1
|
Li S, Zhuang Y, Ji Y, Chen X, He L, Chen S, Luo Y, Shen L, Xiao J, Wang H, Luo C, Peng F, Long H. BRG1 accelerates mesothelial cell senescence and peritoneal fibrosis by inhibiting mitophagy through repression of OXR1. Free Radic Biol Med 2024; 214:54-68. [PMID: 38311259 DOI: 10.1016/j.freeradbiomed.2024.01.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024]
Abstract
Peritoneal mesothelial cell senescence promotes the development of peritoneal dialysis (PD)-related peritoneal fibrosis. We previously revealed that Brahma-related gene 1 (BRG1) is increased in peritoneal fibrosis yet its role in modulating peritoneal mesothelial cell senescence is still unknown. This study evaluated the mechanism of BRG1 in peritoneal mesothelial cell senescence and peritoneal fibrosis using BRG1 knockdown mice, primary peritoneal mesothelial cells and human peritoneal samples from PD patients. The augmentation of BRG1 expression accelerated peritoneal mesothelial cell senescence, which attributed to mitochondrial dysfunction and mitophagy inhibition. Mitophagy activator salidroside rescued fibrotic responses and cellular senescence induced by BRG1. Mechanistically, BRG1 was recruited to oxidation resistance 1 (OXR1) promoter, where it suppressed transcription of OXR1 through interacting with forkhead box protein p2. Inhibition of OXR1 abrogated the improvement of BRG1 deficiency in mitophagy, fibrotic responses and cellular senescence. In a mouse PD model, BRG1 knockdown restored mitophagy, alleviated senescence and ameliorated peritoneal fibrosis. More importantly, the elevation level of BRG1 in human PD was associated with PD duration and D/P creatinine values. In conclusion, BRG1 accelerates mesothelial cell senescence and peritoneal fibrosis by inhibiting mitophagy through repression of OXR1. This indicates that modulating BRG1-OXR1-mitophagy signaling may represent an effective treatment for PD-related peritoneal fibrosis.
Collapse
Affiliation(s)
- Shuting Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Yiyi Zhuang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yue Ji
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaowen Chen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Liying He
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Sijia Chen
- Department of Nephrology and Rheumatology, The First Hospital of Changsha, Changsha, China
| | - Yating Luo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lingyu Shen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Xiao
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huizhen Wang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Congwei Luo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Fenfen Peng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Haibo Long
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Breborowicz A. Iron increases the profibrotic properties of the senescent peritoneal mesothelial cells. Int J Artif Organs 2023; 46:473-480. [PMID: 37564017 DOI: 10.1177/03913988231192123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
BACKGROUND Treatment of anemia in peritoneal dialysis patients often requires intravenous iron supplementation. Iron diffuses into the peritoneal cavity and is injurious to the peritoneum. We studied how intermittent exposure to iron changes the properties of the senescent peritoneal mesothelial cells (MC). METHODS Replicative senescence was induced in MC in control medium (Con) or in control medium with intermittent exposure to iron isomaltoside 15 µg/dL (Con-IIS). After 10 passages properties of MC from both groups were compared to MC not exposed to replicative senescence. RESULTS In senescent MC population doubling time was elongated, intracellular generation of free radicals and staining for β-galactosidase was stronger than in MC not exposed to replicative senescence. All these effects were stronger in MC intermittently exposed to IIS. In these cells intracellular iron content was also higher. Also expression of genes p21 and p53 was stronger in MC intermittently treated with IIS. In senescent cells higher release and expression of IL6 and TGFβ1 was observed and that effect was stronger in MC treated with iron. Senescent MC had reduced fibrinolytic activity, what may predispose to the peritoneal fibrosis. Synthesis of collagen was higher in senescent cells, more in MC treated with iron. CONCLUSION MC aging results in change of their genotype and phenotype which lead to their profibrotic effect. Exposure to iron enhances these changes.
Collapse
Affiliation(s)
- Andrzej Breborowicz
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Greater Poland Voivodeship, Poland
- Department of Histology, Collegium Medicum, Zielona Gora University, Zielona Gora, Poland
| |
Collapse
|
3
|
Proteome-Wide Differential Effects of Peritoneal Dialysis Fluid Properties in an In Vitro Human Endothelial Cell Model. Int J Mol Sci 2022; 23:ijms23148010. [PMID: 35887356 PMCID: PMC9317527 DOI: 10.3390/ijms23148010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 01/27/2023] Open
Abstract
To replace kidney function, peritoneal dialysis (PD) utilizes hyperosmotic PD fluids with specific physico-chemical properties. Their composition induces progressive damage of the peritoneum, leading to vasculopathies, decline of membrane function, and PD technique failure. Clinically used PD fluids differ in their composition but still remain bioincompatible. We mapped the molecular pathomechanisms in human endothelial cells induced by the different characteristics of widely used PD fluids by proteomics. Of 7894 identified proteins, 3871 were regulated at least by 1 and 49 by all tested PD fluids. The latter subset was enriched for cell junction-associated proteins. The different PD fluids individually perturbed proteins commonly related to cell stress, survival, and immune function pathways. Modeling two major bioincompatibility factors of PD fluids, acidosis, and glucose degradation products (GDPs) revealed distinct effects on endothelial cell function and regulation of cellular stress responses. Proteins and pathways most strongly affected were members of the oxidative stress response. Addition of the antioxidant and cytoprotective additive, alanyl-glutamine (AlaGln), to PD fluids led to upregulation of thioredoxin reductase-1, an antioxidant protein, potentially explaining the cytoprotective effect of AlaGln. In conclusion, we mapped out the molecular response of endothelial cells to PD fluids, and provided new evidence for their specific pathomechanisms, crucial for improvement of PD therapies.
Collapse
|
4
|
FK866 Protects Human Dental Pulp Cells against Oxidative Stress-Induced Cellular Senescence. Antioxidants (Basel) 2021; 10:antiox10020271. [PMID: 33578781 PMCID: PMC7916510 DOI: 10.3390/antiox10020271] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 01/12/2023] Open
Abstract
FK866 possesses various functional properties, such as anti-angiogenic, anti-cancer, and anti-inflammatory activities. We previously demonstrated that premature senescence of human dental pulp cells (hDPCs) was induced by hydrogen peroxide (H2O2). The present study aimed to investigate whether H2O2-induced premature senescence of hDPCs is affected by treatment with FK866. We found that FK866 markedly inhibited the senescent characteristics of hDPCs after exposure to H2O2, as revealed by an increase in the number of senescence-associated β-galactosidase (SA-β-gal)-positive hDPCs and the upregulation of the p21 and p53 proteins, which acts as molecular indicators of cellular senescence. Moreover, the stimulatory effects of H2O2 on cellular senescence are associated with oxidative stress induction, such as excessive ROS production and NADPH consumption, telomere DNA damage induction, and upregulation of senescence-associated secretory phenotype factors (IL-1β, IL-6, IL-8, COX-2, and TNF-α) as well as NF-κB activation, which were all blocked by FK866. Thus, FK866 might antagonize H2O2-induced premature senescence of hDPCs, acting as a potential therapeutic antioxidant by attenuating oxidative stress-induced pathologies in dental pulp, including inflammation and cellular senescence.
