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
|
Kocurkova A, Kerberova M, Nesporova K, Lehka K, Sandanusova M, Simek M, Velebny V, Kubala L, Ambrozova G. Endogenously produced hyaluronan contributes to the regulation of peritoneal adhesion development. Biofactors 2023; 49:940-955. [PMID: 37154260 DOI: 10.1002/biof.1957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/05/2023] [Indexed: 05/10/2023]
Abstract
Peritoneal adhesions are postsurgical fibrotic complications connected to peritoneal inflammation. The exact mechanism of development is unknown; however, an important role is attributed to activated mesothelial cells (MCs) overproducing macromolecules of extracellular matrix (ECM), including hyaluronic acid (HA). It was suggested that endogenously-produced HA contributes to the regulation of different fibrosis-related pathologies. However, little is known about the role of altered HA production in peritoneal fibrosis. We focused on the consequences of the increased turnover of HA in the murine model of peritoneal adhesions. Changes of HA metabolism were observed in early phases of peritoneal adhesion development in vivo. To study the mechanism, human MCs MeT-5A and murine MCs isolated from the peritoneum of healthy mice were pro-fibrotically activated by transforming growth factor β (TGFβ), and the production of HA was attenuated by two modulators of carbohydrate metabolism, 4-methylumbelliferone (4-MU) and 2-deoxyglucose (2-DG). The attenuation of HA production was mediated by upregulation of HAS2 and downregulation of HYAL2 and connected to the lower expression of pro-fibrotic markers, including fibronectin and α-smooth muscle actin (αSMA). Moreover, the inclination of MCs to form fibrotic clusters was also downregulated, particularly in 2-DG-treated cells. The effects of 2-DG, but not 4-MU, were connected to changes in cellular metabolism. Importantly, the inhibition of AKT phosphorylation was observed after the use of both HA production inhibitors. In summary, we identified endogenous HA as an important regulator of peritoneal fibrosis, not just a passive player during this pathological process.
Collapse
Affiliation(s)
- Anna Kocurkova
- Department of Experimental Biology, Faculty of Sciences, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Michaela Kerberova
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | | | | | - Miriam Sandanusova
- Department of Experimental Biology, Faculty of Sciences, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Matej Simek
- Contipro a.s., Dolni Dobrouc, Czech Republic
| | | | - Lukas Kubala
- Department of Experimental Biology, Faculty of Sciences, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Gabriela Ambrozova
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| |
Collapse
|
3
|
Kim I, Seo J, Lee DH, Kim YH, Kim JH, Wie MB, Byun JK, Yun JH. Ulmus davidiana 60% edible ethanolic extract for prevention of pericyte apoptosis in diabetic retinopathy. Front Endocrinol (Lausanne) 2023; 14:1138676. [PMID: 37234799 PMCID: PMC10206296 DOI: 10.3389/fendo.2023.1138676] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Diabetic retinopathy (DR) is a disease that causes visual deficiency owing to vascular leakage or abnormal angiogenesis. Pericyte apoptosis is considered one of the main causes of vascular leakage in diabetic retina, but there are few known therapeutic agents that prevent it. Ulmus davidiana is a safe natural product that has been used in traditional medicine and is attracting attention as a potential treatment for various diseases, but its effect on pericyte loss or vascular leakage in DR is not known at all. In the present study, we investigated on the effects of 60% edible ethanolic extract of U. davidiana (U60E) and catechin 7-O-β-D-apiofuranoside (C7A), a compound of U. davidiana, on pericyte survival and endothelial permeability. U60E and C7A prevented pericyte apoptosis by inhibiting the activation of p38 and JNK induced by increased glucose and tumor necrosis factor alpha (TNF-α) levels in diabetic retina. Moreover, U60E and C7A reduced endothelial permeability by preventing pericyte apoptosis in co-cultures of pericytes and endothelial cells. These results suggest that U60E and C7A could be a potential therapeutic agent for reducing vascular leakage by preventing pericyte apoptosis in DR.
Collapse
Affiliation(s)
- Iljin Kim
- Department of Pharmacology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Jieun Seo
- Faculty of Engineering, Yokohama National University, Yokohama, Japan
| | - Dong Hyun Lee
- Department of Ophthalmology, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| | - Yo-Han Kim
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jun-Hyung Kim
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Myung-Bok Wie
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jun-Kyu Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Jang-Hyuk Yun
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| |
Collapse
|
4
|
González-Herrera F, Anfossi R, Catalán M, Gutiérrez-Figueroa R, Maya JD, Díaz-Araya G, Vivar R. Lipoxin A4 prevents high glucose-induced inflammatory response in cardiac fibroblast through FOXO1 inhibition. Cell Signal 2023; 106:110657. [PMID: 36933776 DOI: 10.1016/j.cellsig.2023.110657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
Cardiac cells respond to various pathophysiological stimuli, synthesizing inflammatory molecules that allow tissue repair and proper functioning of the heart; however, perpetuation of the inflammatory response can lead to cardiac fibrosis and heart dysfunction. High concentration of glucose (HG) induces an inflammatory and fibrotic response in the heart. Cardiac fibroblasts (CFs) are resident cells of the heart that respond to deleterious stimuli, increasing the synthesis and secretion of both fibrotic and proinflammatory molecules. The molecular mechanisms that regulate inflammation in CFs are unknown, thus, it is important to find new targets that allow improving treatments for HG-induced cardiac dysfunction. NFκB is the master regulator of inflammation, while FoxO1 is a new participant in the inflammatory response, including inflammation induced by HG; however, its role in the inflammatory response of CFs is unknown. The inflammation resolution is essential for an effective tissue repair and recovery of the organ function. Lipoxin A4 (LXA4) is an anti-inflammatory agent with cytoprotective effects, while its cardioprotective effects have not been fully studied. Thus, in this study, we analyze the role of p65/NFκB, and FoxO1 in CFs inflammation induced by HG, evaluating the anti-inflammatory properties of LXA4. Our results demonstrated that HG induces the inflammatory response in CFs, using an in vitro and ex vivo model, while FoxO1 inhibition and silencing prevented HG effects. Additionally, LXA4 inhibited the activation of FoxO1 and p65/NFκB, and inflammation of CFs induced by HG. Therefore, our results suggest that FoxO1 and LXA4 could be novel drug targets for the treatment of HG-induced inflammatory and fibrotic disorders in the heart.
Collapse
Affiliation(s)
- Fabiola González-Herrera
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Renatto Anfossi
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Mabel Catalán
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Renata Gutiérrez-Figueroa
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juan Diego Maya
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Guillermo Díaz-Araya
- Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Raúl Vivar
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
5
|
Hung H, Tsai S, Sie S, Kuo Y. High glucose enhances lipopolysaccharide‐induced inflammation in cultured BV2 microglial cell line. Immun Inflamm Dis 2022; 10:e610. [PMID: 35478445 PMCID: PMC9017628 DOI: 10.1002/iid3.610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/08/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- Hao‐Chang Hung
- Division of Endocrinology and Metabolism, Department of Internal Medicine Kaohsiung Veterans General Hospital Kaohsiung Taiwan
| | - Sheng‐Feng Tsai
- Institute of Basic Medical Sciences, College of Medicine National Cheng Kung University Tainan Taiwan
- Department of Cell Biology and Anatomy, College of Medicine National Cheng Kung University Tainan Taiwan
| | - Shih‐Ren Sie
- Department of Anesthesiology Ditmanson Medical Foundation Chia‐Yi Christian Hospital Chiayi Taiwan
| | - Yu‐Min Kuo
- Institute of Basic Medical Sciences, College of Medicine National Cheng Kung University Tainan Taiwan
- Department of Cell Biology and Anatomy, College of Medicine National Cheng Kung University Tainan Taiwan
| |
Collapse
|
6
|
Huang HW, Yang CM, Yang CH. Fibroblast Growth Factor Type 1 Ameliorates High-Glucose-Induced Oxidative Stress and Neuroinflammation in Retinal Pigment Epithelial Cells and a Streptozotocin-Induced Diabetic Rat Model. Int J Mol Sci 2021; 22:ijms22137233. [PMID: 34281287 PMCID: PMC8267624 DOI: 10.3390/ijms22137233] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/26/2021] [Accepted: 06/30/2021] [Indexed: 01/20/2023] Open
Abstract
Diabetic retinopathy (DR) is a common complication of diabetes that causes severe visual impairment globally. The pathogenesis of DR is related to oxidative stress and chronic inflammation. The fibroblast growth factor type 1 (FGF-1) mitogen plays crucial roles in cell function, development, and metabolism. FGF-1 is involved in blood sugar regulation and exerts beneficial antioxidative and anti-inflammatory effects on various organ systems. This study investigated the antioxidative and anti-inflammatory neuroprotective effects of FGF-1 on high-glucose-induced retinal damage. The results revealed that FGF-1 treatment significantly reversed the harmful effects of oxidative stress and inflammatory mediators in retinal tissue in a streptozotocin-induced diabetic rat model. These protective effects were also observed in the in vitro model of retinal ARPE-19 cells exposed to a high-glucose condition. We demonstrated that FGF-1 attenuated p38 mitogen-activated protein kinase and nuclear factor-κB pathway activation under the high-glucose condition. Our results indicated that FGF-1 could effectively prevent retinal injury in diabetes. The findings of this study could be used to develop novel treatments for DR that aim to reduce the cascade of oxidative stress and inflammatory signals in neuroretinal tissue.
