alvarez Ruf J, Brandan E, Cabello-Verrugio C. Angiotensin II-induced pro-fibrotic effects require p38MAPK activity and transforming growth factor beta 1 expression in skeletal muscle cells. Int J Biochem Cell Biol 2012; 44:1993-2002. [PMID: 22964022 DOI: 10.1016/j.biocel.2012.07.028] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 10/28/2022]
Abstract
Fibrotic disorders are typically characterised by excessive connective tissue and extracellular matrix (ECM) deposition that preclude the normal healing of different tissues. Several skeletal muscle dystrophies are characterised by extensive fibrosis. Among the factors involved in skeletal muscle fibrosis is angiotensin II (Ang-II), a key protein of the renin-angiotensin system (RAS). We previously demonstrated that myoblasts responded to Ang-II by increasing the ECM protein levels mediated by AT-1 receptors, implicating an Ang-II-induced reactive oxygen species (ROS) by a NAD(P)H oxidase-dependent mechanism. In this paper, we show that in myoblasts, Ang-II induced the increase of transforming growth factor beta 1 (TGF-β1) and connective tissue growth factor (CTGF) expression through its AT-1 receptor. This effect is dependent of the NAD(P)H oxidase (NOX)-induced ROS, as indicated by a decrease of the expression of both pro-fibrotic factors when the ROS production was inhibited via the NOX inhibitor apocynin. The increase in pro-fibrotic factors levels was paralleled by enhanced p38MAPK and ERK1/2 phosphorylation in response to Ang-II. However, only the p38MAPK activity was critical for the Ang-II-induced fibrotic effects, as indicated by the decrease in the Ang-II-induced TGF-β1 and CTGF expression and fibronectin levels by SB-203580, an inhibitor of the p38MAPK, but not by U0126, an inhibitor of ERK1/2 phosphorylation. Furthermore, we showed that the Ang-II-dependent p38MAPK activation, but not the ERK1/2 phosphorylation, was necessary for the NOX-derived ROS. In addition, we demonstrated that TGF-β1 expression was required for the Ang-II-induced pro-fibrotic effects evaluated by using SB-431542, an inhibitor of TGF-βRI kinase activity, and by knocking down TGF-β1 levels by shRNA technique. These results strongly suggest that the fibrotic response to Ang-II is mediated by the AT-1 receptor and requires the p38MAPK phosphorylation, NOX-induced ROS, and TGF-β1 expression increase mediated by Ang-II in skeletal muscle cells.
Collapse
Affiliation(s)
- María Gabriela Morales
- Centro de Regulación Celular y Patología, Centro de Regeneración y Envejecimiento, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Functional deterioration of the peritoneal membrane in patients on peritoneal dialysis has been described as being the result of a combination of neoangiogenesis and fibrosis. Glucose, glucose degradation products, and the unphysiological pH of the dialysate solution contribute to these changes. Although newer solutions clearly perform better in terms of their biocompatibility in an in vitro setting and in animal models, the benefit of such solutions over older solutions in the clinical setting is so far unproven. The difficulties in showing a benefit of the newer, more biocompatible solutions in the clinical setting can be explained by the fact that other factors also affect the properties of the peritoneal membrane. These factors are often neglected in clinical studies, which results in unnoticed differences in case-mix and blurs the potential impact of the novel solutions. However, many of these factors are modifiable, and attention should be paid to them in clinical practice to maintain the integrity of the peritoneal membrane. This Review focuses on factors that potentially influence the integrity of the peritoneal membrane, other than those associated with the peritoneal dialysis fluid itself.