Collapse
|
5
|
Mikuła-Pietrasik J, Niklas A, Uruski P, Tykarski A, Książek K. Mechanisms and significance of therapy-induced and spontaneous senescence of cancer cells. Cell Mol Life Sci 2020; 77:213-229. [PMID: 31414165 PMCID: PMC6970957 DOI: 10.1007/s00018-019-03261-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/24/2019] [Accepted: 07/29/2019] [Indexed: 12/17/2022]
Abstract
In contrast to the well-recognized replicative and stress-induced premature senescence of normal somatic cells, mechanisms and clinical implications of senescence of cancer cells are still elusive and uncertain from patient-oriented perspective. Moreover, recent years provided multiple pieces of evidence that cancer cells may undergo senescence not only in response to chemotherapy or ionizing radiation (the so-called therapy-induced senescence) but also spontaneously, without any external insults. Since the molecular nature of the latter process is poorly recognized, the significance of spontaneously senescent cancer cells for tumor progression, therapy effectiveness, and patient survival is purely speculative. In this review, we summarize the most up-to-date research regarding therapy-induced and spontaneous senescence of cancer cells, by delineating the most important discoveries regarding the occurrence of these phenomena in vivo and in vitro. This review provides data collected from studies on various cancer cell models, and the narration is presented from the broader perspective of the most critical findings regarding the senescence of normal somatic cells.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Długa 1/2 Street, 61-848, Poznan, Poland
| | - Arkadiusz Niklas
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Długa 1/2 Street, 61-848, Poznan, Poland
| | - Paweł Uruski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Długa 1/2 Street, 61-848, Poznan, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Długa 1/2 Street, 61-848, Poznan, Poland
| | - Krzysztof Książek
- Department of Hypertensiology, Angiology and Internal Medicine, Poznan University of Medical Sciences, Długa 1/2 Street, 61-848, Poznan, Poland.
| |
Collapse
|
6
|
Gao L, Fan Y, Zhang X, Yang L, Huang W, Hang T, Li M, Du S, Ma J. Zinc supplementation inhibits the high glucose‑induced EMT of peritoneal mesothelial cells by activating the Nrf2 antioxidant pathway. Mol Med Rep 2019; 20:655-663. [PMID: 31115566 PMCID: PMC6580007 DOI: 10.3892/mmr.2019.10260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 04/11/2019] [Indexed: 01/17/2023] Open
Abstract
The high glucose (HG)-induced epithelial-mesenchymal transition (EMT) of peritoneal mesothelial cells (PMCs) serves an important role in peritoneal fibrosis (PF) during peritoneal dialysis. Our previous study reported that zinc (Zn) supplementation prevented the HG-induced EMT of rat PMCs in vitro. In the present study, the role of Zn in HG-induced EMT was investigated in vivo using a rat model of PF. Additionally, the molecular mechanisms underlying HG-induced EMT were studied in human PMCs (HPMCs). In the rat model of PF, HG treatment increased the glucose transfer capacity and decreased the ultrafiltration volume. Histopathological analysis revealed peritoneal thickening, increased expression of vimentin and decreased expression of E-cadherin. ZnSO4 significantly ameliorated the aforementioned changes, whereas Zn inhibition by clioquinol significantly aggravated the effects of HG on rats. The effects of Zn on HPMCs was assessed using western blot analysis, Transwell assays and flow cytometry. It was revealed that Zn also significantly suppressed the extent of the EMT, and reduced reactive oxygen species production and the migratory ability of HG-induced HPMCs, whereas Zn inhibition by N',N',N',N'-tetrakis (2-pyridylmethyl) ethylenediamine significantly potentiated the HG-induced EMT of HPMCs. HG-stimulated HPMCs exhibited increased expression of nuclear factor-like 2 (Nrf2) in the nucleus, and total cellular NAD(P)H quinone dehydrogenase 1 (NQO1) and heme oxygenase-1 (HO-1), the target proteins of the Nrf2 antioxidant pathway. Zn supplementation further promoted nuclear Nrf2 expression, and increased the expression of target proteins of the Nrf2 antioxidant pathway, whereas Zn depletion decreased nuclear Nrf2, NQO1 and HO-1 expression compared with the HG group. In conclusion, Zn supplementation was proposed to suppress the effects of HG on the EMT by stimulating the Nrf2 antioxidant pathway and subsequently reducing oxidative stress in PMCs.
Collapse
Affiliation(s)
- Lili Gao
- Department of Nephrology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yi Fan
- Department of Nephrology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiuli Zhang
- Department of Nephrology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lina Yang
- Department of Nephrology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Wenyu Huang
- Department of Nephrology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Tianyu Hang
- Department of Nephrology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Mingyang Li
- Department of Nephrology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Shuyan Du
- Department of Nephrology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jianfei Ma
- Department of Nephrology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
7
|
Wiesenhofer FM, Herzog R, Boehm M, Wagner A, Unterwurzacher M, Kasper DC, Alper SL, Vychytil A, Aufricht C, Kratochwill K. Targeted Metabolomic Profiling of Peritoneal Dialysis Effluents Shows Anti-oxidative Capacity of Alanyl-Glutamine. Front Physiol 2019; 9:1961. [PMID: 30719009 PMCID: PMC6348277 DOI: 10.3389/fphys.2018.01961] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 12/28/2018] [Indexed: 01/25/2023] Open
Abstract
Readily available peritoneal dialysis (PD) effluents from PD patients in the course of renal replacement therapy are a potentially rich source for molecular markers for predicting clinical outcome, monitoring the therapy, and therapeutic interventions. The complex clinical phenotype of PD patients might be reflected in the PD effluent metabolome. Metabolomic analysis of PD effluent might allow quantitative detection and assessment of candidate PD biomarkers for prognostication and therapeutic monitoring. We therefore subjected peritoneal equilibration test effluents from 20 stable PD patients, obtained in a randomized controlled trial (RCT) to evaluate cytoprotective effects of standard PD solution (3.86% glucose) supplemented with 8 mM alanyl-glutamine (AlaGln) to targeted metabolomics analysis. One hundred eighty eight pre-defined metabolites, including free amino acids, acylcarnitines, and glycerophospholipids, as well as custom metabolic indicators calculated from these metabolites were surveyed in a high-throughput assay requiring only 10 μl of PD effluent. Metabolite profiles of effluents from the cross-over trial were analyzed with respect to AlaGln status and clinical parameters such as duration of PD therapy and history of previous episodes of peritonitis. This targeted approach detected and quantified 184 small molecules in PD effluent, a larger number of detected metabolites than in all previous metabolomic studies in PD effluent combined. Metabolites were clustered within substance classes regarding concentrations after a 4-h dwell. PD effluent metabolic profiles were differentiated according to PD patient sub-populations, revealing novel changes in small molecule abundance during PD therapy. AlaGln supplementation of PD fluid altered levels of specific metabolites, including increases in alanine and glutamine but not glutamate, and reduced levels of small molecule indicators of oxidative stress, such as methionine sulfoxide. Our study represents the first application of targeted metabolomics to PD effluents. The observed metabolomic changes in PD effluent associated with AlaGln-supplementation during therapy suggested an anti-oxidant effect, and were consistent with the restoration of important stress and immune processes previously noted in the RCT. High-throughput detection of PD effluent metabolomic signatures and their alterations by therapeutic interventions offers new opportunities for metabolome-clinical correlation in PD and for prescription of personalized PD therapy.
Collapse
Affiliation(s)
- Florian M Wiesenhofer
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Rebecca Herzog
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Michael Boehm
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Anja Wagner
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Unterwurzacher
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Seth L Alper
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Boston, MA, United States.,Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Andreas Vychytil
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Christoph Aufricht
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Klaus Kratochwill
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
Wang J, Liu C, Chang X, Qi Y, Zhu Z, Yang X. Fibrosis of mesothelial cell-induced peritoneal implantation of ovarian cancer cells. Cancer Manag Res 2018; 10:6641-6647. [PMID: 30584359 PMCID: PMC6284525 DOI: 10.2147/cmar.s183043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Peritoneal metastasis frequently occurs in patients with advanced ovarian cancer and is the main basis for a poor prognosis. The mechanism underlying preferential ovarian cancer spread to the peritoneum is not well understood. Methods Herein, we investigated the significance and mechanism underlying fibrosis of mesothelial cells promoting peritoneal implantation of ovarian cancer. We have assessed the mesothelial cell fibroblast transformation process in peritoneal tissues of omentum and fibrotic mesothelial cell release of chemokines to promote dissemination by scanning electron microscopy, ELISA, Western blot, and Transwell chamber assay. Results We showed that the fibrosis of mesothelial cells exists in the peritoneum of ovarian cancer patients with peritoneal metastasis. Fibrosis of the mesothelial cells was induced by TGF-β1, which upregulates the CXCL12–CXCR4 and CXCL16–CXCR6 axes of mesothelial cells. Conclusion CXCL12–CXCR4 and CXCL16–CXCR6 may be important signaling pathways closely involved in peritoneal metastasis of ovarian cancer that require further investigation. The findings may lead to developing alternative strategies aimed at preventing and treating the metastasis of ovarian cancer.
