1
|
Yu M, Zhao J, Shan Y, Dai H, Tang L, Sheng L, Zhang L, Sheng M. Genome-wide DNA methylation analysis of Astragalus on the intervention of ID2 promoter via PI3K/Akt signaling pathway in peritoneal fibrosis. Sci Rep 2025; 15:15786. [PMID: 40328830 PMCID: PMC12056223 DOI: 10.1038/s41598-025-96709-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Peritoneal dialysis (PD) is a successful renal replacement therapy for end-stage renal disease. Continuous infiltration of bioincompatible PD fluid causes mesothelial-mesenchymal transition (MMT) of peritoneal mesothelial cells (PMCs), leading to peritoneal fibrosis (PF). DNA methylation has been characterized as an important regulatory mechanism on multiple fibrosis. However, the mechanisms by which DNA methylation regulates PF are not fully understood resulting in a lack of disease-modifying drugs. Astragalus membranaceus (Astragalus) is naturally phytomedicine that has immunoregulation properties. The study aimed to elucidate the underlying mechanisms of Astragalus in regulating DNA methylation and anti-PF capabilities. In vivo PD rat models were established by inducing with high-glucose PD fluid and Astragalus was intraperitoneal injection. Global DNA methylation sequencing was used to compare the DNA methylation status between control and PF rat models. Methylation profiles and KEGG analysis were identified a possible methylated target gene and its correlation pathway. Through real-time PCR and western blotting, candidate markers and pathways were validated in vivo and in vitro. Chromatin immunoprecipitation and luciferase assays were used to identify the prediction of DNA methyltransferase (Dnmts) binding with methylated target gene. The functions of the validated pathways were further investigated using the knockdown or overexpression strategy. In vivo and in vitro, Astragalus treatment showed a protective effect against PF and Dnmts, characterized by improving pathological manifestation, ameliorating MMT markers, and reducing Dnmt1/3a proteins. Inhibitor of DNA-binding 2 (ID2) was investigated in target gene by integrating the mRNA and methylation profiles involved in PF and Astragalus treatment. PF induced the methylation of ID2 that resulted in recruitment of the Dnmt3a and decreased ID2 expression. The increased ID2 expression in response to Astragalus is a consequence of demethylation in promoter. In addition, phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway correlated with PF, knockdown or overexpression of ID2 regulated this pathway and MMT of PMCs. Astragalus ameliorated PF by targeting Dnmt3a mediated ID2 promoter via PI3K/Akt signaling pathway. The epigenetic regulation of DNA methylation existed the critical role in attenuating PF.
Collapse
Affiliation(s)
- Manshu Yu
- Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Junyi Zhao
- Institute of Literature in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yun Shan
- Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Huibo Dai
- Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Lei Tang
- Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Li Sheng
- Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Lu Zhang
- Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Meixiao Sheng
- Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
2
|
Hara D, Sasaki K, Doi S, Ike T, Maeda K, Yoshida M, Takahashi A, Osaki Y, Ishiuchi N, Maeoka Y, Doi T, Chiba T, Nakashima A, Masaki T. Targeting MLL1/WDR5-Mediated Epigenetic Regulation Mitigates Peritoneal Fibrosis by Reducing p16 INK4a. FASEB J 2025; 39:e70543. [PMID: 40232893 PMCID: PMC12059782 DOI: 10.1096/fj.202402382r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/24/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
Peritoneal fibrosis poses a significant challenge to the long-term efficacy of peritoneal dialysis (PD), with emerging evidence highlighting the role of cellular senescence in its pathogenesis. p16INK4a is a cell cycle regulator that has been implicated in cellular senescence. Mixed-lineage leukemia 1 (MLL1) forms a complex with WD-40 repeat protein 5 (WDR5) and exhibits histone H3K4 methyltransferase activity. We have previously shown that inhibition of the MLL1/WDR5 complex reduces p16INK4a expression and attenuates renal senescence after injury in mice. This study aimed to investigate whether inhibiting MLL1/WDR5 attenuates peritoneal senescence, inflammation, and fibrosis in both human samples and in mice with methylglyoxal (MGO)-induced peritoneal fibrosis (MGO-injected mice), while also exploring the associated underlying mechanisms. MLL1/WDR5, histone 3 lysine 4 trimethylation (H3K4me3), and p16INK4a expression were elevated in TGF-β1-stimulated human peritoneal mesothelial cells (HPMCs), non-adherent cells obtained from patients undergoing PD, and the submesothelial compact zones of MGO-injected mice. Notably, p16INK4a expression in these cells was positively correlated with the dialysate/plasma creatinine ratio. Treatment with the MLL1/WDR5 protein-protein interaction inhibitors MM-102 and OICR-9429 reduced H3K4me3 levels and p16INK4a expression, suppressing fibrosis in HPMCs as well as peritoneal fibrosis and inflammation in MGO-injected mice. These inhibitors also improved peritoneal function in MGO-injected mice. Additionally, we demonstrated that MLL1/WDR5-induced H3K4me3 directly regulates p16INK4a gene transcription, and that inhibiting MLL1/WDR5 reduces H3K4me3, thereby suppressing p16INK4a gene transcription. These findings suggest that targeting MLL1/WDR5 activation alleviates peritoneal senescence, inflammation, and fibrosis, highlighting its potential as a promising therapeutic strategy for peritoneal fibrosis.