Collapse
|
33
|
Pathophysiological changes to the peritoneal membrane during PD-related peritonitis: the role of mesothelial cells. Mediators Inflamm 2012; 2012:484167. [PMID: 22577250 PMCID: PMC3337720 DOI: 10.1155/2012/484167] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 01/18/2012] [Accepted: 01/18/2012] [Indexed: 01/08/2023] Open
Abstract
The success of peritoneal dialysis (PD) is dependent on the structural and functional integrity of the
peritoneal membrane. The mesothelium lines the peritoneal membrane and is the first line of
defense against chemical and/or bacterial insult. Peritonitis remains a major complication of PD and
is a predominant cause of technique failure, morbidity and mortality amongst PD patients. With
appropriate antibiotic treatment, peritonitis resolves without further complications, but in some PD
patients excessive peritoneal inflammatory responses lead to mesothelial cell exfoliation and
thickening of the submesothelium, resulting in peritoneal fibrosis and sclerosis. The detrimental
changes in the peritoneal membrane structure and function correlate with the number and severity
of peritonitis episodes and the need for catheter removal. There is evidence that despite clinical
resolution of peritonitis, increased levels of inflammatory and fibrotic mediators may persist in the
peritoneal cavity, signifying persistent injury to the mesothelial cells. This review will describe the
structural and functional changes that occur in the peritoneal membrane during peritonitis and how
mesothelial cells contribute to these changes and respond to infection. The latter part of the review
discusses the potential of mesothelial cell transplantation and genetic manipulation in the
preservation of the peritoneal membrane.
Collapse
|
34
|
Lakshmanan AP, Thandavarayan RA, Watanabe K, Sari FR, Meilei H, Giridharan VV, Sukumaran V, Soetikno V, Arumugam S, Suzuki K, Kodama M. Modulation of AT-1R/MAPK cascade by an olmesartan treatment attenuates diabetic nephropathy in streptozotocin-induced diabetic mice. Mol Cell Endocrinol 2012; 348:104-11. [PMID: 21827824 DOI: 10.1016/j.mce.2011.07.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 07/20/2011] [Accepted: 07/22/2011] [Indexed: 10/17/2022]
Abstract
There is increasing evidence that angiotensin (Ang)-II plays an unprecedented role in diabetic complications. It could also be an important therapeutic target for ameliorating various diseases, especially diabetic nephropathy (DN). We therefore studied the beneficial effects of olmesartan, an Ang-II type 1 receptor (AT-1R) blocker in streptozotocin (150 mg/kg, BW)-induced diabetic kidney disease in mice. The diabetic kidney mice displayed upregulated protein expression levels of AT-1R, AT-2R, ERK-1/2, p-p38 MAPK, p-MAPKAPK-2, ET-1, p-JNK, p-c-Jun, TGF-β1, and gp91-phox, and all of these effects were expectedly downregulated by an olmesartan treatment. Also, immunohistochemical analysis, and Azan-Mallory and HE staining were performed to examine the expression of collagen-III and fibronectin, renal fibrosis, and hypertrophy, respectively. Furthermore, olmesartan treatment significantly abrogated the downregulation of ACE-2 and Ang-(1-7) mas R protein expression in diabetic kidney mice. Considering all these findings together, the AT-1R/MAPK pathway might be a potential therapeutic target in diabetes kidney disease, and olmesartan treatment could have beneficial effects on DN by modulating the AT-1R/MAPK pathway.
Collapse
Affiliation(s)
- Arun Prasath Lakshmanan
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences, Niigata City 956-8603, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Tomino Y. Mechanisms and interventions in peritoneal fibrosis. Clin Exp Nephrol 2011; 16:109-14. [PMID: 21935614 DOI: 10.1007/s10157-011-0533-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 08/22/2011] [Indexed: 11/25/2022]
Abstract
Peritoneal dialysis (PD) is an attractive treatment for patients with end-stage kidney disease (ESKD). However, long-term peritoneal dialysis is associated with development of functional and structural alterations of the peritoneal membrane. Several factors are implicated in the development of peritoneal fibrosis in PD patients. Inflammatory cytokines, which are induced in the peritoneal cavity during peritonitis, may also induce chronic inflammation and fibrosis. Transforming growth factor β1 (TGF-β1) is generally considered to play an important role in peritoneal fibrosis. The objective of this review is to summarize the mechanisms of peritoneal fibrosis using in vitro and in vivo studies, and the current status and future prospects of interventions in the peritoneal fibrosis.