Collapse
Affiliation(s)
- Jinou Wang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China,
| | - Chang Liu
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China,
| | - Xiaoying Chang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China,
| | - Yafei Qi
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China,
| | - Zhi Zhu
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Xianghong Yang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China,
| |
Collapse
|
9
|
Biomarker research to improve clinical outcomes of peritoneal dialysis: consensus of the European Training and Research in Peritoneal Dialysis (EuTRiPD) network. Kidney Int 2017; 92:824-835. [PMID: 28797473 DOI: 10.1016/j.kint.2017.02.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/16/2017] [Accepted: 02/27/2017] [Indexed: 12/15/2022]
Abstract
Peritoneal dialysis (PD) therapy substantially requires biomarkers as tools to identify patients who are at the highest risk for PD-related complications and to guide personalized interventions that may improve clinical outcome in the individual patient. In this consensus article, members of the European Training and Research in Peritoneal Dialysis Network (EuTRiPD) review the current status of biomarker research in PD and suggest a selection of biomarkers that can be relevant to the care of PD patients and that are directly accessible in PD effluents. Currently used biomarkers such as interleukin-6, interleukin-8, ex vivo-stimulated interleukin-6 release, cancer antigen-125, and advanced oxidation protein products that were collected through a Delphi procedure were first triaged for inclusion as surrogate endpoints in a clinical trial. Next, novel biomarkers were selected as promising candidates for proof-of-concept studies and were differentiated into inflammation signatures (including interleukin-17, M1/M2 macrophages, and regulatory T cell/T helper 17), mesothelial-to-mesenchymal transition signatures (including microRNA-21 and microRNA-31), and signatures for senescence and inadequate cellular stress responses. Finally, the need for defining pathogen-specific immune fingerprints and phenotype-associated molecular signatures utilizing effluents from the clinical cohorts of PD patients and "omics" technologies and bioinformatics-biostatistics in future joint-research efforts was expressed. Biomarker research in PD offers the potential to develop valuable tools for improving patient management. However, for all biomarkers discussed in this consensus article, the association of biological rationales with relevant clinical outcomes remains to be rigorously validated in adequately powered, prospective, independent clinical studies.
Collapse
|
10
|
Simon F, Tapia P, Armisen R, Echeverria C, Gatica S, Vallejos A, Pacheco A, Sanhueza ME, Alvo M, Segovia E, Torres R. Human Peritoneal Mesothelial Cell Death Induced by High-Glucose Hypertonic Solution Involves Ca 2+ and Na + Ions and Oxidative Stress with the Participation of PKC/NOX2 and PI3K/Akt Pathways. Front Physiol 2017; 8:379. [PMID: 28659813 PMCID: PMC5468383 DOI: 10.3389/fphys.2017.00379] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 05/22/2017] [Indexed: 01/07/2023] Open
Abstract
Chronic peritoneal dialysis (PD) therapy is equally efficient as hemodialysis while providing greater patient comfort and mobility. Therefore, PD is the treatment of choice for several types of renal patients. During PD, a high-glucose hyperosmotic (HGH) solution is administered into the peritoneal cavity to generate an osmotic gradient that promotes water and solutes transport from peritoneal blood to the dialysis solution. Unfortunately, PD has been associated with a loss of peritoneal viability and function through the generation of a severe inflammatory state that induces human peritoneal mesothelial cell (HPMC) death. Despite this deleterious effect, the precise molecular mechanism of HPMC death as induced by HGH solutions is far from being understood. Therefore, the aim of this study was to explore the pathways involved in HGH solution-induced HPMC death. HGH-induced HPMC death included influxes of intracellular Ca2+ and Na+. Furthermore, HGH-induced HPMC death was inhibited by antioxidant and reducing agents. In line with this, HPMC death was induced solely by increased oxidative stress. In addition to this, the cPKC/NOX2 and PI3K/Akt intracellular signaling pathways also participated in HGH-induced HPMC death. The participation of PI3K/Akt intracellular is in agreement with previously shown in rat PMC apoptosis. These findings contribute toward fully elucidating the underlying molecular mechanism mediating peritoneal mesothelial cell death induced by high-glucose solutions during peritoneal dialysis.
Collapse
Affiliation(s)
- Felipe Simon
- Departamento de Ciencias Biologicas, Facultad de Ciencias Biologicas and Facultad de Medicina, Universidad Andres BelloSantiago, Chile.,Millennium Institute on Immunology and ImmunotherapySantiago, Chile
| | - Pablo Tapia
- Unidad de Paciente Critico, Hospital Clínico Metropolitano de La FloridaSantiago, Chile
| | - Ricardo Armisen
- Centro de Investigación y Tratamiento del Cancer, Facultad de Medicina, Universidad de ChileSantiago, Chile.,Center for Excellence in Precision Medicine Pfizer, Pfizer ChileSantiago, Chile
| | - Cesar Echeverria
- Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo OHigginsSantiago, Chile
| | - Sebastian Gatica
- Departamento de Ciencias Biologicas, Facultad de Ciencias Biologicas and Facultad de Medicina, Universidad Andres BelloSantiago, Chile
| | - Alejandro Vallejos
- Departamento de Ciencias Biologicas, Facultad de Ciencias Biologicas and Facultad de Medicina, Universidad Andres BelloSantiago, Chile
| | - Alejandro Pacheco
- Sección de Nefrología, Departamento de Medicina, Hospital Clínico Universidad de ChileSantiago, Chile
| | - Maria E Sanhueza
- Sección de Nefrología, Departamento de Medicina, Hospital Clínico Universidad de ChileSantiago, Chile
| | - Miriam Alvo
- Sección de Nefrología, Departamento de Medicina, Hospital Clínico Universidad de ChileSantiago, Chile
| | - Erico Segovia
- Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo OHigginsSantiago, Chile
| | - Rubén Torres
- Sección de Nefrología, Departamento de Medicina, Hospital Clínico Universidad de ChileSantiago, Chile.,Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de ChileSantiago, Chile
| |
Collapse
|
11
|
Zhang N, Li Z, Xu K, Wang Y, Wang Z. Resveratrol Protects against High-Fat Diet Induced Renal Pathological Damage and Cell Senescence by Activating SIRT1. Biol Pharm Bull 2017; 39:1448-54. [PMID: 27582325 DOI: 10.1248/bpb.b16-00085] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Obesity-related renal diseases have been a worldwide issue. Effective strategy that prevents high fat-diet induced renal damage is of great significance. Resveratrol, a natural plant polyphenol, is famous for its antioxidant activity, cardioprotective effects and anticancer properties. However whether resveratrol can play a role in the treatment of renal diseases is unknown. In this study, we added resveratrol in normal glucose or high glucose medium and provide evidences that resveratrol protects against high-glucose triggered oxidative stress and cell senescence. Moreover, mice were fed with standard diet, standard diet plus resveratrol, high-fat diet or high-fat diet plus resveratrol for 3 months, and results show that resveratrol treatment prevents high-fat diet induced renal pathological damage by activating SIRT1, a key member in the mammalian sirtuin family that response to calorie restriction life-extension method. This research confirms the potential role of resveratrol in the treatment of renal diseases and may provide an effective and convenient method to mimic the beneficial effects of calorie restriction.