Collapse
Affiliation(s)
- Daisuke Hara
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kensuke Sasaki
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Shigehiro Doi
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Takeshi Ike
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Kazuya Maeda
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Maria Yoshida
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Akira Takahashi
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Yosuke Osaki
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Naoki Ishiuchi
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Yujiro Maeoka
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Toshiki Doi
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
- Division of Nephrology, Ichiyokai Harada Hospital, Hiroshima, Japan
| | - Takuto Chiba
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
3
|
Shi Y, Hu Y, Li J, Chen H, Zhong Q, Ma X, Li X, Zhang S, Zhuang S, Liu N. Inhibition of Caspase-1 Suppresses GSDMD-mediated Peritoneal Mesothelial Cell Pyroptosis and Inflammation in Peritoneal Fibrosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409362. [PMID: 40018871 DOI: 10.1002/smll.202409362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/08/2025] [Indexed: 03/01/2025]
Abstract
Pyroptosis, belonging to programmed cell death, is shown to be mediated by gasdermin D (GSDMD) and gains more and more attention in innate immunity and multiple diseases. However, the role of GSDMD-mediated pyroptosis in peritoneal fibrosis (PF) remains unclear. This study observed NLRP3 inflammasome activation and pyroptosis in the peritoneum of long-term peritoneal dialysis (PD) patients with PF. Moreover, it is found that high glucose (HG) can induce the activation of NLRP3 inflammasome by regulating TLR4/NF-κB and JNK/p38 MAPK signaling in human peritoneal mesothelial cells (HPMCs), leading to subsequent Caspase-1 activation. The cleaved Caspase-1 promoted pyroptosis-related transmembrane pore formation through activating GSDMD-N, and stimulated the HPMCs to secrete inflammatory factors including IL-1β and IL-18. GSDMD global deletion or pharmacologic pretreatment with Caspase-1 specific inhibitor VX-765 effectively inhibited the pyroptosis and inflammation, thereby ameliorating PF. Additionally, treatment with VX-765 and transfected with Caspase-1 siRNA or GSDMD siRNA also inhibited the transmembrane pore formation and inflammatory factors secretion in HG-induced HPMCs. Consistent with these results, delayed treatment with VX-765 also alleviated PF, indicating the therapeutic effect of VX-765. Taken together, the results demonstrate that pyroptosis may be a novel therapeutic target for peritoneal fibrosis.
Collapse
Affiliation(s)
- Yingfeng Shi
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yan Hu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Jinqing Li
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Hui Chen
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qin Zhong
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Xiaoyan Ma
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Xialin Li
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Shasha Zhang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, 02902, USA
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| |
Collapse
|
4
|
Horikoshi K, Sakai N, Oshima M, Yamauchi H, Ikeda M, Hayashi K, Yanagisawa H, Yamamori F, Kajikawa S, Hayashi D, Koshino A, Sako K, Yuasa T, Tamai A, Minami T, Nakagawa S, Kitajima S, Toyama T, Hara A, Shimizu M, Oota S, Ishida Y, Wada T, Iwata Y. Autotaxin concentrations in peritoneal dialysis effluent reflect peritoneal function. Ther Apher Dial 2025; 29:276-284. [PMID: 39326924 DOI: 10.1111/1744-9987.14211] [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: 03/28/2024] [Revised: 06/13/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024]
Abstract
INTRODUCTION Peritoneal equilibration test (PET) has been used to monitor peritoneal function. A more convenient marker would be useful in clinical situations including home medical care. Autotaxin is known to leak into the interstitium as vascular permeability increases during the progression of tissue fibrosis. Therefore, we hypothesized that autotaxin concentrations in peritoneal dialysis (PD) effluent might reflect peritoneal function. METHODS This study enrolled 45 patients undergoing PD from 2016 to 2021. Autotaxin concentrations measured in PD effluent were evaluated for their associations with markers obtained from PET. RESULTS Mean age was 69 years, and 33 patients were men. Univariate and multivariate analyses revealed that autotaxin concentrations are associated with dialysate/plasma creatinine ratio, end/start dialysate glucose ratio, and the dip in the dialysate sodium concentration, a marker of ultrafiltration capacity, at baseline (all p < 0.05). CONCLUSIONS Autotaxin concentrations in PD effluent might be an adjunct marker that reflects peritoneal function.