Collapse
Affiliation(s)
- Hsin-Wei Huang
- Department of Ophthalmology, Wan Fang Hospital, Taipei Medical University, No. 111, Sec. 3, Xinglong Rd., Taipei 11696, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, No. 1, Jen Ai Road Sec. 1, Taipei 100, Taiwan
| | - Chung-May Yang
- Department of Ophthalmology, National Taiwan University Hospital, No. 7, Zhongshan South Road, Taipei 100, Taiwan;
- Department of Ophthalmology, College of Medicine, National Taiwan University, No. 1, Jen Ai Road, Sec. 1, Taipei 100, Taiwan
| | - Chang-Hao Yang
- Department of Ophthalmology, National Taiwan University Hospital, No. 7, Zhongshan South Road, Taipei 100, Taiwan;
- Department of Ophthalmology, College of Medicine, National Taiwan University, No. 1, Jen Ai Road, Sec. 1, Taipei 100, Taiwan
- Correspondence: ; Tel.: +886-2-2312-3456 (ext. 62131); Fax: +886-2-2393-4420
| |
Collapse
|
7
|
Abstract
Peritoneal fibrosis (PF) is an important issue in peritoneal dialysis (PD) because it remains one of the leading causes of patient drop-out from PD. In this review, we focus on in vitro approaches to the pathogenesis and therapeutic potential of PF and on associated clinical implications. Representative Asian studies, initiated since mid-1990s, that have investigated matrix accumulation in peritoneal tissue possibly leading to PF in the PD population will be highlighted as examples to learn how to apply this research tool. As compared with data from well-designed clinical trials, observations from in vitro models may be far from becoming solid evidence; however, they do cast new light on options for investigations into therapeutic pharmaceuticals.
Collapse
Affiliation(s)
- Kuan-Yu Hung
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuan-Dun Wu
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tun-Jun Tsai
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
8
|
Abstract
Peritoneal dialysis (PD) solutions using glucose as osmotic agent have been used for more than two decades as effective treatment for patients with end-stage renal disease. Although alternative osmotic agents such as amino acids and macromolecular solutions, including polypeptides and glucose polymers, are now available, glucose is still the most widely used osmotic agent in PD. It has been shown to be safe, effective, readily metabolized, and inexpensive. On the other hand, it is widely assumed that exposure of the peritoneal membrane to high glucose concentrations contributes to both structural and functional changes in the dialyzed peritoneal membrane. As in diabetes, glucose, either directly or indirectly through the generation of glucose degradation products or the formation of advanced glycation end products, may contribute to peritoneal membrane failure. Although efforts to reduce glucose toxicity have been made for years, only a few suggestions, such as dual-bag systems with bicarbonate as buffer system, have found broader acceptance. Recently, some interesting new approaches to the problem of glucose-related toxicity have been made, but further investigations will be necessary before they can be used clinically. This review will focus on adverse effects of glucose in PD solutions and summarize different aspects of glucotoxicity and potential therapeutic interventions.
Collapse
Affiliation(s)
- Thomas Sitter
- Department of Nephrology, Medizinische Poliklinik–Innenstadt, Klinikum der Universität München, Germany
| | - Matthias Sauter
- Department of Nephrology, Medizinische Poliklinik–Innenstadt, Klinikum der Universität München, Germany
| |
Collapse
|
9
|
The Role of Cathepsin B in Peritoneal Fibrosis due to Peritoneal Dialysis. Int J Nephrol 2019; 2019:4150656. [PMID: 31815017 PMCID: PMC6878782 DOI: 10.1155/2019/4150656] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 07/20/2019] [Accepted: 10/26/2019] [Indexed: 11/24/2022] Open
Abstract
Glucose-containing peritoneal dialysis (PD) solution causes peritoneal fibrosis (PF) characterized by accumulation of extracellular matrix (ECM) in the submesothelial layer. Cathepsin B is a lysosomal cysteine protease that degrades ECM, but its role in the PF remains unclear. Thus, we investigated the role of cathepsin B in PF. Procathepsin B was measured in the 73 PD effluents of 68 patients. Procathepsin B and cathepsin B after exposure of glucose and the effects of cathepsin B on the expression of matrix metalloproteinases (MMPs), tissue inhibitor of metalloproteinases (TIMPs), and urokinase-type plasminogen activator (uPA) were measured in the supernatant of cultured human peritoneal mesothelial cells (HPMCs). The effect of cathepsin B and its inhibitor, cystatin C, on PF was investigated in the murine model. Procathepsin B was measured at 3.6 μg/L in serum and 5.4 μg/L in PD effluent and positively correlated to the cancer antigen (CA) 125. The treatment with 4.25% glucose increased procathepsin B by 3.1-fold and cathepsin B by 5.9-fold in the HPMCs. Cathepsin B induced the secretion of MMP-1, -2, and -3 and TIMP-1 in the HPMCs, but uPA was not excreted. In the PF murine models, cathepsin B reduced the thickness of the submesothelial layer and cystatin C attenuated the effect of cathepsin B. HPMCs secrete cathepsin B with exposure of PD solution, and cathepsin B might help protect against PF.
Collapse
|
10
|
Hernández B, Fuentes E, Palomo I, Alarcón M. Increased platelet function during frailty. Exp Hematol 2019; 77:12-25.e2. [DOI: 10.1016/j.exphem.2019.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/24/2019] [Accepted: 08/26/2019] [Indexed: 12/19/2022]
|
11
|
Kuroiwa T, Matsumoto M, Kato R, Nimura A, Yoshii T, Okawa A, Fujita K. Activation of cancer-related and mitogen-activated protein kinase signaling pathways in human mature osteoblasts isolated from patients with type 2 diabetes. Bone Rep 2019; 10:100199. [PMID: 30891471 PMCID: PMC6406057 DOI: 10.1016/j.bonr.2019.100199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/05/2019] [Accepted: 02/20/2019] [Indexed: 01/04/2023] Open
Abstract
Diabetes mellitus is a disease of glucose metabolism, and it adversely affects bone metabolism and increases the risk of cancer development. Previously, we reported a method for the direct isolation of human mature osteoblasts and indicated that osteoblasts were associated with type 2 diabetes mellitus-related signaling pathways. In addition, a recent report suggested that osteoblasts are involved in glucose metabolism. Thus, we sought to examine the effects of diabetes on osteoblast signaling in vivo. We recruited eight patients with type 2 diabetes and eight non-diabetic individuals. We isolated human mature osteoblasts from the resected femoral heads during orthopaedic surgery and extracted their RNA. We compared the gene expression between the two groups by RNA microarray and pathway analyses. Microarray analysis showed significant differences in 885 of 19,463 genes between the two groups (p < 0.05), and pathway analysis revealed that pathways related to cancer and the mitogen-activated protein kinase signaling pathway were significantly activated in the diabetes group (p < 0.01). These preliminary findings suggest that diabetes affects intracellular signaling in human mature osteoblasts and that osteoblasts might not only play a key role in the regulation of bone and glucose metabolism, but might also be related to cancer metabolism. We plan to conduct further studies to examine signaling in diabetic osteoblasts and to further investigate the genes and pathways identified here. Compared microarray data from in vivo DM and healthy control osteoblasts MAPK and cancer-related signaling genes were enriched in DM osteoblasts. DM may increase cancer risk by activating cancer-related pathways in osteoblasts.