Collapse
Affiliation(s)
- Nannan Zhang
- MOE Key Laboratory of Protein Sciences, Department of Pharmacology, School of Medicine, Tsinghua University
| | | | | | | | | |
Collapse
|
12
|
Li Y, Zhou Q, Pei C, Liu B, Li M, Fang L, Sun Y, Li Y, Meng S. Hyperglycemia and Advanced Glycation End Products Regulate miR-126 Expression in Endothelial Progenitor Cells. J Vasc Res 2016; 53:94-104. [DOI: 10.1159/000448713] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/26/2016] [Indexed: 11/19/2022] Open
|
13
|
Bartosova M, Rudolf A, Pichl S, Schmidt K, Okun JG, Straub BK, Rutkowski R, Witowski J, Schmitt CP. Increased storage and secretion of phosphatidylcholines by senescent human peritoneal mesothelial cells. Clin Exp Nephrol 2015; 20:544-551. [PMID: 26527208 DOI: 10.1007/s10157-015-1192-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/24/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND/AIMS Human peritoneal mesothelial cells (HPMC) secrete phosphatidylcholines (PC) which form a lipid bilayer lining the peritoneum. They prevent frictions and adhesions and act as a barrier to the transport of water-soluble solutes while permitting water flux. PC may play an essential role in peritoneal integrity and function, the role of PD induced HPMC senescence on PC homeostasis, however, is unknown. METHODS HPMC cell lines were isolated from four non-uremic patients. Expression of the three PC synthesis genes (rt-PCR), and cellular storage and secretion of PC (ESI-mass-spectrometry) were analyzed in young and senescent HPMC (>Hayflick-limit). RESULTS Senescent cells displayed significantly altered morphology; flow cytometry demonstrated extensive staining for senescence-associated beta galactosidase. Nine different PC were detected in HPMC with palmitoyl-myristoyl phosphatidylcholine (PMPC) being most abundant. In senescent HPMC mRNA expression of the three key PC synthesis genes was 1.5-, 2.4- and 6-fold increased as compared to young HPMC, with the latter, phosphatidylcholine cytidylyltransferase, being rate limiting. Intracellular storage of the nine PC was 75-450 % higher in senescent vs. young HPMC, PC secretion rates were 100-300 % higher. Intracellular PC concentrations were not correlated with the PC secretion rates. Electron microscopy demonstrated lamellar bodies, the primary storage site of PC, in senescent but not in young cells. CONCLUSION Senescent HPMC store and secrete substantially more PC than young cells. Our findings indicate a novel protective mechanism, which should counteract peritoneal damage induced by chronic exposure to PD fluids.
Collapse
Affiliation(s)
- Maria Bartosova
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | | | - Sebastian Pichl
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Kathrin Schmidt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Jürgen G Okun
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Beate K Straub
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | - Claus P Schmitt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany.
| |
Collapse
|
14
|
Pirmoradi L, Mohammadi MT, Safaei A, Mesbah F, Dehghani GA. Does the relief of glucose toxicity act as a mediator in proliferative actions of vanadium on pancreatic islet beta cells in streptozocin diabetic rats? IRANIAN BIOMEDICAL JOURNAL 2015; 18:173-80. [PMID: 24842144 PMCID: PMC4048482 DOI: 10.6091/ibj.1329.2014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background: Data shows vanadium protects pancreatic beta cells (BC) from diabetic animals. Whether this effect is direct or through the relief of glucose toxicity is not clear. This study evaluated the potential effect of oral vanadyl sulfate (vanadium) on glycemic status and pancreatic BC of normal and diabetic rats. Methods: Rats were divided into five groups of normal and diabetic. Diabetes was induced with streptozocin (40 mg/kg, i.v.). Normal rats used water (CN) or vanadium (1 mg/ml VOSO4, VTN). Diabetic rats used water (CD), water plus daily neutral protamine Hagedorn insulin injection (80 U/kg, ITD) or vanadium (VTD). Blood samples were taken for blood glucose (BG, mg/dL) and insulin (ng/dL) measurements. After two months, the pancreata of sacrificed rats were prepared for islet staining. Results: Pre-treated normal BG was 88 ± 2, and diabetic BG was 395 ± 9. The final BG in CD, VTD, and ITD was 509 ± 22, 138 ± 14, and 141 ± 14, respectively. Insulin in VTN (0.75 ± 0.01) and VTD (0.78 ± 0.01) was similar, higher than CD (0.51 ± 0.07) but lower than CN (2.51 ± 0.02). VTN islets compared to CN had larger size and denser central core insulin immunoreactivity with plentiful BC. CD and ITD islets were atrophied and had scattered insulin immunoreactivity spots and low BC mass. VTD islets were almost similar to CN. Conclusion: Besides insulin-like activity, vanadium protected pancreatic islet BC, and the relief of glucose toxicity happening with vanadium had a little role in this action.
Collapse
Affiliation(s)
- Leila Pirmoradi
- Dept. of Physiology, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Akbar Safaei
- Dept. of Pathology, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fakhardin Mesbah
- Dept. of Anatomy, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholam Abbas Dehghani
- Dept. of Physiology, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran.,Dept. of Pathology, Nemazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
15
|
Mesothelial cell: a multifaceted model of aging. Ageing Res Rev 2013; 12:595-604. [PMID: 23415666 DOI: 10.1016/j.arr.2013.01.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 01/30/2013] [Indexed: 01/13/2023]
Abstract
Human peritoneal mesothelial cells (HPMCs) dominate within the peritoneal cavity and thus play a central role in a variety of intraperitoneal processes, including the transport of water and solutes, inflammation, host response, angiogenesis, and extracellular matrix remodeling. In addition, they contribute to the development of abdominal adhesions, peritonitis, endometriosis, cancer cell metastases, and peritoneal dialysis complications. For less than a decade the primary cultures of omental HPMCs have also been used as an experimental tool in studies on cellular aging. This paper provides the first comprehensive overview of the current state of art on molecular mechanisms underlying HPMC senescence in vitro. Special attention is paid to the causes of the very fast dynamics of HPMC senescence, and in particular to the role of non-telomeric DNA damage, the autocrine activity of TGF-β1, and the causative effects of oxidative stress. In addition, some clinical manifestations of HPMC senescence will be discussed, including its interplay with organismal aging, peritoneal dialysis, and cancer progression.
Collapse
|
16
|
l-Cysteine improves survival and growth of mesothelial cells after freezing. Cell Biol Int 2013; 33:1155-64. [DOI: 10.1016/j.cellbi.2009.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 06/29/2009] [Accepted: 08/17/2009] [Indexed: 11/18/2022]
|
17
|
Higashi Y, Abe K, Kuzumoto T, Hara T, Miyamoto K, Murata T, Ishikawa E, Nomura S, Horiuchi T. Characterization of peritoneal dialysis effluent-derived cells: diagnosis of peritoneal integrity. J Artif Organs 2012; 16:74-82. [PMID: 23274627 DOI: 10.1007/s10047-012-0673-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 11/01/2012] [Indexed: 11/26/2022]
Abstract
To assess the integrity of the peritoneal membrane, we characterized the functionality of the cellular components derived from peritoneal dialysis effluent (PDE). About 3 % of all cells collected from the PDE attached to the plastic dish, and 97.1 ± 3.1 % of the adherent cells expressed CK-18 (PDE-HPMC). A typical cobble-stone-like morphology under neutralized PD solution was obtained over 65 out of 74 primary cultures (88 %) while only 53 % under acidic PD solution in a previous report by Yanez-Mo et al. However, 26.6 ± 10.3 % of PDE-HPMC expressed the EMT marker α-SMA. Transepithelial resistance (TER) as a marker of cell polarity was 34 % lower than that of omentum-derived(OM)-HPMC. We found a significant decrease in the rate of PDE-HPMC proliferation compared to OM-HPMC, accompanied by a significant increase of cell area within the tertiary passage. Comparison of TER, α-SMA and SA-β-Gal between CAPD durations suggests that cell polarity weakens with increased duration of CAPD, reflecting the occurrence of EMT and cell senescence. We conclude that functional characterization of cellular components in PDE reflects how well the peritoneum is preserved.