Collapse
Affiliation(s)
- Yasuhiko Tomino
- Division of Nephrology, Department of Internal Medicine, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
36
|
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: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
37
|
Aguirre AR, Abensur H. Protective measures against ultrafiltration failure in peritoneal dialysis patients. Clinics (Sao Paulo) 2011; 66:2151-7. [PMID: 22189743 PMCID: PMC3226613 DOI: 10.1590/s1807-59322011001200023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 08/18/2011] [Indexed: 01/17/2023] Open
Abstract
Ultrafiltration failure in patients undergoing peritoneal dialysis is a condition with an incidence that increases over time. It is related to increased cardiovascular morbidity and mortality and is a major cause of the abandonment of the treatment technique. Because the number of patients undergoing renal replacement therapy is increasing with society aging and because approximately 10% of this population is treated with peritoneal dialysis, this matter is becoming more common in everyday practice for clinicians involved in the care of patients with chronic renal failure. In this review, we summarize the available measures used to prevent and treat ultrafiltration failure and the current state of research in the field, both in the experimental and clinical settings, focusing on the possible clinical applications of recent findings.
Collapse
Affiliation(s)
- Anna Rita Aguirre
- Hospital das Clínicas, Universidade de São Paulo, Nephrology Division, São Paulo, SP, Brazil.
| | | |
Collapse
|
38
|
Ward BC, Panitch A. Abdominal Adhesions: Current and Novel Therapies. J Surg Res 2011; 165:91-111. [DOI: 10.1016/j.jss.2009.09.015] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 08/17/2009] [Accepted: 09/04/2009] [Indexed: 12/20/2022]
|
39
|
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: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
40
|
Alonso F, Krattinger N, Mazzolai L, Simon A, Waeber G, Meda P, Haefliger JA. An angiotensin II- and NF-kappaB-dependent mechanism increases connexin 43 in murine arteries targeted by renin-dependent hypertension. Cardiovasc Res 2010; 87:166-76. [PMID: 20110337 PMCID: PMC2883896 DOI: 10.1093/cvr/cvq031] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 01/20/2010] [Accepted: 01/25/2010] [Indexed: 01/17/2023] Open
Abstract
AIMS Connexins (Cxs) play a role in the contractility of the aorta wall. We investigated how connexins of the endothelial cells (ECs; Cx37, Cx40) and smooth muscle cells (SMCs; Cx43, Cx45) of the aorta change during renin-dependent and -independent hypertension. METHODS AND RESULTS We subjected both wild-type (WT) mice and mice lacking Cx40 (Cx40(-/-)), to either a two-kidney, one-clip procedure or to N-nitro-l-arginine-methyl-ester treatment, which induce renin-dependent and -independent hypertension, respectively. All hypertensive mice featured a thickened aortic wall, increased levels of Cx37 and Cx45 in SMC, and of Cx40 in EC (except in Cx40(-/-) mice). Cx43 was up-regulated, with no effect on its S368 phosphorylation, only in the SMCs of renin-dependent models of hypertension. Blockade of the renin-angiotensin system of Cx40(-/-) mice normalized blood pressure and prevented both aortic thickening and Cx alterations. Ex vivo exposure of WT aortas, carotids, and mesenteric arteries to physiologically relevant levels of angiotensin II (AngII) increased the levels of Cx43, but not of other Cx. In the aortic SMC line of A7r5 cells, AngII activated kinase-dependent pathways and induced binding of the nuclear factor-kappa B (NF-kappaB) to the Cx43 gene promoter, increasing Cx43 expression. CONCLUSION In both large and small arteries, hypertension differently regulates Cx expression in SMC and EC layers. Cx43 is selectively increased in renin-dependent hypertension via an AngII activation of the extracellular signal-regulated kinase and NF-kappaB pathways.