Collapse
Affiliation(s)
- Tomoyuki Kuroiwa
- Department of Orthopaedic and Spinal Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Megumi Matsumoto
- Laboratory of Cell and Molecular Bioengineering, Division of Biosciences, Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Room 302, Pharmaceutical Sciences Building Graduate School of Pharmaceutical Sciences, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8601, Japan
| | - Ryuji Kato
- Laboratory of Cell and Molecular Bioengineering, Division of Biosciences, Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Room 302, Pharmaceutical Sciences Building Graduate School of Pharmaceutical Sciences, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8601, Japan
| | - Akimoto Nimura
- Department of Functional Joint Anatomy, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Toshitaka Yoshii
- Department of Orthopaedic and Spinal Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Atsushi Okawa
- Department of Orthopaedic and Spinal Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Koji Fujita
- Department of Orthopaedic and Spinal Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| |
Collapse
|
12
|
Bender TO, Riesenhuber A, Endemann M, Herkner K, Witowski J, Jörres A, Aufricht C. Correlation between HSP-72 Expression and IL-8 Secretion in Human Mesothelial Cells. Int J Artif Organs 2018; 30:199-203. [PMID: 17417758 DOI: 10.1177/039139880703000304] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Cytotoxicity of peritoneal dialysis fluid (PDF) and peritoneal inflammation are currently regarded as the two major culprits for chronic mesothelial injury and peritoneal membrane failure. In this study, we correlated induction of HSP-72, as a marker of the cellular stress response, to secretion of IL-8, as a marker for pro-inflammatory cytokines, in mesothelial cells upon sublethal PDF exposure. Methods Primary omental cell cultures of human mesothelial cells were subjected to sublethal PDF exposure times (CAPD2, Fresenius, Germany). At the end of a 24 hour recovery period, induction of HSP-72 in the cell homogenate and IL-8 secretion in the supernatant was assessed by immunodensitometry and ELISA, respectively. Results PDF exposure times from 15 min to 60 min resulted in progressively increased HSP-72 expression levels (267 vs 320 vs 419% of controls, p<0.05 vs controls) as well as increased IL-8 secretion (323 vs 528 vs 549% of controls, p<0.05 vs controls) with full cell viability (MTT unchanged to control). HSP-72 expression was statistically significantly correlated with IL-8 secretion. Conclusions: The significant correlation between HSP-72 expression and IL-8 secretion suggests that the regulation of pro-inflammatory pathways in mesothelial cells exposed to PDF may represent an integral part of their stress response. Future studies to investigate the cellular regulatory mechanism involved are warranted.
Collapse
Affiliation(s)
- T O Bender
- Department of Nephrology and Medical Intensive Care, Universitätsklinikum Charité, Campus Virchow-Klinikum, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
13
|
Guo Y, Sun L, Xiao L, Gou R, Fang Y, Liang Y, Wang R, Li N, Liu F, Tang L. Aberrant Wnt/Beta-Catenin Pathway Activation in Dialysate-Induced Peritoneal Fibrosis. Front Pharmacol 2017; 8:774. [PMID: 29163160 PMCID: PMC5670149 DOI: 10.3389/fphar.2017.00774] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/13/2017] [Indexed: 01/15/2023] Open
Abstract
Peritoneal dialysis (PD)-associated peritoneal fibrosis is a chronic progress which induces ultrafiltration failure. It remains a challenge to prevent the progression of PD-associated fibrosis in clinic practice. Wnt/β-catenin pathway plays important role in many severe fibrotic diseases, here we investigated its contribution to the development of peritoneal damage. We isolated mesothelial cells (MC) from the effluent of PD patients and found that the expressions of Wnt1, Wnt5a, β-catenin, and LEF1 were increased in patients with more than 1-year PD compared with patients who just started with PD (<1 month). The elevated expressions of Wnts and β-catenin were accompanied with changes in the expressions of E-cadherin, α-SMA, COL-I, and FN mRNA and proteins, which are known related to mesothelial-mesenchymal transition (MMT). In addition, treatment with high glucose significantly increased the expression of Wnt1, Wnt5a, β-catenin, and LEF1 as well as the expression of α-SMA, COL-I, and FN in human peritoneal mesothelial cells (HPMC), whereas the expression of E-cadherin was reduced. Dickkopf-1 (DKK-1) is an endogenous inhibitor of Wnt/β-catenin signaling. Overexpression of DKK1 transgene significantly decreased the expression of β-catenin and attenuated the process of MMT as indicated by the decreased expression of α-SMA, COL-I, and FN and the increased expression of E-cadherin. Furthermore, TGF-β1 treatment significantly activated the Wnt/β-catenin pathway in HPMCs, while DKK1 blocked the TGF-β1-induced Wnt signaling activation and significantly inhibited the process of MMT. These data suggest that the canonical Wnt/β-catenin pathway plays an important role in the MMT and fibrosis induced by PD.
Collapse
Affiliation(s)
- Yuanyuan Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin Sun
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Li Xiao
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Rong Gou
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yudong Fang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Liang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruiqiang Wang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ningjun Li
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, United States
| | - Fuyou Liu
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Lin Tang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
14
|
Kiss K, Baghy K, Spisák S, Szanyi S, Tulassay Z, Zalatnai A, Löhr JM, Jesenofsky R, Kovalszky I, Firneisz G. Chronic hyperglycemia induces trans-differentiation of human pancreatic stellate cells and enhances the malignant molecular communication with human pancreatic cancer cells. PLoS One 2015; 10:e0128059. [PMID: 26010611 PMCID: PMC4444240 DOI: 10.1371/journal.pone.0128059] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/23/2015] [Indexed: 12/12/2022] Open
Abstract
Background Diabetes mellitus is linked to pancreatic cancer. We hypothesized a role for pancreatic stellate cells (PSC) in the hyperglycemia induced deterioration of pancreatic cancer and therefore studied two human cell lines (RLT-PSC, T3M4) in hyperglycemic environment. Methodology/Principal Findings The effect of chronic hyperglycemia (CHG) on PSCs was studied using mRNA expression array with real-time PCR validation and bioinformatic pathway analysis, and confirmatory protein studies. The stress fiber formation (IC: αSMA) indicated that PSCs tend to transdifferentiate to a myofibroblast-like state after exposure to CHG. The phosphorylation of p38 and ERK1/2 was increased with a consecutive upregulation of CDC25, SP1, cFOS and p21, and with downregulation of PPARγ after PSCs were exposed to chronic hyperglycemia. CXCL12 levels increased significantly in PSC supernatant after CHG exposure independently from TGF-β1 treatment (3.09-fold with a 2.73-fold without TGF-β1, p<0.05). The upregualtion of the SP1 transcription factor in PSCs after CHG exposure may be implicated in the increased CXCL12 and IGFBP2 production. In cancer cells, hyperglycemia induced an increased expression of CXCR4, a CXCL12 receptor that was also induced by PSC’s conditioned medium. The receptor-ligand interaction increased the phosphorylation of ERK1/2 and p38 resulting in activation of MAP kinase pathway, one of the most powerful stimuli for cell proliferation. Certainly, conditioned medium of PSC increased pancreatic cancer cell proliferation and this effect could be partially inhibited by a CXCR4 inhibitor. As the PSC conditioned medium (normal glucose concentration) increased the ERK1/2 and p38 phosphorylation, we concluded that PSCs produce other factor(s) that influence(s) pancreatic cancer behaviour. Conclusions Hyperglycemia induces increased CXCL12 production by the PSCs, and its receptor, CXCR4 on cancer cells. The ligand-receptor interaction activates MAP kinase signaling that causes increased cancer cell proliferation and migration.