Collapse
Affiliation(s)
- Yo Higashi
- Division of Chemistry for Materials, Faculty of Engineering, Graduate School of Mie University, 1577 Kurima-Machiyacho, Tsu, Mie, 514-8507, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zhang X, Wang J, Fan Y, Yang L, Wang L, Ma J. Zinc supplementation attenuates high glucose-induced epithelial-to-mesenchymal transition of peritoneal mesothelial cells. Biol Trace Elem Res 2012; 150:229-35. [PMID: 22639383 DOI: 10.1007/s12011-012-9451-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 05/10/2012] [Indexed: 01/12/2023]
Abstract
Zinc (Zn) plays an important role in preventing many types of epithelial-to-mesenchymal transition (EMT)-driven fibrosis in vivo. But its function in the EMT of the peritoneal mesothelial cells (PMCs) remains unknown. Here, we studied the Zn effect on the high glucose (HG)-induced EMT in the rat PMCs (RPMCs) and the underlying molecular mechanisms. We found that Zn supplementation significantly inhibited TGF-β1 and ROS production, and attenuated the HG-induced EMT in the RPMCs, likely through inhibition of MAPK, NF-κB, and TGF-β/Smad pathways.
Collapse
Affiliation(s)
- Xiuli Zhang
- Department of Nephrology, the First Affiliated Hospital, China Medical University, 155th Nanjing North Street, Shenyang, Liaoning, 110001, People's Republic of China
| | | | | | | | | | | |
Collapse
|
19
|
Yokoi H, Kasahara M, Mori K, Kuwabara T, Toda N, Yamada R, Namoto S, Yamamoto T, Seki N, Souma N, Yamaguchi T, Sugawara A, Mukoyama M, Nakao K. Peritoneal fibrosis and high transport are induced in mildly pre-injured peritoneum by 3,4-dideoxyglucosone-3-ene in mice. Perit Dial Int 2012; 33:143-54. [PMID: 23123666 DOI: 10.3747/pdi.2011.00033] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Peritoneal dialysis (PD) solution contains high concentrations of glucose and glucose degradation products (GDPs). One of several GDPs--3,4-dideoxyglucosone-3-ene (3,4-DGE)--was recently identified as the most reactive and toxic GDP in PD fluids. In vitro, 3,4-DGE has been shown to induce mesothelial cell damage; however, its role in peritoneal fibrosis in vivo remains unclear. In the present study, we intraperitoneally administered chlorhexidine gluconate (CG) for mild peritoneal injury, and we then injected 3,4-DGE [38 μmol/L (low concentration) or 145 μmol/L (high concentration)] 5 times weekly for 4 weeks. Significant thickening of the parietal peritoneal membrane was observed only when treatment with low or high concentrations of 3,4-DGE occurred after CG administration, but not when either CG or 3,4-DGE alone was given. The combination of CG and 3,4-DGE also caused upregulation of messenger RNA expression of transforming growth factor β1, connective tissue growth factor, fibronectin, collagen type 1 α1 chain, alpha smooth muscle actin (α-SMA), vascular endothelial growth factor 164, NADPH oxidase 1 and 4, p22phox, p47phox, and gp91phox in peritoneal tissue. Treatment with CG alone was sufficient to cause significant F4/80-positive macrophage infiltration, appearance of α-SMA-positive cells, and vessel formation in the submesothelial layer. Addition of 3,4-DGE markedly enhanced those changes and induced apoptosis, mainly in leukocytes. The concentration of 3,4-DGE in the abdominal cavity declined more rapidly in CG-treated mice than in PBS-treated mice. Peritoneal membrane permeability determined by peritoneal equilibration test showed high transport conditions in peritoneum treated with both CG and 3,4-DGE. These results indicate that, when mild peritoneal damage is already present, 3,4-DGE causes peritoneal thickening and fibrosis, resulting in deterioration of peritoneal membrane function.
Collapse
Affiliation(s)
- Hideki Yokoi
- Department of Medicine and Clinical Science,Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Zhang S, Cai G, Fu B, Feng Z, Ding R, Bai X, Liu W, Zhuo L, Sun L, Liu F, Chen X. SIRT1 is required for the effects of rapamycin on high glucose-inducing mesangial cells senescence. Mech Ageing Dev 2012; 133:387-400. [PMID: 22561310 DOI: 10.1016/j.mad.2012.04.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2011] [Revised: 04/13/2012] [Accepted: 04/26/2012] [Indexed: 01/30/2023]
Abstract
The mTOR deregulation has a role in chronic kidney disease including diabetic nephropathy. SIRT1 is an important participant in renal cytoprotective responses to aging and stress. However, whether both mTOR and SIRT1 are involved in high glucose-inducing mesangial cells (MCs) senescence still remains to be explored. Hence we investigate the potential functional interrelationship between these two proteins in high glucose-inducing MCs senescence. High glucose increased mTOR expression and activity, but decreased SIRT1 expression and activity. The level of mTOR was increased significantly, while the SIRT1 expression and activity was declined significantly with serial cell culture passage. The siRNA-SIRT1 and nicotinamide promoted MCs senescence. NAD or resveratrol arrested high glucose-inducing MCs senescence. Meanwhile, the effects of NAD or resveratrol on high glucose-inducing MCs senescence were also completely blocked by SiRNA-SIRT1. Rapamycin arrested MCs senescence induced by high glucose and prevented MCs senescence with serial cell culture passage, and meanwhile increased the SIRT1 expression and activity. Moreover, the effects of rapamycin on MCs senescence induced by high glucose were also completely blocked by treating cells with niacinamide or siRNA-SIRT1. These findings provide support for the hypothesis that SIRT1 is required for the effects of rapamycin on high glucose-inducing MCs senescence.
Collapse
Affiliation(s)
- Sifang Zhang
- Department of Nephrology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Hong MN, Kim BC, Ko YG, Lee YS, Hong SC, Kim T, Pack JK, Choi HD, Kim N, Lee JS. Effects of 837 and 1950 MHz radiofrequency radiation exposure alone or combined on oxidative stress in MCF10A cells. Bioelectromagnetics 2012; 33:604-11. [PMID: 22549623 DOI: 10.1002/bem.21731] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 03/31/2012] [Indexed: 12/13/2022]
Abstract
The aim of this study was to determine whether the exposure to either single or multiple radio-frequency (RF) radiation frequencies could induce oxidative stress in cell cultures. Exposures of human MCF10A mammary epithelial cells to either a single frequency (837 MHz alone or 1950 MHz alone) or multiple frequencies (837 and 1950 MHz) were conducted at specific absorption rate (SAR) values of 4 W/kg for 2 h. During the exposure period, the temperature in the exposure chamber was maintained isothermally. Intracellular levels of reactive oxygen species (ROS), the antioxidant enzyme activity of superoxide dismutase (SOD), and the ratio of reduced/oxidized glutathione (GSH/GSSG) showed no statistically significant alterations as the result of either single or multiple RF radiation exposures. In contrast, ionizing radiation-exposed cells, used as a positive control, showed evident changes in all measured biological endpoints. These results indicate that single or multiple RF radiation exposure did not elicit oxidative stress in MCF10A cells under our exposure conditions.