Collapse
Affiliation(s)
- Keisuke Horikoshi
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Norihiko Sakai
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
- Division of Blood Purification, Kanazawa University Hospital, Kanazawa, Japan
| | - Megumi Oshima
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroyuki Yamauchi
- Department of Nephrology, Seika Town National Health Insurance Hospital, Kyoto, Japan
| | - Megumi Ikeda
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kaho Hayashi
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroyoshi Yanagisawa
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Fumitaka Yamamori
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Sho Kajikawa
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Daiki Hayashi
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Akihiko Koshino
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Keisuke Sako
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Takahiro Yuasa
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Akira Tamai
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Taichiro Minami
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Shiori Nakagawa
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Shinji Kitajima
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
- Division of Blood Purification, Kanazawa University Hospital, Kanazawa, Japan
| | - Tadashi Toyama
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Akinori Hara
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Miho Shimizu
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Satoshi Oota
- Department of Internal medicine, Toyama City Hospital, Toyama, Japan
| | - Yoichi Ishida
- Department of Internal medicine, Toyama City Hospital, Toyama, Japan
| | - Takashi Wada
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yasunori Iwata
- Department of Nephrology and Rheumatology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
5
|
Gouveia PQ, Fanelli C, Ornellas FM, Garnica MR, Francini ALR, Murata GM, Matheus LHG, Morales MM, Noronha IL. Adipose Tissue Stem Cells (ASCs) and ASC-Derived Extracellular Vesicles Prevent the Development of Experimental Peritoneal Fibrosis. Cells 2025; 14:436. [PMID: 40136685 PMCID: PMC11941392 DOI: 10.3390/cells14060436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 03/27/2025] Open
Abstract
Cell therapy utilizing mesenchymal stromal cells (MSCs) through paracrine mechanisms holds promise for regenerative purposes. Peritoneal fibrosis (PF) is a significant complication of peritoneal dialysis. Various strategies have been proposed to protect the peritoneal membrane (PM). This study explores the effectiveness of adipose-tissue-derived stem cells (ASCs) and extracellular vesicles (EVs) at mitigating PF using a rat model of PF induced by chlorhexidine gluconate. ASC and EV treatments effectively prevented an increase in the thickness of the PM and diminished the number of myofibroblasts, fibronectin expression, collagen III expression, and PF-related factors such as TGF-β and FSP-1. Smad3 gene expression decreased in the treatment groups, whereas Smad7 gene expression increased in treated animals. In addition, ASC and EV injections showed potent anti-inflammatory effects. Glucose transport through the PM remained unaffected in relation to the PF group; both treatments promoted an increase in ultrafiltration (UF) capacity. The PF+EVs treated group showed the highest increase in UF capacity. Another critical aspect of ASC and EV treatments was their impact on neoangiogenesis in the PM which is vital for UF capacity. Although the treated groups displayed a significant decrease in VEGF expression in the PM, peritoneal function remained effective. In conclusion, within the experimental PF model, both ASC and EV treatments demonstrated anti-inflammatory effects and comparably hindered the progression of PF. The EV treatment exhibited superior preservation of peritoneal function, along with enhanced UF capacity. These findings suggest the potential of ASCs and EVs as novel therapeutic approaches to prevent the development of PF associated with peritoneal dialysis.
Collapse
Affiliation(s)
- Priscila Q. Gouveia
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| | - Camilla Fanelli
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| | - Felipe M. Ornellas
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| | - Margoth R. Garnica
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| | - Ana L. R. Francini
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| | - Gilson M. Murata
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| | - Luiz H. G. Matheus
- Laboratory of Carbohydrate and Radioimmunoassay, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil;
| | - Marcelo M. Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-853, Brazil;
| | - Irene L. Noronha
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| |
Collapse
|
6
|
Chen J, Wan Z, Cao M, Huang Y, Li Y, Wu W, Guo C, Huang Z, Ou S. 68Ga-FAPI Small Animal PET/CT in Rats with Peritoneal Fibrosis and the Therapeutic Effect of Sodium Butyrate. Mol Pharm 2025; 22:1329-1338. [PMID: 39935001 DOI: 10.1021/acs.molpharmaceut.4c00998] [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: 02/13/2025]
Abstract
Peritoneal fibrosis (PF) is a common complication in peritoneal dialysis patients with end-stage renal disease. This study established a rat model of PF, used 68Ga-FAPI PET/CT imaging to visualize PF, and evaluated the therapeutic effects and mechanism of action of sodium butyrate. The rat model of PF (n = 20) was induced by hyperglycemic peritoneal dialysate combined with lipopolysaccharide, the control group (n = 20) was given the same amount of normal saline, and the intervention group (n = 20) was given sodium butyrate by intraperitoneal injection. At 2, 4, 6, and 8 weeks, a peritoneal equilibration test was performed, and peritoneal tissues were collected for histological staining. Three rats from each group were randomly selected for 68Ga-FAPI small animal PET/CT imaging. Compared with control rats, model group rats presented a decreased ultrafiltration volume, increased maximum glucose transport (P < 0.05), increased peritoneal thickness and fibrosis area, and upregulated α-SMA, COL I, TGF-β1, Smad3, and p-Smad3 expression in peritoneal tissues (P < 0.05) in a time-dependent manner. The sodium butyrate group improved peritoneal transport function (P < 0.05), alleviated collagen deposition, and downregulated α-SMA, COL I, TGF-β1, Smad3, and p-Smad3 while increasing Smad7 expression in peritoneal tissues (P < 0.05). 68Ga uptake was markedly increased in the model group (P < 0.05) but was reduced after sodium butyrate treatment (P < 0.05). The SUVmax was positively correlated with peritoneal thickness; maximum glucose transport; and α-SMA, COL I, and FAP-α expression (r = 0.871, 0.845, 0.843, 0.659, 0.926) but negatively correlated with ultrafiltration volume (r= -0.894). In summary, 68Ga-FAPI PET/CT could be a promising noninvasive approach for assessing peritoneal fibrosis that is superior to and safer than peritoneal biopsy. Sodium butyrate may attenuate peritoneal fibrosis by regulating the TGF-β1/Smad3 signaling pathway.