Collapse
MESH Headings
- Angiotensin II/metabolism
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Angiotensin-Converting Enzyme Inhibitors/pharmacology
- Animals
- Antihypertensive Agents/pharmacology
- Aorta/drug effects
- Aorta/metabolism
- Aorta/physiopathology
- Binding Sites
- Blood Pressure
- Carotid Arteries/metabolism
- Carotid Arteries/physiopathology
- Cell Line
- Connexin 43/genetics
- Connexin 43/metabolism
- Connexins/deficiency
- Connexins/genetics
- Disease Models, Animal
- Endothelial Cells/metabolism
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Genes, Reporter
- Hypertension, Renovascular/drug therapy
- Hypertension, Renovascular/etiology
- Hypertension, Renovascular/metabolism
- Hypertension, Renovascular/physiopathology
- Mesenteric Arteries/metabolism
- Mesenteric Arteries/physiopathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Smooth Muscle/metabolism
- NF-kappa B/metabolism
- NG-Nitroarginine Methyl Ester
- Nephrectomy
- Phosphorylation
- Promoter Regions, Genetic
- Rats
- Renin/blood
- Time Factors
- Transfection
- Up-Regulation
- Gap Junction alpha-5 Protein
Collapse
Affiliation(s)
- Florian Alonso
- Service of Internal Medicine, Laboratory of Experimental Medicine 19-135S, University Hospital, CHUV-1011 Lausanne, Switzerland
| | - Nathalie Krattinger
- Service of Internal Medicine, Laboratory of Experimental Medicine 19-135S, University Hospital, CHUV-1011 Lausanne, Switzerland
| | - Lucia Mazzolai
- Service of Internal Medicine, Laboratory of Experimental Medicine 19-135S, University Hospital, CHUV-1011 Lausanne, Switzerland
| | - Alexander Simon
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Gérard Waeber
- Service of Internal Medicine, Laboratory of Experimental Medicine 19-135S, University Hospital, CHUV-1011 Lausanne, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva, School of Medicine, CMU, 1211 Genève 4, Switzerland
| | - Jacques-Antoine Haefliger
- Service of Internal Medicine, Laboratory of Experimental Medicine 19-135S, University Hospital, CHUV-1011 Lausanne, Switzerland
| |
Collapse
|
41
|
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: 2.0] [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
|
42
|
Yokoyama Y, Masaki T, Kiribayashi K, Nakashima A, Kokoroishi K, Ogawa T, Kohno N, Yorioka N. 15-Deoxy-Delta12,14-prostaglandin J2 inhibits angiotensin II-induced fibronectin expression via hepatocyte growth factor induction in human peritoneal mesothelial cells. Ther Apher Dial 2010; 14:43-51. [PMID: 20438519 DOI: 10.1111/j.1744-9987.2009.00702.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
15-Deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)) is an endogenous peroxisome proliferator-activated receptor gamma (PPARgamma) agonist that suppresses progressive matrix deposition; however, little is known about the effects of 15d-PGJ(2) on human peritoneal mesothelial cells (HPMCs). We investigated the following: (i) the expression of PPARgamma; (ii) the effect of 15d-PGJ(2) on angiotensin II (Ang II)-induced fibronectin (FN) expression and secretion; (iii) the effect of 15d-PGJ(2) (with or without Ang II and with or without the specific PPARgamma antagonist GW9662) and pioglitazone, a synthetic PPARgamma agonist, on hepatocyte growth factor (HGF) expression and secretion; (iv) the effect of HGF on Ang II-induced FN expression and secretion; (v) the expression of c-Met (a specific HGF receptor) and its phospho-signal; and (vi) the involvement of HGF in the effect produced by 15d-PGJ(2) using selective c-Met inhibitor PHA-665752. The presence of PPARgamma was detected by western blot analysis. 15d-PGJ(2) inhibited Ang II-induced FN expression and increased HGF expression, even in the presence of Ang II. This effect of HGF expression was completely prevented by co-treatment with GW9662. Additionally, upregulation of HGF secretion induced by 15d-PGJ(2) and HGF production induced by pioglitazone was revealed. We demonstrated the presence of c-Met, and presented evidence that HGF inhibits Ang II-induced FN expression and activates phosphorylation of c-Met, which is blocked by PHA-665752; 15d-PGJ(2) also activated c-Met phosphorylation. Furthermore, PHA-665752 attenuates the inhibitory effects of 15d-PGJ(2) on FN secretion. These findings suggest that 15d-PGJ(2) has a novel and potent antifibrotic effect in HPMC and this action is likely mediated by HGF.