Collapse
Affiliation(s)
- Mi-Na Hong
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Mikuła-Pietrasik J, Kuczmarska A, Kucińska M, Murias M, Wierzchowski M, Winckiewicz M, Staniszewski R, Bręborowicz A, Książek K. Resveratrol and its synthetic derivatives exert opposite effects on mesothelial cell-dependent angiogenesis via modulating secretion of VEGF and IL-8/CXCL8. Angiogenesis 2012; 15:361-76. [PMID: 22451299 PMCID: PMC3409370 DOI: 10.1007/s10456-012-9266-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 03/13/2012] [Indexed: 11/19/2022]
Abstract
We examined the effect of resveratrol (RVT) and its two derivatives (3,3′,4,4′-tetrahydroxy-trans-stilbene and 3,3′,4,4′,5,5′-hexahydroxy-trans-stilbene) on human peritoneal mesothelial cell (HPMC)-dependent angiogenesis in vitro. To this end, angiogenic activity of endothelial cells (HUVEC, HMVEC, and HMEC-1) was monitored upon their exposure to conditioned medium (CM) from young and senescent HPMCs treated with stilbenes or to stilbenes themselves. Results showed that proliferation and migration of endothelial cells were inhibited in response to indirect (HPMC-dependent) or direct RVT activity. This effect was associated with decreased secretion of VEGF and IL-8/CXCL8 by HPMCs treated with RVT, which confirmed the experiments with recombinant forms of these angiogenic agents. Angiogenic activity of endothelial cells treated with CM from HPMCs exposed to RVT analogues was more effective. Improved migration was particularly evident in cells exposed to CM from senescent HPMCs. Upon direct treatment, RVT derivatives stimulated proliferation (but not migration) of HUVECs, and failed to affect the behaviour of HMVEC and HMEC-1 cells. These compounds stimulated production of VEGF and IL-8/CXCL8 by HPMCs. Studies with neutralizing antibodies against angiogenic factors revealed that augmented angiogenic reactions of endothelial cells exposed to CM from HPMC treated with RVT analogues were related to enhanced production of VEGF and IL-8/CXCL8. Collectively, these findings indicate that RVT and its synthetic analogues divergently alter the secretion of the angiogenic factors by HPMCs, and thus modulate HPMC-dependent angiogenic responses in the opposite directions. This may have implications for the attempts of practical employment of the stilbenes for treatment of pathologies proceeding with abnormal vascularisation of the peritoneal tissue.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Pathophysiology, Poznań University of Medical Sciences, Święcickiego 6 Str., 60-781, Poznań, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Glucose but not N-acetylglucosamine accelerates in vitro senescence of human peritoneal mesothelial cells. Int J Artif Organs 2011; 34:489-94. [PMID: 21725930 DOI: 10.5301/ijao.2011.8467] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2011] [Indexed: 11/20/2022]
Abstract
BACKGROUND Preservation of the mesothelial cells (MCs) is crucial for longevity of the peritoneal dialysis membrane. Glucose accelerates aging of MC and we tested whether N-acetylglucosamine (NAG) has an identical effect. METHODS Replicative aging of MCs was studied during 10 passages performed every three days in cells cultured in standard medium or in medium supplemented with Glucose 30 mmol/L or NAG 30 mmol/L. Changes in population doubling time and ß-galactosidase activity were used as an index of aging and compared with other cellular parameters. RESULTS Repeated passages of MC cause their aging, as reflected by prolongation of the population doubling time, increased ß-galactosidase activity, oxidative stress and release of cytokines. Healing of injured mesothelial monolayer is impaired in senescent cells. Glucose accelerates in vitro aging of MC, whereas NAG does not cause this effect. CONCLUSIONS Replacement of glucose with NAG in the dialysis fluid can slow down aging of MC.
Collapse
|
24
|
Cao D, Zhang M, Jiang C, Xue L, Sun C. Protection of Tanshinone IIA to human peritoneal mesothelial cells (HPMC) through delaying cellular senescence induced by high glucose. Ren Fail 2011; 34:88-94. [PMID: 22032700 DOI: 10.3109/0886022x.2011.623497] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Long-term peritoneal exposure to high glucose in the peritoneal dialysis (PD) solution may potentiate the development of peritoneal fibrosis in PD patients. The most important factor leading to peritoneal fibrosis may be injury of human peritoneal mesothelial cells (HPMC). Little is known about senescence of HPMC. It has been reported that Tanshinone IIA can ameliorate fibrosis. Whether Tanshinone IIA may delay senescence and protect HPMC against high glucose is not clear. The aim of this study is to investigate the protective role of Tanshinone IIA in senescence of HPMC induced by high glucose. METHODS HPMC were isolated and cultured with Roswell Park Memorial Institute 1640 medium containing high glucose concentrations (2.5%) and Tanshinone IIA (50 μmol/L and 100 μmol/L). The effects of high glucose and Tanshinone IIA on cellular senescence of HPMC were examined by observing cell generation, growth rate, cell cycle, positive rate of senescence-associated β-galactosidase (SA-β-gal) staining, telomere length, and expression of p16 and p21. RESULTS Compared with the control cells, HPMC cultured in high glucose showed decreased cell generations by four to five and suppression of growth rate, and the cell cycle was stopped at G1 phase. The positive rate of SA-β-gal staining was increased; the telomere length was shortened; and the expressions of p16 and p21 were increased. The characteristics in morphology of senescent cells appeared earlier. Tanshinone IIA may delay the process of senescence of HPMC induced by high glucose by increasing cell generations and growth rate, decreasing the rate of G1 phase and the positive rate of SA-β-gal staining, lengthening the telomere, and decreasing the expression of p16 and p21. CONCLUSIONS Tanshinone IIA may protect HPMC through delaying cellular senescence induced by high glucose.
Collapse
Affiliation(s)
- Dongwei Cao
- Department of Nephrology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, PR China
| | | | | | | | | |
Collapse
|
25
|
Chepelev NL, Bennitz JD, Wright JS, Smith JC, Willmore WG. Oxidative modification of citrate synthase by peroxyl radicals and protection with novel antioxidants. J Enzyme Inhib Med Chem 2010; 24:1319-31. [PMID: 19795928 DOI: 10.3109/14756360902852586] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In mammals, aging is linked to a decline in the activity of citrate synthase (CS; E.C. 2.3.3.1), the first enzyme of the citric acid cycle. We used 2,2'-azobis(2-amidinopropane) dihydrochloride (AAPH), a water-soluble generator of peroxyl and alkoxyl radicals, to investigate the susceptibility of CS to oxidative damage. Treatment of isolated mitochondria with AAPH for 8-24 h led to CS inactivation; however, the activity of aconitase, a mitochondrial enzyme routinely used as an oxidative stress marker, was unaffected. In addition to enzyme inactivation, AAPH treatment of purified CS resulted in dityrosine formation, increased protein surface hydrophobicity, and loss of tryptophan fluorescence. Propyl gallate, 1,8-naphthalenediol, 2,3-naphthalenediol, ascorbic acid, glutathione, and oxaloacetate protected CS from AAPH-mediated inactivation, with IC(50) values of 9, 14, 34, 37, 150, and 160 muM, respectively. Surprisingly, the antioxidant epigallocatechin gallate offered no protection against AAPH, but instead caused CS inactivation. Our results suggest that the current practice of using the enzymatic activity of CS as an index of mitochondrial abundance and the use of aconitase activity as an oxidative stress marker may be inappropriate, especially in oxidative stress-related studies, during which alkyl peroxyl and alkoxyl radicals can be generated.
Collapse
|
26
|
Yang WH, Park SY, Ji S, Kang JG, Kim JE, Song H, Mook-Jung I, Choe KM, Cho JW. O-GlcNAcylation regulates hyperglycemia-induced GPX1 activation. Biochem Biophys Res Commun 2009; 391:756-61. [PMID: 19944066 DOI: 10.1016/j.bbrc.2009.11.133] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 11/19/2009] [Indexed: 10/20/2022]
Abstract
Hyperglycemia induces activation of glutathione peroxidase 1 (GPX1), an anti-oxidant enzyme essential for cell survival during oxidative stress. However, the mechanism of GPX1 activation is unclear. Here, we report that hyperglycemia-induced protein glycosylation by O-linked N-acetylglucosamine (O-GlcNAc) is crucial for activation of GPX1 and for its binding to c-Abl and Arg kinases. GPX1 itself is modified with O-GlcNAc on its C-terminus. We also demonstrate that pharmacological injection of the O-GlcNAcase inhibitor NTZ induces GPX1 activation in the mouse liver. Our findings suggest a crucial role for GPX1 and its O-GlcNAc modification in hyperglycemia and diabetes mellitus.