Collapse
Affiliation(s)
- Jingyi Chen
- Department of Nephrology, The First People's Hospital Of Guangyuan Guangyuan, Sichuan 628000, China
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zibei Wan
- Department of Nuclear Medicine and Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Mengxia Cao
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yuexi Huang
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Li
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Weihua Wu
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chunmei Guo
- Department of Nuclear Medicine and Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhanwen Huang
- Department of Nuclear Medicine and Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Santao Ou
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
7
|
Sun CY, Hsieh YT, Lu SC, Huang CYF. Prevention and Treatment of Peritoneal Dialysis-Associated Fibrosis with Intraperitoneal Anti-Fibrotic Therapy in Experimental Peritoneal Fibrosis. Pharmaceuticals (Basel) 2025; 18:188. [PMID: 40006006 PMCID: PMC11859390 DOI: 10.3390/ph18020188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Long-term peritoneal dialysis (PD) often results in peritoneal damage and fibrosis, impairing peritoneal membrane function and leading to ultrafiltration failure. This study aimed to explore the therapeutic potential of nintedanib and pirfenidone in preventing and treating PD-associated peritoneal fibrosis using experimental models. METHODS An animal model of peritoneal fibrosis and cultured mesothelial cells were utilized to evaluate the effects of nintedanib and pirfenidone. Histological analysis, molecular techniques, and RNA sequencing were employed to assess the fibrosis, inflammation, and gene expression. The key outcomes included changes in the peritoneal structure, inflammatory markers, and transcriptional regulation. RESULTS Induced peritoneal fibrosis resulted in significant structural and histological changes. Treatment with nintedanib and pirfenidone effectively prevented peritoneal thickening and reduced excessive fibrosis deposition. Both agents ameliorated the inflammatory responses by lowering inflammatory marker expression, inhibiting cytokine activity, and decreasing macrophage infiltration. Molecular analyses revealed the suppression of inflammation-related transcription regulators and cytokine receptors. RNA sequencing identified glucose-induced gene expression changes and demonstrated significant modulation by the treatments. In animal studies with established fibrosis, these agents reduced peritoneal inflammation and slowed fibrosis progression. CONCLUSIONS This study demonstrates that intraperitoneal administration of nintedanib and pirfenidone shows promise as an anti-fibrosis therapy for preventing and treating peritoneal fibrosis associated with PD. These findings highlight the potential of targeted interventions to improve the long-term outcomes for PD patients.
Collapse
Affiliation(s)
- Chiao-Yin Sun
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 61363, Taiwan
- Kidney Research Center and Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
| | - Yu-Ting Hsieh
- Department of Medical Research, Chang Gung Memorial Hospital, Keelung 83301, Taiwan; (Y.-T.H.); (S.-C.L.)
| | - Shang-Chieh Lu
- Department of Medical Research, Chang Gung Memorial Hospital, Keelung 83301, Taiwan; (Y.-T.H.); (S.-C.L.)
| | - Chi-Ying F. Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang-Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
8
|
Sun Y, Huang Q, Sun J, Zhou H, Guo D, Peng L, Lin H, Li C, Shang H, Wang T, Chen Y, Huang Y, Hu C, Hu Z, Lu Y, Peng H. Mucosal-Associated Invariant T (MAIT) Cell-Mediated Immune Mechanisms of Peritoneal Dialysis-Induced Peritoneal Fibrosis and Therapeutic Targeting. J Am Soc Nephrol 2025:00001751-990000000-00539. [PMID: 39874111 DOI: 10.1681/asn.0000000627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
Key Points
Peritoneal mucosal-associated invariant T (MAIT) cells were characterized by single-cell RNA sequencing, histological imaging, and flow cytometry.Activation of MAIT cells modulated glucose metabolism in mesothelial cells by TCRVα7.2-MHC class 1–related protein 1 signaling and triggered peritoneal fibrogenesis.Pharmacological inhibition of MAIT cell function by acetyl-6-formylpterin mitigated peritoneal fibrosis.
Background
Peritoneal fibrosis is a serious complication of long-term peritoneal dialysis (PD) and abdominal surgeries, yet effective treatments remain elusive. Given the known roles of mucosal-associated invariant T (MAIT) cells in immune responses and fibrotic diseases, we investigated their involvement in PD-induced peritoneal fibrosis to identify potential therapeutic targets.
Methods
We used single-cell RNA sequencing and flow cytometry to characterize the activation and function of peritoneal MAIT cells in patients undergoing long-term PD. Our investigation focused on the molecular pathways activated by these cells, particularly the MHC class 1–related protein 1 (MR1)-mediated interaction with mesothelial cells and subsequent activation of the mTOR complex 1 signaling pathway. We further assessed the effect of inhibiting MAIT cells on fibrogenesis using both in vitro models and Mr1 knockout mice.