Collapse
Affiliation(s)
- Katalin Kiss
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Kornélia Baghy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Sándor Spisák
- Children's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Szilárd Szanyi
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
- School of Ph.D. Studies, Semmelweis University, Budapest, Hungary
| | - Zsolt Tulassay
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Attila Zalatnai
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - J.-Matthias Löhr
- Karolinska Institutet, Gastrocentrum, Karolinska University Hospital, Stockholm, Sweden
| | - Ralf Jesenofsky
- University of Heidelberg, Medical Campus Mannheim, Dept. of Medicine II, Mannheim, Germany
| | - Ilona Kovalszky
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gábor Firneisz
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
15
|
Qing Q, Zhang S, Chen Y, Li R, Mao H, Chen Q. High glucose-induced intestinal epithelial barrier damage is aggravated by syndecan-1 destruction and heparanase overexpression. J Cell Mol Med 2015; 19:1366-74. [PMID: 25702768 PMCID: PMC4459850 DOI: 10.1111/jcmm.12523] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 11/27/2014] [Indexed: 02/06/2023] Open
Abstract
Syndecan-1 (Sdc1) and its endo-beta-d-glucuronidase heparanase (HPSE) are implicated in maintenance of intestinal epithelial barrier (IEB), but their alterations and roles in high-glucose/hyperglycaemia (HG) conditions have not been fully investigated. This study aimed to determine the expression pattern, the possible regulation mechanism of Sdc1 and HPSE in HG conditions, and their potential effects on IEB. Therefore, diabetic mice/cell models were developed, and tissue/serum samples, cell lysate and culture supernatants were harvested. The expression of Sdc1 and HPSE in control, HG and designated interventions groups were detected. Phosphorylations of mitogen-activated protein kinase signalling pathway (MAPK), the expressions of Occludin and ZO-1, and the levels of transepithelial electrical resistance (TEER) were measured and monitored. The results showed that in HG conditions, intestinal tissue and cellular Sdc1 were significantly decreased, but the expression of HPSE, and soluble Sdc1 in serum and culture supernatants were remarkably increased. Such alterations of Sdc1 and HPSE were associated with solely p38 MAPK activation, and were correlated with the reductions of Occludin, ZO-1 and TEER. Heparin (Sdc1 analogue) and SB203580 (a p38 MAPK inhibitor), instead of insulin, alleviated Sdc1 destruction and HPSE overexpression, and effectively prevented against the reductions of tight junctions and the abnormality of intestinal permeability in HG conditions. In conclusion, we confirm the unique alterations of Sdc1 and HPSE in HG conditions, and found their interactions with p38 MAPK activation and IEB. These indicate that Sdc1/HPSE modulation can be viewed as an important complementary treatment for relieving HG-induced gastrointestinal damage.
Collapse
Affiliation(s)
- Qing Qing
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shaoheng Zhang
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Chen
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Gastroenterology, Guangzhou, China
| | - Runhua Li
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Gastroenterology, Guangzhou, China
| | - Hua Mao
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qikui Chen
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
16
|
Janus kinase signaling activation mediates peritoneal inflammation and injury in vitro and in vivo in response to dialysate. Kidney Int 2014; 86:1187-96. [PMID: 25007168 DOI: 10.1038/ki.2014.209] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 04/17/2014] [Accepted: 04/24/2014] [Indexed: 11/08/2022]
Abstract
Peritoneal membrane pathology limits long-term peritoneal dialysis (PD). Here, we tested whether JAK/STAT signaling is implicated and if its attenuation might be salutary. In cultured mesothelial cells, PD fluid activated, and the pan-JAK inhibitor P6 reduced, phospho-STAT1 and phospho-STAT3, periostin secretion, and cleaved caspase-3. Ex vivo, JAK was phosphorylated in PD effluent cells from long-term but not new PD patients. MCP-1 and periostin were increased in PD effluent in long term compared with new patients. In rats, twice daily, PD fluid infusion induced phospho-JAK, mesothelial cell hyperplasia, inflammation, fibrosis, and hypervascularity after 10 days of exposure to PD fluid. Concomitant instillation of a JAK1/2 inhibitor virtually completely attenuated these changes. Thus, our studies directly implicate JAK/STAT signaling in the mediation of peritoneal membrane pathology as a consequence of PD.
Collapse
|
17
|
Lu Y, Shen H, Shi X, Feng S, Wang Z, Shi Y. Hydrogen sulfide ameliorates high-glucose toxicity in rat peritoneal mesothelial cells by attenuating oxidative stress. Nephron Clin Pract 2014; 126:157-65. [PMID: 24863338 DOI: 10.1159/000358436] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 12/12/2013] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND/AIMS Continuous exposure of the peritoneal membrane to high-glucose (HG) peritoneal dialysis fluids (PDFs) can produce peritoneal mesothelial cells (PMCs) injury. It has been demonstrated that hydrogen sulfide (H2S), the third endogenous gaseous mediator identified after nitric oxide and carbon monoxide, exhibits a potent protective effect on cell activity. We studied the toxic effects of HG PDFs and their reversal by H2S on cultures of rat PMCs. METHODS Synchronized confluent rat PMCs were incubated with 2.5% glucose PDFs with or without NaHS, an H2S donor. Cell viability was assessed by methyl thiazolyl tetrazolium assay and flow cytometry. The level of phospho-p38 mitogen-activated protein kinase (MAPK) was analyzed by immunoblotting. p53, Bax and Bcl-2 mRNA expressions by rat PMCs were detected by real-time PCR. The levels of reactive oxygen species (ROS), superoxide dismutase (SOD) activity and caspase-3 activity were measured. RESULTS Exposure of rat PMCs to 2.5% glucose PDFs for 24 h resulted in a significant induction of apoptosis, which was attenuated by NaHS. NaHS also restored the 2.5% glucose PDF-induced increase in phospho-p38 MAPK (indices of cellular toxicity). Further investigation of the apoptotic mechanisms in rat PMCs demonstrated that HG activated caspase-3 and upregulated Bax, while it downregulated Bcl-2. All the above responses were prevented by pretreatment with NaHS. Moreover, NaHS reversed the 2.5% glucose PDF-induced increase in ROS generation and decrease in SOD activity. CONCLUSIONS These findings suggest that HG PDFs significantly inhibit rat PMC viability, leading to peritoneal injury. H2S exhibits a potent anti-apoptotic ability by attenuating oxidative stress and inhibiting caspase-3 activation, which in turn restores peritoneal injury.
Collapse
Affiliation(s)
- Ying Lu
- Department of Nephrology, Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | | | | | | | | | | |
Collapse
|
18
|
Pérez-Martínez J, Pérez-Martínez FC, Carrión B, Masiá J, Ortega A, Simarro E, Nam-Cha SH, Ceña V. Aliskiren prevents the toxic effects of peritoneal dialysis fluids during chronic dialysis in rats. PLoS One 2012; 7:e36268. [PMID: 22558414 PMCID: PMC3338692 DOI: 10.1371/journal.pone.0036268] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Accepted: 04/04/2012] [Indexed: 11/26/2022] Open
Abstract
The benefits of long-term peritoneal dialysis (PD) in patients with end-stage renal failure are short-lived due to structural and functional changes in the peritoneal membrane. In this report, we provide evidence for the in vitro and in vivo participation of the renin-angiotensin-aldosterone system (RAAS) in the signaling pathway leading to peritoneal fibrosis during PD. Exposure to high-glucose PD fluids (PDFs) increases damage and fibrosis markers in both isolated rat peritoneal mesothelial cells and in the peritoneum of rats after chronic dialysis. In both cases, the addition of the RAAS inhibitor aliskiren markedly improved damage and fibrosis markers, and prevented functional modifications in the peritoneal transport, as measured by the peritoneal equilibrium test. These data suggest that inhibition of the RAAS may be a novel way to improve the efficacy of PD by preventing inflammation and fibrosis following peritoneal exposure to high-glucose PDFs.