Collapse
Affiliation(s)
- Won Ho Yang
- Department of Biology, Yonsei University, Seoul 120-749, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Katakura Y, Udono M, Katsuki K, Nishide H, Tabira Y, Ikei T, Yamashita M, Fujiki T, Shirahata S. Protein kinase C delta plays a key role in cellular senescence programs of human normal diploid cells. J Biochem 2009; 146:87-93. [PMID: 19279193 DOI: 10.1093/jb/mvp046] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
In the present study, we clarified that transforming growth factor beta (TGF-beta) induces cellular senescence in human normal diploid cells, TIG-1, and identified protein kinase Cs (PKCs) as downstream mediators of TGF-beta-induced cellular senescence. Among PKCs, we showed that PKC-delta induced cellular senescence in TIG-1 cells and was activated in replicatively and prematurely senescent TIG-1 cells. The causative role of PKC-delta in cellular senescence programs was demonstrated using a kinase negative PKC-delta and small interfering RNA against PKC-delta. Furthermore, PKC-delta was shown to function in human telomerase reverse transcriptase (hTERT) gene repression. These results indicate that PKC-delta plays a key role in cellular senescence programs, and suggest that the induction of senescence and hTERT repression are coordinately regulated by PKC-delta.
Collapse
Affiliation(s)
- Yoshinori Katakura
- Department of Genetic Resources Technology, Kyushu University, Fukuoka, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ksiazek K, Mikula-Pietrasik J, Korybalska K, Dworacki G, Jörres A, Witowski J. Senescent peritoneal mesothelial cells promote ovarian cancer cell adhesion: the role of oxidative stress-induced fibronectin. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1230-40. [PMID: 19246646 DOI: 10.2353/ajpath.2009.080613] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Adhesion of ovarian cancer cells to the peritoneal mesothelium is a key step in the malignant progression of the disease. In an in vitro study, we showed that the adherence of ovarian cancer cells (of the OVCAR-3, SKOV-3, and A2780 cell lines) to senescent human omentum-derived peritoneal mesothelial cells (HOMCs) was greater than to early passage cells. The process was mediated primarily by the increased interaction of the alpha5beta1 integrin on cancer cells with HOMC-associated fibronectin (FN). In comparison with early passage HOMCs, senescent cells exhibited increased FN mRNA expression levels and produced significantly more FN. To assess the effect of senescence-associated oxidative stress on FN release, HOMCs were rendered senescent by exposure to an oxidant, tert-butyl hydroperoxide. Treatment with tert-butyl hydroperoxide resulted in a significant increase in HOMC FN mRNA and protein expression levels. The effect of oxidative stress on FN synthesis was found to be mediated by transforming growth factor-beta1, whose signaling pathway was controlled at upstream and downstream levels by p38 MAPK. The activity of p38 MAPK increased markedly in senescent HOMCs. Treatment of HOMCs with antioxidants significantly attenuated senescence-associated increases in p38 MAPK activity, production of both transforming growth factor-beta1 and FN, and ovarian cancer cell adhesion. These data indicate that oxidative stress that accompanies senescence may increase FN production by HOMCs and thus facilitate binding and dissemination of ovarian cancer cells.
Collapse
Affiliation(s)
- Krzysztof Ksiazek
- Department of Pathophysiology, Poznan University of Medical Sciences, Swiecickiego 6, 60-781 Poznan, Poland
| | | | | | | | | | | |
Collapse
|
29
|
Kuo HT, Chen HW, Hsiao HH, Chen HC. Heat shock response protects human peritoneal mesothelial cells from dialysate-induced oxidative stress and mitochondrial injury. Nephrol Dial Transplant 2009; 24:1799-809. [PMID: 19126751 DOI: 10.1093/ndt/gfn718] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Chronic peritoneal dialysis (PD) is one of the major therapies for uremic patients. However, the peritoneal mesothelial cells (PMCs) are subject to the injury by bioincompatible dialysates. The aim of this study is to investigate the protective roles and mechanisms of heat shock response in PMCs. METHODS Primary cultured human PMCs (HPMCs) were subjected to commercial peritoneal dialysates. The cell viability was assayed by MTT test and Annexin V assay. The expression of HSPs was detected by Western blots analysis. Intracellular hydrogen peroxide and superoxide anion were detected using H(2)DCFDA and dHE probe, respectively, with flow cytometry. The mitochondrial membrane potential (DeltaPsim) of HPMCs was evaluated using JC1 probe with flow-cytometry. RESULTS Exposure of HPMCs to 1.5%, 2.5%, and 4.25% dextrose, and 7.5% icodextrin dialysates, respectively, for 60 min resulted in significantly accumulation of intracellular reactive oxygen species (ROS), DeltaPsim loss, and cell death in HPMCs. Amino acid dialysates exhibited no significant cytotoxicity. Adjusting the acidity in 1.5% dextrose and icodextrin dialysate significantly attenuated the dialysate-induced ROS generation and cell death in HPMCs. Heat pretreatment (41 degrees C, 30 minutes), which induced HSP 27 and 72 syntheses, significantly attenuated the dialysate-induced intracellular ROS accumulation, Dym loss, and cell death in HPMCs. CONCLUSIONS In conclusion, the acidic bioincompatible dialysates induce oxidative stress, DeltaPsim loss, and subsequent cell death in HPMCs. Amino acid dialysates is more biocompatible than glucose and icodextrin dialysates to HPMCs. Heat shock response protects HPMCs from the bioincompatible dialysates-induced cellular damage.
Collapse
Affiliation(s)
- Hung-Tien Kuo
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Faculty of Renal Care, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | |
Collapse
|
30
|
Ksiazek K, Mikula-Pietrasik J, Olijslagers S, Jörres A, von Zglinicki T, Witowski J. Vulnerability to oxidative stress and different patterns of senescence in human peritoneal mesothelial cell strains. Am J Physiol Regul Integr Comp Physiol 2008; 296:R374-82. [PMID: 19036828 DOI: 10.1152/ajpregu.90451.2008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Both the ascites fluid-derived mesothelial cell line LP-9 and primary cultures of human omentum-derived mesothelial cells (HOMCs) are commonly used in experimental studies. However, they seem to have a different replicative potential in vitro. In the present study, we have attempted to determine the causes of this discrepancy. HOMCs were found to divide fewer times and enter senescence earlier than LP-9 cells. This effect was coupled with earlier increases in the expression of senescence-associated-beta-galactosidase and cell cycle inhibitors p16INK4a and p21WAF1. Moreover, almost 3 times as many early-passage HOMCs as LP-9 cells bore senescence-associated DNA damage foci. In sharp contrast to LP-9 cells, the foci present in HOMCs localized predominantly outside the telomeres, and the HOMC telomere length did not significantly shorten during senescence. Compared with LP-9 cells, HOMCs were found to enter senescence with significantly lower levels of lipofuscin and damaged DNA, and markedly decreased glutathione contents. In addition, early-passage HOMCs generated significantly more reactive oxygen species either spontaneously or in response to exogenous oxidants. These results indicate that compared with LP-9 cells, HOMCs undergo stress-induced telomere-independent premature senescence, which may result from increased vulnerability to oxidative DNA injury.