Results
Our study revealed that long-term PD significantly enhanced the activation of MAIT cells, particularly the proinflammatory MAIT17 subtype. These activated cells contributed to peritoneal fibrogenesis by binding to the MR1 receptor on mesothelial cells, which triggered hyperglycolysis through the mTOR complex 1 pathway, ultimately leading to fibrogenesis. Notably, we demonstrated that blocking the MR1–MAIT interaction, either through genetic knockout or pharmacological inhibition with acetyl-6-formylpterin, effectively mitigated fibrosis.
Conclusions
This study identified MAIT cells as crucial drivers of PD-induced peritoneal fibrosis.
Collapse
Affiliation(s)
- Yuxiang Sun
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Qiang Huang
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Juan Sun
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hu Zhou
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dandan Guo
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Long Peng
- Division of Cardiovascular Medicine, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongchun Lin
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Canming Li
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongli Shang
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tongtong Wang
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanxu Chen
- Organ Transplant Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yong Huang
- Division of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Cheng Hu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhaoyong Hu
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Yan Lu
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hui Peng
- Nephrology Division, Department of Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| |
Collapse
|
9
|
Wei YS, Tsai SY, Lin SL, Chen YT, Tsai PS. Methylglyoxal-Stimulated Mesothelial Cells Prompted Fibroblast-to-Proto-Myofibroblast Transition. Int J Mol Sci 2025; 26:813. [PMID: 39859527 PMCID: PMC11766140 DOI: 10.3390/ijms26020813] [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: 12/25/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
During long-term peritoneal dialysis, peritoneal fibrosis (PF) often happens and results in ultrafiltration failure, which directly leads to the termination of dialysis. The accumulation of extracellular matrix produced from an increasing number of myofibroblasts was a hallmark characteristic of PF. To date, glucose degradation products (GDPs, i.e., methylglyoxal (MGO)) that appeared during the heating and storage of the dialysate are considered to be key components to initiating PF, but how GDPs lead to the activation of myofibroblast in fibrotic peritoneum has not yet been fully elucidated. In this study, mesothelial cell line (MeT-5A) and fibroblast cell line (MRC-5) were used to investigate the transcriptomic and proteomic changes to unveil the underlying mechanism of MGO-induced PF. Our transcriptomic data from the MGO-stimulated mesothelial cells showed upregulation of genes involved in pro-inflammatory, apoptotic, and fibrotic pathways. While no phenotypic changes were noted on fibroblasts after direct MGO, supernatant from MGO-stimulated mesothelial cells promoted fibroblasts to change into proto-myofibroblasts, activated fibroblasts in the first stage toward myofibroblasts. In conclusion, this study showed that MGO-stimulated mesothelial cells promoted fibroblast-to-proto-myofibroblast transition; however, additional involvement of other factors or cells (e.g., macrophages) may be needed to complete the transformation into myofibroblasts.
Collapse
Affiliation(s)
- Yu-Syuan Wei
- Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan;
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Su-Yi Tsai
- Department of Life Science, College of Life Science, National Taiwan University, Taipei 10617, Taiwan;
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei 10617, Taiwan;
| | - Shuei-Liong Lin
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei 10617, Taiwan;
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan;
| | - Yi-Ting Chen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan;
- Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Pei-Shiue Tsai
- Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan;
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei 10617, Taiwan;
| |
Collapse
|
10
|
Diao X, Zhan C, Ye H, Wu H, Yi C, Lin J, Mao H, Chen W, Yang X. Single-cell transcriptomic reveals the peritoneal microenvironmental change in long-term peritoneal dialysis patients with ultrafiltration failure. iScience 2024; 27:111383. [PMID: 39687014 PMCID: PMC11647153 DOI: 10.1016/j.isci.2024.111383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 07/02/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
The microenvironmental changes in peritoneal dialysis effluent (PDE) after long-term vintage (LV) of PD in patients with ultrafiltration failure (LV_UF) are unclear. Single-cell sequencing revealed that peritoneal neutrophils were elevated in LV_UF patients, while MRC1-macrophage subcluster decreased compared with PD patients with short vintage (SV) and LV without ultrafiltration failure (LV_NOT_UF). Compared with the LV_NOT_UF group, the upregulated differentially expressed genes (DEGs) of monocytes/macrophages in the LV_UF group were involved in inflammatory response and EMT progress. LV_UF patients had a higher proportion of epithelial-like mesothelial cells (E-MCs), which were characterized by autophagy activation, inflammation, and upregulation of neutrophil- and autophagy-related DEGs compared to the LV_NOT_UF group. Additionally, mesenchymal-like MCs and AQP1 expression were reduced in the LV_UF group compared with the other groups. Both neutrophils and monocytes/macrophages interacted with MCs. Our study provides insights into the roles of peritoneal mesothelial cells and inflammatory cells in PD patients with UF.