Collapse
Affiliation(s)
- Yukio Yokoyama
- Department of Molecular and Internal Medicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
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: 1.8] [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
|
44
|
Angiotensin-(1–7) enhances angiotensin II induced phosphorylation of ERK1/2 in mouse bone marrow-derived dendritic cells. Mol Immunol 2009; 46:355-61. [DOI: 10.1016/j.molimm.2008.10.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Revised: 09/30/2008] [Accepted: 10/22/2008] [Indexed: 11/20/2022]
|
45
|
Kawai T, Masaki T, Doi S, Arakawa T, Yokoyama Y, Doi T, Kohno N, Yorioka N. PPAR-gamma agonist attenuates renal interstitial fibrosis and inflammation through reduction of TGF-beta. J Transl Med 2009; 89:47-58. [PMID: 19002105 DOI: 10.1038/labinvest.2008.104] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Thiazolidinediones (TZDs), synthetic peroxisome proliferator-activated receptor (PPAR)-gamma ligands, have a central role in insulin sensitization and adipogenesis. It has been reported that TZDs exert protective effects in both diabetic and nondiabetic models of renal disease, although the exact mechanism is not well understood. In particular, only a few studies have reported the renoprotective effects of TZDs in nondiabetic models of tubulointerstitial fibrosis and inflammation. Therefore, we investigated the anti-fibrotic and anti-inflammatory effects of the TZD troglitazone in the mouse model of unilateral ureteral obstruction (UUO). C57BL/6J mice underwent UUO and were studied after 3 and 7 days. Animals were divided into three groups and received control vehicle, troglitazone (150 mg/kg per day) or troglitazone (300 mg/kg per day) by gavage. Kidneys were harvested for morphological, mRNA and protein analysis. Reverse-transcriptase-PCR was used to assess the expression of transforming growth factor-beta1 (TGF-beta1) and the TGF-beta1 type I receptor (TGF beta R-I). Protein expression was assessed by western blotting (TGF beta R-I) and immunostaining (TGF beta R-I, alpha-smooth muscle actin (alpha-SMA), type I collagen (collagen I), F4/80, and proliferating cell nuclear antigen (PCNA)). The expression of alpha-SMA, collagen I, and F4/80 was decreased in mice treated with troglitazone compared with the control group. The numbers of PCNA-positive interstitial cells were decreased in mice treated with troglitazone. TGF-beta1 mRNA and TGF beta R-I mRNA and protein expression were decreased in the group treated with troglitazone compared with the control group. The beneficial effects of troglitazone treatment were also dose dependent. PPAR-gamma agonist significantly reduced TGF-beta and attenuated renal interstitial fibrosis and inflammation in the model of UUO.
Collapse
Affiliation(s)
- Toru Kawai
- Department of Molecular and Internal Medicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Tang J, Zhan C, Zhou J. Effects of tanshinone IIA on transforming growth factor beta1-Smads signal pathway in renal interstitial fibroblasts of rats. ACTA ACUST UNITED AC 2008; 28:539-42. [PMID: 18846334 DOI: 10.1007/s11596-008-0511-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2008] [Indexed: 11/25/2022]
Abstract
The effects of tanshinone IIA (TSN) on transforming growth factor beta1 (TGFbeta1) signal transduction in renal interstitial fibroblasts of rats were studied in order to investigate its mechanism in prevention of renal interstitial fibrosis. Rat renal fibroblasts of the line NRK/49F were cultured in vitro, stimulated with 5 ng/mL TGFbeta1 and pretreated with 10(-6), 10(-5), 10(-4) mol/L TSN respectively. The mRNA levels of fibronectin (FN) were examined by RT-PCR. The protein expression of FN and Smads was detected by Western blot. TGFbeta1 induced the expression of FN mRNA and Smads in a time-dependent manner in a certain range. Compared with pre-stimulation, the FN mRNA and protein levels were increased by 1.1 times and 1.5 times respectively (P<0.01, P<0.01), and the protein expression of phosphorylated Smad2/3 (p-Smad2/3) increased by 7 times at the end of TGFbeta1 stimulation (P<0.01). TSN pretreatment may down-regulate the FN and p-Smad2/3 expression in a dose-dependent manner. 10(-6) mol/L TSN pretreatment had no effect on the FN and p-Smad2/3 expression (both P>0.05). After pretreatment with 10(-5) and 10(-4) mol/L TSN, the FN mRNA levels were decreased by 28.1% and 43.8% respectively (P<0.05, P<0.01), the FN protein levels were decreased by 40% and 44% respectively (P<0.05, P<0.05), and the p-Smad2/3 protein expression were decreased by 40% and 65% respectively (P<0.05, P<0.01). The inhibitory effect of TSN on renal interstitial fibrosis may be related to its blocking effect on TGFbeta1-Smads signal pathway in renal interstitial fibroblasts.