Collapse
Affiliation(s)
- Juan Pérez-Martínez
- Department of Nephrology, Complejo Hospitalario Universitario, Albacete, Spain
| | | | - Blanca Carrión
- Department of Research and Development, NanoDrugs, S.L., Parque Científico y Tecnológico, Albacete, Spain
| | - Jesús Masiá
- Department of Nephrology, Complejo Hospitalario Universitario, Albacete, Spain
| | - Agustín Ortega
- Department of Nephrology, Complejo Hospitalario Universitario, Albacete, Spain
| | - Esther Simarro
- Department of Clinical Chemistry, Complejo Hospitalario Universitario, Albacete, Spain
| | - Syong H. Nam-Cha
- Department of Pathology, Complejo Hospitalario Universitario, Albacete, Spain
| | - Valentín Ceña
- Unidad Asociada Neurodeath, Departamento de Ciencias Médicas, CSIC-Universidad de Castilla-La Mancha, Albacete, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
19
|
Fang D, Guan H, Liu J, Wei G, Ke W, Yao B, Xiao H, Li Y. Early intensive insulin therapy attenuates the p38 pathway in the renal cortex and indices of nephropathy in diabetic rats. Endocr J 2012; 59:81-90. [PMID: 22068113 DOI: 10.1507/endocrj.ej11-0057] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In this rodent study, we compared the effects of early versus late intensive insulin therapy on diabetic nephropathy and potential causal mechanisms. Diabetes was induced in rats by high-fat diet and low-dose streptozotocin. Intensive insulin therapy was initiated in the early intensive insulin therapy groups as soon as diabetes was confirmed and lasted for 8 (8wEI group) and 16 weeks (16wEI group). In the late insulin therapy group (LI group), intensive insulin treatment was initiated 8 weeks later and lasted for 8 weeks. Age-matched diabetic rats (8wDM group and 16wDM group) and non-diabetic rats (8wNC group and 16wNC group) served as controls. Histological analysis, real-time PCR, and western blot were performed in renal cortex specimens. Glomerular hypertrophy and mesangial matrix expansion were prominent in the 16wDM and LI groups while the EI groups remained normal and similar to the 16wNC group. Western blots revealed that p38 MAPK activities in the EI groups decreased significantly, whereas the level in the LI group was markedly higher than the 16wEI group, and not different from the DM groups. Activities of MKK3/6, CREB and MKP-1 proteins as well as CREB and MKP-1 mRNA showed a similar pattern. Therefore, we concluded that early intensive insulin treatment and attainment of good glycemic control counteracted some renal molecular pathways associated with epigenetic metabolic memory to minimize risk of diabetic nephropathy. However, late insulin therapy did not abrogate the increased renal cortical p38 MAPK pathway activation in diabetic rats and led to glomerular hypertrophy and extracellular matrix expansion.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetic Nephropathies/prevention & control
- Gene Expression Regulation/drug effects
- Glomerular Mesangium/drug effects
- Glomerular Mesangium/pathology
- Humans
- Hypertrophy
- Hypoglycemic Agents/therapeutic use
- Insulin, Isophane/therapeutic use
- Insulin, Regular, Human/therapeutic use
- Isophane Insulin, Human
- Kidney Cortex/drug effects
- Kidney Cortex/metabolism
- Kidney Cortex/pathology
- Kidney Glomerulus/drug effects
- Kidney Glomerulus/pathology
- MAP Kinase Signaling System/drug effects
- Male
- Phosphorylation/drug effects
- Protein Processing, Post-Translational/drug effects
- RNA, Messenger/metabolism
- Random Allocation
- Rats
- Rats, Sprague-Dawley
- Time Factors
- p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Donghong Fang
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Interleukin-1 receptor-mediated inflammation impairs the heat shock response of human mesothelial cells. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1544-55. [PMID: 21435443 DOI: 10.1016/j.ajpath.2010.12.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 11/22/2010] [Accepted: 12/30/2010] [Indexed: 11/22/2022]
Abstract
Bioincompatibility of peritoneal dialysis fluids (PDF) limits their use in renal replacement therapy. PDF exposure harms mesothelial cells but induces heat shock proteins (HSP), which are essential for repair and cytoprotection. We searched for cellular pathways that impair the heat shock response in mesothelial cells after PDF-exposure. In a dose-response experiment, increasing PDF-exposure times resulted in rapidly increasing mesothelial cell damage but decreasing HSP expression, confirming impaired heat shock response. Using proteomics and bioinformatics, simultaneously activated apoptosis-related and inflammation-related pathways were identified as candidate mechanisms. Testing the role of sterile inflammation, addition of necrotic cell material to mesothelial cells increased, whereas addition of the interleukin-1 receptor (IL-1R) antagonist anakinra to PDF decreased release of inflammatory cytokines. Addition of anakinra during PDF exposure resulted in cytoprotection and increased chaperone expression. Thus, activation of the IL-1R plays a pivotal role in impairment of the heat shock response of mesothelial cells to PDF. Danger signals from injured cells lead to an elevated level of cytokine release associated with sterile inflammation, which reduces expression of HSP and other cytoprotective chaperones and exacerbates PDF damage. Blocking the IL-1R pathway might be useful in limiting damage during peritoneal dialysis.
Collapse
|
21
|
Carrión B, Pérez–Martínez FC, Monteagudo S, Pérez–Carrión MD, Gómez–Roldán C, Ceña V, Pérez–Martínez J. Atorvastatin Reduces High Glucose Toxicity in Rat Peritoneal Mesothelial Cells. Perit Dial Int 2011; 31:325-31. [DOI: 10.3747/pdi.2010.00164] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective Continuous exposure of the peritoneal membrane to high glucose dialysis solutions can produce functional alterations in this membrane. We studied the toxic effects of high glucose (50 mmol/L and 83 mmol/L) and its reversal by atorvastatin (0.5 – 5 μmol/L) on cultures of rat peritoneal mesothelial cells (PMCs). Methods Rat PMCs were harvested from the peritonea of male Sprague–Dawley rats and grown in M199 medium supplemented with 10% fetal bovine serum. The effects of high glucose (50 mmol/L and 83 mmol/L) on levels of reactive oxygen species (ROS), on caspase 3 activity, and on phospho-p38 mitogen-activated protein kinase (MAPK) in the cultures were evaluated. Results Exposure to high glucose (for 4, 8, and 24 hours) increased intracellular levels of ROS and phospho-p38 MAPK (indices of cellular toxicity). Atorvastatin blocked these toxic effects of high glucose, being more effective against 50 mmol/L glucose (protective effects were observed above 0.5 μmol/L) than against 83 mmol/L (protective effects were observed above 2.5 μmol/L). Atorvastatin was also able to prevent glucose-induced increase in caspase 3 activity. Conclusions The present study shows that high glucose may promote oxidative stress and may activate apoptotic pathways in rat PMCs. These toxic effects could be reversed by atorvastatin.
Collapse
Affiliation(s)
| | | | | | | | | | - Valentín Ceña
- Department of Nephrology, Complejo Hospitalario Universitario Albacete, Albacete
- Unidad Asociada Neurodeath, CSIC–Universidad de Castilla–La Mancha, Departamento de Ciencias Médicas, Albacete, Spain
| | | |
Collapse
|
22
|
Aoki S, Makino J, Nagashima A, Takezawa T, Nomoto N, Uchihashi K, Matsunobu A, Sanai T, Sugihara H, Toda S. Fluid flow stress affects peritoneal cell kinetics: possible pathogenesis of peritoneal fibrosis. Perit Dial Int 2011; 31:466-76. [PMID: 21532005 DOI: 10.3747/pdi.2010.00157] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Peritoneal fibrosis is an essential precursor condition to the development of encapsulating peritoneal sclerosis (EPS). This serious complication leads to a high mortality rate in peritoneal dialysis (PD) patients. Although several factors, including highly concentrated glucose in the dialysis solution, are believed to be potent agents for peritoneal fibrosis, the underlying mechanism remains unclear. During PD, the dialysis solution continuously generates fluid flow stress to the peritoneum under peristalsis and body motion. Fluid flow stress has been implicated as playing a critical role in the physiologic responses of many cell types. We therefore hypothesized that fluid flow stress may be involved in the pathogenesis of peritoneal fibrosis leading to EPS. METHODS To generate fluid flow stress, culture containers were placed on a rotatory shaker in a thermostatic chamber. In this system, the shaker rotated at a speed of 25 rpm with a radius of 1.5 cm. Mesothelial cells were cultured in low-glucose (1000 mg/L) or high-glucose (4500 mg/L) complete medium with and without flow stress. RESULTS Fluid flow stress promoted hyperplasia and epithelial-mesenchymal transition (EMT) of mesothelial cells independent of glucose concentration. Fluid flow stress inhibited expression of ERK (extracellular signal-regulated kinase) and p38 MAPK (mitogen-activated protein kinase) in mesothelial cells. Administration of ERK and p38 MAPK inhibitors replicated the stress-induced morphology of mesothelial cells. CONCLUSIONS The present data indicate that fluid flow stress promotes hyperplasia and EMT of mesothelial cells via the MAPK axis, suggesting that fluid flow stress may be involved in the pathogenesis of peritoneal fibrosis.