Collapse
Affiliation(s)
- Xiangwen Diao
- Department of Emergency, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Cuixia Zhan
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Hongjian Ye
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Haishan Wu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Chunyan Yi
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Jianxiong Lin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| | - Xiao Yang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, Guangdong, China
| |
Collapse
|
11
|
Lu Y, Gao L, Zhang W, Zeng Y, Hu J, Song K. Caffeic acid phenethyl ester restores mitochondrial homeostasis against peritoneal fibrosis induced by peritoneal dialysis through the AMPK/SIRT1 pathway. Ren Fail 2024; 46:2350235. [PMID: 38721924 PMCID: PMC11086008 DOI: 10.1080/0886022x.2024.2350235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 04/27/2024] [Indexed: 05/12/2024] Open
Abstract
Increasing evidence suggests that peritoneal fibrosis induced by peritoneal dialysis (PD) is linked to oxidative stress. However, there are currently no effective interventions for peritoneal fibrosis. In the present study, we explored whether adding caffeic acid phenethyl ester (CAPE) to peritoneal dialysis fluid (PDF) improved peritoneal fibrosis caused by PD and explored the molecular mechanism. We established a peritoneal fibrosis model in Sprague-Dawley rats through intraperitoneal injection of PDF and lipopolysaccharide (LPS). Rats in the PD group showed increased peritoneal thickness, submesothelial collagen deposition, and the expression of TGFβ1 and α-SMA. Adding CAPE to PDF significantly inhibited PD-induced submesothelial thickening, reduced TGFβ1 and α-SMA expression, alleviated peritoneal fibrosis, and improved the peritoneal ultrafiltration function. In vitro, peritoneal mesothelial cells (PMCs) treated with PDF showed inhibition of the AMPK/SIRT1 pathway, mitochondrial membrane potential depolarization, overproduction of mitochondrial reactive oxygen species (ROS), decreased ATP synthesis, and induction of mesothelial-mesenchymal transition (MMT). CAPE activated the AMPK/SIRT1 pathway, thereby inhibiting mitochondrial membrane potential depolarization, reducing mitochondrial ROS generation, and maintaining ATP synthesis. However, the beneficial effects of CAPE were counteracted by an AMPK inhibitor and siSIRT1. Our results suggest that CAPE maintains mitochondrial homeostasis by upregulating the AMPK/SIRT1 pathway, which alleviates oxidative stress and MMT, thereby mitigating the damage to the peritoneal structure and function caused by PD. These findings suggest that adding CAPE to PDF may prevent and treat peritoneal fibrosis.
Collapse
Affiliation(s)
- Ying Lu
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Luyan Gao
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenwen Zhang
- Department of Nephrology, Zibo City Hospital Combined of Traditional Chinese and Western Medicine, Zibo, China
| | - Ying Zeng
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Kai Song
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
12
|
Zheng L, Chen W, Yao K, Xie Y, Liao C, Lin Y, Zhou T. Efficacy of mesenchymal stem cells in the treatment of peritoneal fibrosis in animal models: a systematic review and meta-analysis. Ren Fail 2024; 46:2438863. [PMID: 39681477 PMCID: PMC11650619 DOI: 10.1080/0886022x.2024.2438863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Peritoneal fibrosis is a serious complication of long-term peritoneal dialysis, often resulting in functional deterioration and withdrawal from therapy. Mesenchymal stem cells (MSCs) have demonstrated immunomodulatory and antifibrotic effects in various models. This meta-analysis evaluated the efficacy of MSCs therapy in animal models of peritoneal fibrosis. METHODS A comprehensive search of PubMed, the Cochrane Library, Web of Science, and EMBASE was conducted for studies published up to April 27, 2024. Two independent reviewers (LQZ and WMC) screened studies for inclusion, extracted data, and analyzed outcomes using RevMan 5.3 and STATA 17.0. RESULT Fifteen studies met the inclusion criteria. MSC therapy significantly reduced inflammatory cytokines, including IL-6, TGF-β (SMD = -1.79, 95% CI: -2.32, -1.25, p < 0.00001), and TNF-α (SMD = -1.57, 95% CI: -2.71, -0.44, p = 0.006) levels. Additionally, MSCs reduced submesothelial thickness (MD = -63.14, 95% CI: -78.52, -47.76, p < 0.00001), Collagen I and Collagen III levels. MSCs treatment also improved ultrafiltration capacity (MD = 1.21, 95% CI: 0.64, 1.77, p < 0.0001), D/D0 of glucose and E-cadherin levels. However, no significant differences were observed in VEGF, D/P of Na, D/P of BUN, D/P of protein, or glucose mass transfer between the MSCs treatment group and the control group. CONCLUSION MSC therapy significantly improves peritoneal function and attenuates fibrotic and inflammatory responses in animal models. These findings highlight the potential of MSCs as a promising therapeutic strategy for managing peritoneal fibrosis in clinical settings.