Collapse
Affiliation(s)
- Krzysztof Ksiazek
- Department of Pathophysiology, Poznan Univ. of Medical Sciences, Swiecickiego 6, 60-781 Poznan, Poland
| | | | | | | | | | | |
Collapse
|
31
|
Witowski J, Ksiazek K, Jörres A. Glucose-Induced Mesothelial Cell Senescence and Peritoneal Neoangiogenesis and Fibrosis. Perit Dial Int 2008. [DOI: 10.1177/089686080802805s07] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Janusz Witowski
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
- Department of Pathophysiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Krzysztof Ksiazek
- Department of Pathophysiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Achim Jörres
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| |
Collapse
|
32
|
Ksiazek K, Piatek K, Witowski J. Impaired response to oxidative stress in senescent cells may lead to accumulation of DNA damage in mesothelial cells from aged donors. Biochem Biophys Res Commun 2008; 373:335-9. [DOI: 10.1016/j.bbrc.2008.06.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2008] [Accepted: 06/10/2008] [Indexed: 01/09/2023]
|
33
|
Bender TO, Witowski J, Ksiazek K, Jörres A. Comparison of icodextrin- and glucose-based peritoneal dialysis fluids in their acute and chronic effects on human peritoneal mesothelial cells. Int J Artif Organs 2008; 30:1075-82. [PMID: 18203069 DOI: 10.1177/039139880703001207] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Icodextrin-based peritoneal dialysis fluids (PDFs) display several features that may potentially improve their biocompatibility compared to conventional glucose-containing solutions. So far, however, the studies assessing the biocompatibility profile of icodextrin toward human peritoneal mesothelial cells (HPMC) has produced mixed results. The present study was performed to examine the acute and chronic impact of icodextrin on HPMC in vitro in comparison with standard glucose-based PDF. METHODS Omentum-derived HPMC were either acutely pre-exposed to or incubated chronically (for up to 10 days) in the presence of icodextrin-PDF. Parallel cultures were treated with conventional PDFs containing either 1.5% or 4.25% glucose. All fluids were tested at neutral pH. HPMC were assessed for viability, proliferation, IL-6 secretion and generation of reactive oxygen species (ROS). RESULTS Incubation in the presence of icodextrin-PDF significantly reduced HPMC proliferation in a manner similar to that of 1.5% glucose-PDF. In addition, exposure to icodextrin-PDF impaired viability and IL-6 release from HPMC. This effect occurred both after the short pre-treatment with neat icodextrin-PDF for 1-4 hours and after prolonged incubation (up to 10 days) in media supplemented with icodextrin-PDF (1:1). The dysfunction of icodextrin-treated HPMC was of the magnitude that was between the effects exerted by 1.5%- and 4.25%-glucose PDF. Furthermore, exposure of HPMC to icodextrin-PDF induced a dose-dependent increase in ROS generation which was comparable to that produced by 1.5%-glucose PDF. CONCLUSION Exposure to icodextrin-PDF may impair viability and function of HPMC. The detrimental effects of icodextrin-PDF are at least as serious as those produced by conventional heat-sterilized low glucose-based PDF.
Collapse
Affiliation(s)
- T O Bender
- Department of Nephrology and Medical Intensive Care, Charité University Hospital, Virchow Clinic Campus, Berlin, Germany
| | | | | | | |
Collapse
|
34
|
Ksiazek K, Passos JF, Olijslagers S, von Zglinicki T. Mitochondrial dysfunction is a possible cause of accelerated senescence of mesothelial cells exposed to high glucose. Biochem Biophys Res Commun 2008; 366:793-9. [DOI: 10.1016/j.bbrc.2007.12.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Accepted: 12/04/2007] [Indexed: 01/31/2023]
|
35
|
Ciszewicz M, Wu G, Tam P, Polubinska A, Breborowicz A. Changes in peritoneal mesothelial cells phenotype after chronic exposure to glucose or N-acetylglucosamine. Transl Res 2007; 150:337-42. [PMID: 18022595 DOI: 10.1016/j.trsl.2007.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Revised: 07/10/2007] [Accepted: 07/13/2007] [Indexed: 10/22/2022]
Abstract
Glucose is commonly used as an osmotic solute in peritoneal dialysis fluids despite vast knowledge about deleterious peritoneal and systemic effects of that solute. N-acetylglucosamine (NAG) is a solute of the comparable size to glucose, with strong anti-inflammatory properties. We compared the chronic in vitro effect of both solutes on phenotype of peritoneal mesothelial cells. Experiments were performed of primary cultures of human peritoneal mesothelial cells, which were cultured over 4 weeks in medium supplemented either with glucose 45 mmol/L (GLU) or with NAG 45 mmol/L (NAG). Generation of reactive oxygen species (ROS) in cells was studied, as well as their ability to proliferate, synthesis of cytokines, fibronectin, and factors regulating peritoneal fibrinolysis. Cells cultured in the presence of glucose 45 mmol/L generated more ROS (+73% vs control, P < 0.01), whereas NAG did not stimulate generation of ROS. GLU caused hypertrophy of mesothelial cells (+53% vs control, P < 0.001) and prolonged their population doubling time (+16% vs control, P < 0.01); NAG did not cause significant changes in these parameters. Healing of mesothelial monolayer after mechanical injury was impaired in GLU treated cells: (-48% vs control, P < 0.001 and -40% vs NAG, P < 0.05). Synthesis of Il-6, vascular endothelial growth factor (VEGF), transforming growth factor beta (TGFbeta), and fibronectin was higher in GLU group as compared with control: + 86%, P < 0.001, +38%, P < 0.05, +51%, P < 0.001, +38%, P < 0.05, respectively. In the presence of NAG, these parameters were comparable with the control group, but at the same time NAG stimulated synthesis of hyaluronan: +116% versus control, P < 0.001 and + 96% versus GLU, P < 0.01. Treatment with GLU resulted in decline of tissue plasminogen activator/plasminogen activator inhibitor-1 (t-PA/PAI-1) ratio by 23% versus control, P < 0.001, whereas NAG increased that parameter by 43%, P < 0.01 versus control. Glucose, contrary to NAG, induces oxidative stress and proinflammatory and profibrotic changes in mesothelial cells. NAG seems to be more biocompatible osmotic solute than glucose.
Collapse
Affiliation(s)
- Maria Ciszewicz
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | | | | |
Collapse
|
36
|
Ksiazek K, Passos JF, Olijslagers S, Saretzki G, Martin-Ruiz C, von Zglinicki T. Premature senescence of mesothelial cells is associated with non-telomeric DNA damage. Biochem Biophys Res Commun 2007; 362:707-11. [PMID: 17720141 DOI: 10.1016/j.bbrc.2007.08.047] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Accepted: 08/08/2007] [Indexed: 11/19/2022]
Abstract
Human peritoneal mesothelial cells (HPMCs) senesce in vitro after barely few population doublings. In this report, we show that senescence of HPMCs is associated with increased accumulation of gamma-H2A.X foci, which reveal DNA double-strand breaks. Of note, already early-passage cultures contain a considerable fraction (44+/-10%) of cells bearing gamma-H2A.X foci. The gamma-H2A.X foci localize predominantly to non-telomeric DNA, either in young or senescent cells. Moreover, HPMCs seem to have unusually short telomeres (approximately 3.5 kbp) despite the presence of active telomerase. These telomeres do not shorten during senescence, but the activity of telomerase decreases to undetectable levels. In addition, senescence of HPMCs is associated with mitochondrial dysfunction, as manifested by increased production of reactive oxygen species and reduced mitochondrial membrane potential. These results may indicate that premature senescence of HPMCs is largely related to oxidative stress-induced DNA damage in non-telomeric regions of the genome.
Collapse
Affiliation(s)
- Krzysztof Ksiazek
- Department of Pathophysiology, University of Medical Sciences, Swiecickiego 6, 60781 Poznan, Poland.
| | | | | | | | | | | |
Collapse
|