Collapse
Affiliation(s)
- Shigehisa Aoki
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, Saga, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Washida N, Wakino S, Tonozuka Y, Homma K, Tokuyama H, Hara Y, Hasegawa K, Minakuchi H, Fujimura K, Hosoya K, Hayashi K, Itoh H. Rho-kinase inhibition ameliorates peritoneal fibrosis and angiogenesis in a rat model of peritoneal sclerosis. Nephrol Dial Transplant 2011; 26:2770-9. [PMID: 21378147 DOI: 10.1093/ndt/gfr012] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Peritoneal fibrosis (PF) and angiogenesis are typical morphological changes, leading to loss of peritoneal functions in patients undergoing peritoneal dialysis. The small G protein, Rho, and its downstream effector Rho-kinase have been shown to be involved in the tissue fibrosis process. This study was undertaken to investigate the role of Rho-kinase in the pathogenesis of these alterations. METHODS PF was induced by intraperitoneal administration of chlorhexidine (CHX) in male rats (CHX group). These rats were treated with a Rho-kinase inhibitor, fasudil (Fas group). Human pleural mesothelial cells, MeT-5A cells, were stimulated by glucose with or without another Rho-kinase inhibitor, Y-27632. RESULTS Peritoneal damage including peritoneal thickening, fibrous changes, macrophage migration and angiogenesis were evident in the CHX group and were ameliorated in the Fas group. The expression of markers of tissue fibrosis, such as transforming growth factor (TGF)-β, fibronectin and α-smooth muscle cell actin, were increased in the CHX group and were downregulated by fasudil. Similar results were also seen with an inducer of angiogenesis, vascular endothelial growth factor (VEGF). Rho-kinase was activated in the peritoneum of the CHX group, which was inhibited by fasudil. In MeT-5A cells, high glucose increased TGF-β expression and VEGF secretion, which were blocked by Y-27632. CONCLUSIONS The activation of Rho-kinase is involved in peritoneal damage at multiple stages including tissue fibrosis and angiogenesis. The inhibition of Rho-kinase constitutes a novel strategy for the treatment of PF.
Collapse
Affiliation(s)
- Naoki Washida
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Riesenhuber A, Kratochwill K, Bender TO, Vargha R, Kasper DC, Herzog R, Salzer E, Aufricht C. Peritoneal dialysis fluid induces p38-dependent inflammation in human mesothelial cells. Perit Dial Int 2010; 31:332-9. [PMID: 21193553 DOI: 10.3747/pdi.2009.00206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Noninfectious upregulation of proinflammatory pathways in mesothelial cells may represent an integral part of their stress response upon exposure to peritoneal dialysis fluids (PDF). OBJECTIVE The aim of this study was to evaluate the role of the stress-inducible mitogen-activated protein kinase (MAPK) p38 in regulation of inflammatory and stress responses in mesothelial cells following in vitro exposure to PDF. MATERIALS AND METHODS Human peritoneal mesothelial cells were exposed to Dianeal PD4 or Physioneal (Baxter AG, Vienna, Austria) containing 1.36% glucose and then allowed to recover. Phosphorylation of p38, induction of heat shock protein-70 (HSP70), release of lactate dehydrogenase (LDH), secretion of interleukin (IL)-8, gene transcription, and mRNA stability were assessed with and without the MAPK p38 inhibitor SB203580. RESULTS Exposure to Dianeal resulted in phosphorylation of p38 within 30 minutes (309% of control, p < 0.05) and increased IL-8 release (370% of control, p < 0.05), HSP70 expression (151% of control, p < 0.05), and LDH release (180% of control, p < 0.05). Exposure to Physioneal resulted in attenuated changes in IL-8, HSP70, and LDH. Addition of the p38 inhibitor SB203580 to Dianeal resulted in dampened IL-8 release (-55%; p < 0.05) and basal HSP70 expression, and unchanged LDH release. Effects of p38 on IL-8 were at transcriptional, posttranscriptional, and translational levels. CONCLUSION These data confirm concordant p38-dependent upregulation of IL-8 and HSP70 following exposure to bioincompatible PDF. The MAPK p38 pathway therefore links proinflammatory processes and the cellular stress response in human peritoneal mesothelial cells.
Collapse
|
25
|
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
|
26
|
Ogata R, Hiramatsu N, Hayakawa K, Nakajima S, Yao J, Kobayashi T, Kitamura M. Impairment of MCP-1 expression in mesothelial cells exposed to peritoneal dialysis fluid by osmotic stress and acidic stress. Perit Dial Int 2010; 31:80-9. [PMID: 20448242 DOI: 10.3747/pdi.2009.00159] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Bacterial peritonitis is one of the most frequent complications in patients on peritoneal dialysis. In the present study, we investigated effects of peritoneal dialysis fluid (PDF) on mesothelial cell recruitment of macrophages, focusing especially on unphysiological properties of PDF. METHODS Human and murine mesothelial cells were exposed to PDF or individual properties of PDF (low pH, high glucose concentration, hyperosmolality, high lactate concentration) in vitro and in vivo, treated with inflammatory stimuli, and subjected to analyses of monocyte chemoattractant protein-1 (MCP-1), nuclear factor-κB (NF-κB), mitogen-activated protein (MAP) kinases, and MAP kinase phosphatase-1 (MKP-1). RESULTS We found that intraperitoneal administration of PDF suppressed expression of MCP-1 and infiltration of mononuclear cells in the peritoneum of mice following injection with lipopolysaccharide. Among the unphysiological properties of PDF, low pH and hyperosmolality caused blunted induction of MCP-1 in cytokine-stimulated mesothelial cells. The attenuated response was ascribed to suppression of NF-κB by low pH and inhibition of p38 MAP kinase by hyperosmolality. Furthermore, the attenuated phosphorylation of p38 MAP kinase by osmotic stress was associated with induction of MKP-1. CONCLUSION These results suggest a possibility that mesothelial cells exposed to PDF exhibit attenuated MCP-1 expression and consequent impairment of macrophage recruitment through dual mechanisms, that is, inhibition of NF-κB by acidic stress and blunted activation of p38 MAP kinase by osmotic stress. In patients on peritoneal dialysis, blunted expression of chemokines may lead to perturbation of bacterial clearance by macrophages in the peritoneal cavity.
Collapse
Affiliation(s)
- Ryouji Ogata
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Schilte MN, Celie JW, ter Wee PM, Beelen RH, van den Born J. Factors Contributing to Peritoneal Tissue Remodeling in Peritoneal Dialysis. Perit Dial Int 2009. [DOI: 10.1177/089686080902900604] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Peritoneal dialysis (PD) is associated with functional and structural changes of the peritoneal membrane. In this review we describe factors contributing to peritoneal tissue remodeling, including uremia, peritonitis, volume loading, the presence of a catheter, and the PD fluid itself. These factors initiate recruitment and activation of peritoneal cells such as macrophages and mast cells, as well as activation of peritoneal cells, including mesothelial cells, fibroblasts, and endothelial cells. We provide an overview of cytokines, growth factors, and other mediators involved in PD-associated changes. Activation of downstream pathways of cellular modulators can induce peritoneal tissue remodeling, leading to ultrafiltration loss. Identification of molecular pathways, cells, and cytokines involved in the development of angiogenesis, fibrosis, and membrane failure may lead to innovative therapeutic strategies that can protect the peritoneal membrane from the consequences of long-term PD.