Collapse
Affiliation(s)
- Lingqian Zheng
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Wenmin Chen
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Kaijin Yao
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yina Xie
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Chunling Liao
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yongda Lin
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Tianbiao Zhou
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
13
|
Hu Y, Zhou C, Zhong Q, Li X, Li J, Shi Y, Ma X, Jiang D, Wang Y, Zhuang S, Liu N. LCZ696, an angiotensin receptor-neprilysin inhibitor, ameliorates epithelial-mesenchymal transition of peritoneal mesothelial cells and M2 macrophage polarization. Ren Fail 2024; 46:2392849. [PMID: 39165231 PMCID: PMC11340223 DOI: 10.1080/0886022x.2024.2392849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/11/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024] Open
Abstract
AIMS To investigate the effects and mechanisms of LCZ696, an angiotensin receptor-neprilysin inhibitor (ARNI), on epithelial-mesenchymal transition (EMT) of peritoneal mesothelial cells and on macrophage M2 polarization. METHODS We examined the effects of LCZ696 in a 4.25% high glucose peritoneal dialysis fluid (PDF)-induced peritoneal fibrosis (PF) mouse model, and explored the mechanisms of LCZ696 on human peritoneal mesothelial cells (HPMCs) stimulated by TGF-β1 (5 ng/mL) and on Raw264.7 cells stimulated by IL-4 (10 ng/mL). To further elucidate the mechanism, we treated HPMCs with the conditioned medium of Raw264.7 cells. RESULTS LCZ696 effectively improved PF and inhibited the process of EMT in PDF mice. In vitro, LCZ696 also significantly alleviated the EMT of TGF-β1 induced HPMCs, although there was no statistically significant difference when compared to the Valsartan treatment group. Moreover, LCZ696 ameliorates the increased expression of Snail and Slug, two nuclear transcription factors that drive the EMT. Mechanistically, TGF-β1 increased the expression of TGFβRI, p-Smad3, p-PDGFRβ and p-EGFR, while treatment with LCZ696 abrogated the activation of TGF-β/Smad3, PDGFRβ and EGFR signaling pathways. Additionally, exposure of Raw264.7 to IL-4 results in increasing expression of Arginase-1, CD163 and p-STAT6. Treatment with LCZ696 inhibited IL-4-elicited M2 macrophage polarization by inactivating the STAT6 signaling pathway. Furthermore, we observed that LCZ696 inhibits EMT by blocking TGF-β1 secretion from M2 macrophages. CONCLUSION Our study demonstrated that LCZ696 improves PF and ameliorates TGF-β1-induced EMT of HPMCs by blocking TGF-β/Smad3, PDGFRβ and EGFR pathways. Meanwhile, LCZ696 also inhibits M2 macrophage polarization by regulating STAT6 pathway.
Collapse
Affiliation(s)
- Yan Hu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Canxin Zhou
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qin Zhong
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xialin Li
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinqing Li
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yingfeng Shi
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyan Ma
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Daofang Jiang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, USA
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Chen YW, Liao CT, Wu MY, Huang NJ, Cherng YG, Wu MS, Hsu YH, Chen CH. Pressure induces peritoneal fibrosis and inflammation through CD44 signaling. Ren Fail 2024; 46:2384586. [PMID: 39082695 PMCID: PMC11293264 DOI: 10.1080/0886022x.2024.2384586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 08/03/2024] Open
Abstract
Peritoneal dialysis (PD) is a widely used sustainable kidney replacement therapy. Prolonged use of PD fluids is associated with mesothelial-mesenchymal transition, peritoneal fibrosis, and eventual ultrafiltration (UF) failure. However, the impact of pressure on the peritoneum remains unclear. In the present study, we hypothesized increased pressure is a potential contributing factor to peritoneal fibrosis and investigated the possible mechanisms. In vitro experiments found that pressurization led to a mesenchymal phenotype, the expression of fibrotic markers and inflammatory factors in human mesothelial MeT-5A cells. Pressure also increased cell proliferation and augmented cell migration potential in MeT-5A cells. The mouse PD model and human peritoneum equilibrium test (PET) data both showed a positive association between higher pressure and increased small solute transport, along with decreased net UF. Mechanistically, we found that significant upregulation of CD44 in mesothelial cells upon pressurization. Notably, the treatment of CD44 neutralizing antibodies prevented pressure-induced phenotypic changes in mesothelial cells, while a CD44 inhibitor oligo-fucoidan ameliorated pressure-induced peritoneal thickening, fibrosis, and inflammation in PD mice. To conclude, intraperitoneal pressure results in peritoneal fibrosis in PD via CD44-mediated mesothelial changes and inflammation. CD44 blockage can be utilized as a novel preventive approach for PD-related peritoneal fibrosis and UF failure.