Collapse
Affiliation(s)
- Jinhui Tang
- Department of Pediatrics, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | | | | |
Collapse
|
47
|
Ito T, Yorioka N. Peritoneal damage by peritoneal dialysis solutions. Clin Exp Nephrol 2008; 12:243-249. [PMID: 18274701 DOI: 10.1007/s10157-008-0032-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Accepted: 12/09/2007] [Indexed: 11/26/2022]
Abstract
Continuous ambulatory peritoneal dialysis is a well-accepted treatment for end-stage renal disease, but its long-term success is limited. Peritoneal sclerosis is still one of the most important complications of long-term peritoneal dialysis and the low biocompatibility of peritoneal dialysis solutions plays a major role in the development of such sclerosis. In this review, we summarize recent experimental data about the biocompatibility of peritoneal dialysis solutions.
Collapse
Affiliation(s)
- Takafumi Ito
- Section of Nephrology, Department of Internal Medicine, Faculty of Medicine, Shimane University, 85-1, Enya-cho, Izumo, 693-8501, Japan
| | - Noriaki Yorioka
- Department of Advanced Nephrology, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| |
Collapse
|
48
|
Doi S, Masaki T, Arakawa T, Takahashi S, Kawai T, Nakashima A, Naito T, Kohno N, Yorioka N. Protective effects of peroxisome proliferator-activated receptor gamma ligand on apoptosis and hepatocyte growth factor induction in renal ischemia-reperfusion injury. Transplantation 2007; 84:207-213. [PMID: 17667812 DOI: 10.1097/01.tp.0000269614.21367.3f] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Renal ischemia-reperfusion injury affects the long-term outcome of renal graft survival. Thiazolidinediones (TZDs), synthetic peroxisome proliferator-activated receptor (PPAR)-gamma ligands, have been shown to exert therapeutic effects upon renal ischemia-reperfusion injury far beyond their use as insulin sensitizers. It has also been reported that hepatocyte growth factor (HGF) has a beneficial effect on renal ischemia-reperfusion injury and that TZDs induce increased HGF mRNA expression and protein secretion. We investigated the effect of troglitazone, one of the TZDs, in a rat model of renal ischemia-reperfusion injury. METHODS A 45-minute period of warm renal ischemia was induced by bilateral clamping at 37 degrees C with rats being sacrificed before the onset of ischemia and at 2, 4, 6, and 12 hr after reperfusion. The expression of PPAR-gamma was measured by reverse-transcriptase polymerase chain reaction (RT-PCR) and western blotting while the production of HGF was investigated by RT-PCR and immunohistochemistry. The effect of troglitazone treatment on the level of apoptosis was determined by staining for cleaved caspase-3 and single-stranded DNA (ssDNA). RESULTS The numbers of cleaved caspase-3 and ssDNA positive cells were decreased in rats treated with troglitazone. The production of HGF mRNA and protein was most intense at 4 hr. The expression of PPAR-gamma and HGF was increased in the group treated with troglitazone compared with the control group. CONCLUSIONS.: Pretreatment of rats with the PPAR-gamma ligand troglitazone decreased apoptotic cell death in renal ischemia-reperfusion injury as a result of the induction of HGF.