Collapse
Affiliation(s)
- Margot N. Schilte
- Departments of Molecular Cell Biology and Immunology VU University Medical Center, Amsterdam, The Netherlands
| | - Johanna W.A.M Celie
- Departments of Molecular Cell Biology and Immunology VU University Medical Center, Amsterdam, The Netherlands
| | - Piet M. ter Wee
- Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - Robert H.J. Beelen
- Departments of Molecular Cell Biology and Immunology VU University Medical Center, Amsterdam, The Netherlands
| | - Jacob van den Born
- Departments of Molecular Cell Biology and Immunology VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Jung DS, Li JJ, Kwak SJ, Lee SH, Park J, Song YS, Yoo TH, Han SH, Lee JE, Kim DK, Moon SJ, Kim YS, Han DS, Kang SW. FR167653 inhibits fibronectin expression and apoptosis in diabetic glomeruli and in high-glucose-stimulated mesangial cells. Am J Physiol Renal Physiol 2008; 295:F595-604. [DOI: 10.1152/ajprenal.00624.2007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous in vitro studies suggest that the p38 MAPK pathway may be involved in the pathogenesis of diabetic nephropathy, but the consequences of the inhibition of the p38 MAPK pathway have not been well elucidated in diabetic (DM) glomeruli. This study was undertaken to investigate the effect of p38 MAPK inhibitor, FR167653, on fibronectin expression and apoptosis in DM glomeruli and in high-glucose-stimulated mesangial cells (MC). In vivo, 32 Sprague-Dawley rats were injected with diluent (control, N = 16) or streptozotocin intraperitoneally (DM, N = 16). Eight rats from each group were treated with FR167653 for 3 mo. In vitro, rat MC were exposed to medium containing 5.6 mM glucose or 30 mM glucose [high glucose (HG)] with or without 10−6 M FR167653 for 24 h. Fibronectin mRNA and protein expression were determined by real-time PCR and Western blot, respectively. Western blot for apoptosis-related molecules, terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling assay, and Hoechst 33342 staining were performed to determine apoptosis. FR167653 ameliorated the increases in fibronectin-to-GAPDH mRNA ratio and protein expression in DM glomeruli by 89 and 79% and in HG-stimulated MC by 70 and 91%, respectively ( P < 0.05). Under diabetic conditions, Bcl-2 protein expression was decreased, whereas cleaved caspase-3 protein expression was increased ( P < 0.05), and these changes were inhibited by FR167653 treatment. Apoptotic cells were also significantly increased in DM glomeruli and in HG-stimulated MC ( P < 0.05), and FR167653 ameliorated these increases in apoptotic cells, both in vivo and in vitro. In conclusion, these findings suggest that the inhibition of the p38 MAPK pathway has a beneficial effect on the development of diabetic nephropathy by inhibiting the increase in fibronectin expression and apoptosis.
Collapse
|
29
|
Hung KY, Huang JW, Chiang CK, Tsai TJ. Preservation of peritoneal morphology and function by pentoxifylline in a rat model of peritoneal dialysis: molecular studies. Nephrol Dial Transplant 2008; 23:3831-40. [PMID: 18614818 DOI: 10.1093/ndt/gfn369] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND High-glucose (HG) content of dialysate accelerated peritoneal fibrosis. We investigated in vitro mechanisms and the in vivo potential of pentoxifylline (PTX) to prevent this fibrogenic process. METHODS For human peritoneal mesothelial cell (HPMC) culture, a normal-glucose (NG, 5.5 mM) or HG (138 mM) medium was established through pilot experiments. The rat peritoneal dialysis (PD) model consists of four groups (n = 8): group 1, intraperitoneal (IP) HG (4.25%) solution; group 2, as group 1 plus daily IP PTX (4 mg/in 1 h); group 3, IP PTX and group 4 as control. RESULTS In HPMC culture, PTX significantly prevented HG-stimulated gene and protein production of collagen and transforming growth factor-beta1 (TGF-ss1) (reduction rate of 72-81%). The p38 mitogen-activated protein kinase (MAPK) pathway was activated significantly in HG-treated HPMCs. Blockade of p38 MAPK by SB203580 (25 microM) or PTX (300 microg/ml) resulted in an effective suppression of collagen and TGF-ss1 gene expression in HG-cultured HPMCs. In PD experimental animals, peritoneal thickness and collagen expression in the peritoneum were significantly increased in HG-treated rats, and was attenuated by PTX (P < 0.01). Impaired peritoneal ultrafiltration (1.9 +/- 0.5 ml versus 2.4 +/- 0.4 ml, P < 0.05) and stimulated proinflammatory IL-6, MCP-1 and TGF-beta1 activation were observed in HG-treated rats. PTX well preserved the functional characteristics of peritoneum and cytokine profiles. CONCLUSIONS These in vitro and in vivo data suggest that PTX may have therapeutic benefits for the prevention or retardation of peritoneal fibrosis.
Collapse
Affiliation(s)
- Kuan-Yu Hung
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan, Republic of China
| | | | | | | |
Collapse
|
30
|
Kiribayashi K, Masaki T, Naito T, Ogawa T, Ito T, Yorioka N, Kohno N. Angiotensin II induces fibronectin expression in human peritoneal mesothelial cells via ERK1/2 and p38 MAPK. Kidney Int 2005; 67:1126-35. [PMID: 15698454 DOI: 10.1111/j.1523-1755.2005.00179.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND The renin-angiotensin system has been implicated in the pathogenesis of fibrosis in various organs. However, its involvement in peritoneal fibrosis, a crucial complication of peritoneal dialysis, is unclear. Human peritoneal mesothelial cells (HPMC) play a major role in peritoneal fibrosis by producing extracellular matrix (ECM). However, there is scant data regarding the effect of angiotensin II (Ang II) on ECM expression and signal transduction pathways in HPMC. METHODS The concentration of Ang II in the peritoneal dialysis effluent was measured by radioimmunoassay. We investigated the expression of Ang II type 1 (AT1) and type 2 (AT2) receptors by HPMC. We also examined the effect of Ang II upon fibronectin production by HPMC, and dissected the receptor and intracellular signaling pathways involved. RESULTS Ang II levels in the peritoneal dialysis effluent at the onset of peritonitis were 30 times higher than baseline levels. HPMC expression of AT1 and AT2 receptors was confirmed at the mRNA and protein level by reverse transcriptase-polymerase chain reaction (PCR), Western blotting, and immunocytochemistry. Quantitative reverse transcriptase-PCR and Western blotting showed that 10 nmol/L Ang II increased fibronectin mRNA expression followed by secretion of fibronectin protein. This response was completely inhibited by the AT1 receptor antagonist RNH6270, while the AT2 receptor antagonist PD123319 had no effect. Ang II-induced fibronectin expression was mediated by the activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase (p38 MAPK), but not c-Jun N-terminal kinase. CONCLUSION These results indicate the potential importance of ERK1/2 and p38 MAPK signaling pathways in Ang II-induced fibronectin expression in HPMC, and suggest the therapeutic potential of AT1 receptor blockers in the prevention or treatment of peritoneal fibrosis in patients on peritoneal dialysis.