Collapse
Affiliation(s)
- Shigehiro Doi
- Department of Molecular and Internal Medicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Aroeira LS, Aguilera A, Sánchez-Tomero JA, Bajo MA, del Peso G, Jiménez-Heffernan JA, Selgas R, López-Cabrera M. Epithelial to Mesenchymal Transition and Peritoneal Membrane Failure in Peritoneal Dialysis Patients: Pathologic Significance and Potential Therapeutic Interventions. J Am Soc Nephrol 2007; 18:2004-13. [PMID: 17568021 DOI: 10.1681/asn.2006111292] [Citation(s) in RCA: 260] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Peritoneal dialysis (PD) is a form of renal replacement and is based on the use of the peritoneum as a semipermeable membrane across which ultrafiltration and diffusion take place. Nevertheless, continuous exposure to bioincompatible PD solutions and episodes of peritonitis or hemoperitoneum cause acute and chronic inflammation and injury to the peritoneal membrane, which progressively undergoes fibrosis and angiogenesis and, ultimately, ultrafiltration failure. The pathophysiologic mechanisms that are involved in peritoneal functional impairment have remained elusive. Resident fibroblasts and infiltrating inflammatory cells have been considered the main entities that are responsible for structural and functional alterations of the peritoneum. Recent findings, however, demonstrated that new fibroblastic cells may arise from local conversion of mesothelial cells (MC) by epithelial-to-mesenchymal transition (EMT) during the inflammatory and repair responses that are induced by PD and pointed to MC as protagonists of peritoneal membrane deterioration. Submesothelial myofibroblasts, which participate in inflammatory responses, extracellular matrix accumulation, and angiogenesis, can originate from activated resident fibroblasts and from MC through EMT. This heterogeneous origin of myofibroblasts reveals new pathogenic mechanisms and offers novel therapeutic possibilities. This article provides a comprehensive review of recent advances on understanding the mechanisms that are implicated in peritoneal structural alterations, which have allowed the identification of the EMT of MC as a potential therapeutic target of membrane failure.
Collapse
Affiliation(s)
- Luiz S Aroeira
- Unidad de Biología Molecular, Hospital Universitario de la Princesa, C/ Diego de León 62, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Katsutani M, Ito T, Masaki T, Kohno N, Yorioka N. Glucose-based PD solution, but not icodextrin-based PD solution, induces plasminogen activator inhibitor-1 and tissue-type plasminogen activator in human peritoneal mesothelial cells via ERK1/2. Ther Apher Dial 2007; 11:94-100. [PMID: 17381529 DOI: 10.1111/j.1744-9987.2007.00423.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Peritoneal dialysis (PD) solutions containing glucose are considered to cause peritoneal fibrosis. Plasminogen activator inhibitor-1 (PAI-1) and tissue-type plasminogen activator (t-PA) participate in fibrogenesis of various organs, and human peritoneal mesothelial cells (HPMC) can produce PAI-1 and t-PA following glucose stimulation. Icodextrin has been widely used as an alternative osmotic agent. In this study, we investigated whether icodextrin-based PD solution reduced the production of PAI-1 and t-PA by HPMC. We also examined the involvement of extracellular signal-regulated kinase 1/2 (ERK1/2). Glucose-based PD solutions increased the production of PAI-1 and t-PA by HPMC, whereas icodextrin-based PD solution exerted lesser effects. Glucose-based PD solutions activated ERK1/2, and PD98059 inhibited the production of PAI-1 and t-PA-responses not observed with icodextrin-based PD solution. In conclusion, glucose-based PD solutions, unlike icodextrin-based PD solution, induce overproduction of PAI-1 and t-PA via the ERK1/2 pathway.
Collapse
Affiliation(s)
- Masahira Katsutani
- Department of Molecular and Internal Medicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | |
Collapse
|