Collapse
Affiliation(s)
- Kei Kiribayashi
- Department of Molecular and Internal Medicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan.
| | | | | | | | | | | | | |
Collapse
|
31
|
Lee HB, Yu MR, Song JS, Ha H. Reactive oxygen species amplify protein kinase C signaling in high glucose-induced fibronectin expression by human peritoneal mesothelial cells. Kidney Int 2004; 65:1170-9. [PMID: 15086456 DOI: 10.1111/j.1523-1755.2004.00491.x] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND We previously demonstrated that high glucose up-regulates fibronectin mRNA and protein expression by human peritoneal mesothelial cells (HPMC) through activation of protein kinase C (PKC). PKC is known to induce cellular reactive oxygen species (ROS) and PKC-dependent activation of the reduced form of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase has recently been shown to be responsible, in part, for increased oxidative stress in diabetes. On the other hand, high glucose-induced mitochondrial overproduction of superoxide anion was found to activate PKC. We, therefore, hypothesized that high glucose-induced activation of PKC in HPMC may increase cellular ROS and ROS, in turn, may activate PKC and thus provide signal amplification in high glucose-induced fibronectin up-regulation in HPMC. METHODS The role of ROS in high glucose- and PKC-induced fibronectin expression was examined by quantification of cellular ROS after stimulation with high glucose and phorbol 12-myristate 13-acetate (PMA), by the effect of hydrogen peroxide (H(2)O(2)) and PMA on fibronectin expression, and finally by inhibition of ROS and PKC. The source of cellular ROS was further examined by inhibition of NADPH oxidase and mitochondrial metabolism. RESULTS D-glucose increased dichlorofluorescein (DCF)-sensitive cellular ROS in HPMC in a dose-dependent manner. l-glucose did not induce ROS generation and cytochalasin B completely blocked high glucose-induced ROS generation, suggesting that glucose uptake, but not media hyperosmolality, is required in ROS generation in HPMC. PMA increased cellular ROS and fibronectin secretion. A single dose of H(2)O(2) or H(2)O(2) continuously generated by glucose oxidase up-regulated fibronectin expression [corrected]. Antioxidants trolox and catalase inhibited high glucose- and PMA-induced fibronectin mRNA and protein expression. Inhibition of PKC inhibited high glucose-and H(2)O(2)-induced fibronectin secretion. NADPH oxidase inhibitors (diphenyleneiodinium and apocynin) and an inhibitor of mitochondrial electron transport chain subunit I (rotenone) all effectively inhibited high glucose-induced cellular ROS generation and fibronectin secretion. CONCLUSION The present data demonstrate that high glucose increases cellular ROS in HPMC through activation of PKC, NADPH oxidase, and mitochondrial metabolism and that ROS, thus generated, up-regulate fibronectin expression by HPMC. ROS are not only downstream but also upstream signaling molecules to PKC and provide signal amplification in high glucose-induced fibronectin expression by HPMC. The present data imply that cellular ROS may be potential therapeutic targets in progressive accumulation of extracellular matrix in the peritoneal tissue of long-term peritoneal dialysis patients using high glucose-containing peritoneal dialysis solutions.
Collapse
Affiliation(s)
- Hi Bahl Lee
- Hyonam Kidney Laboratory, Soon Chun Hyang University, Seoul, Korea
| | | | | | | |
Collapse
|
32
|
Campbell M, Allen WE, Sawyer C, Vanhaesebroeck B, Trimble ER. Glucose-Potentiated Chemotaxis in Human Vascular Smooth Muscle Is Dependent on Cross-Talk Between the PI3K and MAPK Signaling Pathways. Circ Res 2004; 95:380-8. [PMID: 15242975 DOI: 10.1161/01.res.0000138019.82184.5d] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Atheroma formation involves the movement of vascular smooth muscle cells (VSMC) into the subendothelial space. The aim of this study was to determine the involvement of PI3K and MAPK pathways and the importance of cross-talk between these pathways, in glucose-potentiated VSMC chemotaxis to serum factors. VSMC chemotaxis occurred in a serum gradient in 25 mmol/L glucose (but not in 5 mmol/L glucose) in association with increased phosphorylation (activation) of Akt and ERK1/2 in PI3K and MAPK pathways, respectively. Inhibitors of these pathways blocked chemotaxis, as did an mTOR inhibitor. VSMC expressed all class IA PI3K isoforms, but microinjection experiments demonstrated that only the p110β isoform was involved in chemotaxis. ERK1/2 phosphorylation was reduced not only by MAPK pathway inhibitors but also by PI3K and mTOR inhibitors; when PI3K was inhibited, ERK phosphorylation could be induced by microinjected activated Akt, indicating important cross-talk between the PI3K and ERK1/2 pathways. Glucose-potentiated phosphorylation of molecules in the p38 and JNK MAPK pathways inhibited these pathways but did not affect chemotaxis. The statin, mevinolin, blocked chemotaxis through its effects on the MAPK pathway. Mevinolin-inhibited chemotaxis was restored by farnesylpyrophosphate but not by geranylgeranylpyrophosphate; in the absence of mevinolin, inhibition of farnesyltransferase reduced ERK phosphorylation and blocked chemotaxis, indicating a role for the Ras family of GTPases (MAPK pathway) under these conditions. In conclusion, glucose sensitizes VSMC to serum, inducing chemotaxis via pathways involving p110β-PI3K, Akt, mTOR, and ERK1/2 MAPK. Cross-talk between the PI3K and MAPK pathways is necessary for VSMC chemotaxis under these conditions.
Collapse
MESH Headings
- Alkyl and Aryl Transferases/antagonists & inhibitors
- Androstadienes/pharmacology
- Anthracenes/pharmacology
- Antibodies, Monoclonal/pharmacology
- Cells, Cultured/cytology
- Cells, Cultured/drug effects
- Cells, Cultured/metabolism
- Chemotaxis/drug effects
- Chemotaxis/physiology
- Chromones/pharmacology
- Class I Phosphatidylinositol 3-Kinases
- Farnesyltranstransferase
- Flavonoids/pharmacology
- Glucose/pharmacology
- Humans
- Imidazoles/pharmacology
- Isoenzymes/antagonists & inhibitors
- Isoenzymes/physiology
- JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors
- JNK Mitogen-Activated Protein Kinases/physiology
- Lovastatin/pharmacology
- MAP Kinase Kinase 4
- MAP Kinase Signaling System/drug effects
- Mitogen-Activated Protein Kinase 1/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 1/physiology
- Mitogen-Activated Protein Kinase 3/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 3/physiology
- Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors
- Mitogen-Activated Protein Kinase Kinases/physiology
- Morpholines/pharmacology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- Phosphatidylinositol 3-Kinases/physiology
- Phosphoinositide-3 Kinase Inhibitors
- Polyisoprenyl Phosphates/pharmacology
- Protein Kinases/physiology
- Protein Serine-Threonine Kinases/physiology
- Proto-Oncogene Proteins/physiology
- Proto-Oncogene Proteins c-akt
- Pyridines/pharmacology
- Sesquiterpenes
- Sirolimus/pharmacology
- TOR Serine-Threonine Kinases
- Wortmannin
- p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
- p38 Mitogen-Activated Protein Kinases/physiology
- ras Proteins/physiology
Collapse
Affiliation(s)
- Malcolm Campbell
- Department of Clinical Biochemistry and Metabolic Medicine, Queen's University Belfast, Institute of Clinical Science, Royal Victoria Hospital, Grosvenor Road, Belfast, UK
| | | | | | | | | |
Collapse
|
33
|
Abstract
In vitro biocompatibility performance of Physioneal. toneal dialysis (PD) has been a successful and effective form of chronic renal replacement therapy since its introduction over 20 years ago. Despite its overall success, there is a growing body of evidence that suggests shortcomings in the preservation of membrane integrity. This has led to the development of several second-generation PD solutions that demonstrate improved biocompatibility. Physioneal, a neutral pH, bicarbonate/lactate-buffered solution, was one of the first of these new PD solutions to become commercially available. This review will focus on one of the first preclinical stages in the development of Physioneal: studies on in vitro biocompatibility testing. Studies in leukocyte, mesothelial cell, and fibroblast populations demonstrated significantly improved biocompatibility of neutral pH, bicarbonate/lactate-based solutions compared to conventional solutions. The solutions contributed to improved leukocyte viability and response to bacterial infection (e.g., phagocytosis, superoxide radical generation, and endotoxin-stimulated cytokine release). Studies on peritoneal mesothelial cells demonstrate improved cell viability, proliferation, and response to proinflammatory stimuli, and a reduced potential for angiogenesis and peritoneal fibrosis, all suggesting a better preservation of membrane structure and function. The bicarbonate/lactate-based solutions demonstrated decreased cytotoxicity and preserved cell growth in fibroblast cultures as well. In vitro biocompatibility testing has clearly demonstrated that neutral pH, bicarbonate/lactate-buffered Physioneal solutions are superior to conventional solutions in preserving cell viability and function in cell populations that contribute to peritoneal homeostasis. This positive assessment now provides a foundation and rationale for moving forward with the next stages in preclinical testing: in vivo animal models and human ex vivo studies.
Collapse
Affiliation(s)
- Catherine M Hoff
- Renal Division Research, Baxter Healthcare Corporation, McGaw Park, Illinois 60085-6730, USA.
| |
Collapse
|