Collapse
Affiliation(s)
- Yu-Wei Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Chia-Te Liao
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Mei-Yi Wu
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Nai-Jen Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yih-Giun Cherng
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mai-Szu Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Yung-Ho Hsu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Hsin Kuo Min Hospital, Taipei Medical University, Taoyuan City, Taiwan
| | - Cheng-Hsien Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
- Department of Internal Medicine, Division of Nephrology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
15
|
Xu M, Huan J, Zhu L, Xu J, Song K. The neutrophil percentage-to-albumin ratio is an independent risk factor for poor prognosis in peritoneal dialysis patients. Ren Fail 2024; 46:2294149. [PMID: 38178381 PMCID: PMC10773631 DOI: 10.1080/0886022x.2023.2294149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
AIM This study aimed to investigate the predictive ability of the neutrophil percentage-to-albumin Ratio (NPAR) concerning all-cause mortality and cardio-cerebrovascular mortality in patients undergoing peritoneal dialysis (PD). METHODS We included a total of 807 PD patients from the Peritoneal Dialysis Center of the Second Affiliated Hospital of Soochow University between January 2009 and December 2019 in this study. Patients were categorized into three groups based on their baseline NPAR. The Kaplan-Meier method, multivariate Cox proportional hazard model, and Fine-Gray competing risk model were employed to examine the relationship between NPAR level and all-cause mortality and cardio-cerebrovascular mortality among PD patients. Furthermore, the ROC curve and calibration plots were utilized to compare the performance between NPAR and other conventional indicators. RESULTS The mean follow-up period was 38.2 months. A total of 243 (30.1%) patients passed away, with 128 (52.7%) succumbing to cardio-cerebrovascular diseases. The mortality rates of the Middle and High NPAR groups were significantly greater than that of the Low NPAR group (p < 0.001), and NPAR was independently associated with all-cause mortality and cardio-cerebrovascular mortality. Receiver Operating Characteristic (ROC) analysis indicated that the Area Under the Curve (AUC) of NPAR (0.714) was significantly superior to those of C-reactive protein (CRP) (0.597), neutrophil to lymphocyte ratio (NLR) (0.589), C-reactive protein to albumin ratio (CAR) (0.698) and platelet to lymphocyte ratio (PLR) (0.533). CONCLUSION NPAR served as an independent predictive marker for all-cause mortality and cardio-cerebrovascular mortality in PD patients. Moreover, NPAR demonstrated superior predictive potential compared to CRP, CAR, NLR, and PLR.
Collapse
Affiliation(s)
- Mingfan Xu
- Department of Nephrology, The Second Affiliated Hospital of Soochow UniversityChina, China
| | - Jingjia Huan
- Department of Nephrology, The Second Affiliated Hospital of Soochow UniversityChina, China
| | - Lujie Zhu
- Department of Nephrology, The Second Affiliated Hospital of Soochow UniversityChina, China
| | - Jiachun Xu
- Department of Nephrology, The Second Affiliated Hospital of Soochow UniversityChina, China
| | - Kai Song
- Department of Nephrology, The Second Affiliated Hospital of Soochow UniversityChina, China
| |
Collapse
|
16
|
Wu T, Yu Z, Dai J, Li J, Ning F, Liu X, Zhu N, Zhang X. JPH203 alleviates peritoneal fibrosis via inhibition of amino acid-mediated mTORC1 signaling. Biochem Biophys Res Commun 2024; 734:150656. [PMID: 39362029 DOI: 10.1016/j.bbrc.2024.150656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND AND AIMS The mesothelial-mesenchymal transition (MMT) of mesothelial cells has been recognized as a critical process during progression of peritoneal fibrosis (PF). Despite its crucial role in amino acid transport and metabolism, the involvement of L-type amino acid transporter 1 (LAT1) and the potential therapeutic role of its inhibitor, JPH203, in fibrotic diseases remain unexplored. Considering the paucity of research on amino acid-mediated mTORC1 activation in PF, our study endeavors to elucidate the protective effects of JPH203 against PF and explore the involvement of amino acid-mediated mTORC1 signaling in this context. METHODS We established the transforming growth factor beta 1 (TGF-β1) induced MMT model in primary human mesothelial cells and the peritoneal dialysis fluid (PDF) induced PF model in mice. The therapeutic effects of JPH203 on PF were then examined on these two models by real-time quantitative polymerase chain reaction, western blotting, immunofluorescence staining, Masson's trichrome staining, H&E staining, picro-sirius red staining, and immunohistochemistry. The involvement of amino acid-mediated mTORC1 signaling was screened by RNA sequencing and further verified by western blotting in vitro. RESULTS LAT1 was significantly upregulated and JPH203 markedly attenuated fibrotic phenotype both in vitro and in vivo. RNA-seq unveiled a significant enrichment of mTOR signaling pathway in response to JPH203 treatment. Western blotting results indicated that JPH203 alleviates PF by inhibiting amino acid-mediated mTORC1 signaling, which differs from the direct inhibition observed with rapamycin. CONCLUSION JPH203 alleviates PF by inhibiting amino acid-mediated mTORC1 signaling.
Collapse
Affiliation(s)
- Tiangang Wu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Zanzhe Yu
- Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Junhao Dai
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiayang Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Fengling Ning
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xin Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Nan Zhu
- Department of Nephrology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China; School of Pharmacy, East China Normal University, Shanghai, China.
| |
Collapse
|
17
|
Shinkai Y, Sasaki K, Tamura R, Ike T, Takahashi A, Osaki Y, Ishiuchi N, Maeoka Y, Nakashima A, Masaki T. Selective activation of PPARα by pemafibrate mitigates peritoneal inflammation and fibrosis through suppression of NLRP3 inflammasome and modulation of inflammation. Sci Rep 2024; 14:23816. [PMID: 39394435 PMCID: PMC11470028 DOI: 10.1038/s41598-024-74340-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/25/2024] [Indexed: 10/13/2024] Open
Abstract
Peritoneal inflammation and fibrosis remain major challenges to the long-term maintenance of peritoneal dialysis. Pemafibrate, a selective peroxisome proliferator-activated receptor α (PPARα) modulator, has been implicated in the management of fibrosis-related disorders. We investigated whether pemafibrate ameliorates peritoneal inflammation and fibrosis and explored the underlying mechanisms in mice with methylglyoxal (MGO)-induced peritoneal fibrosis (MGO mice). MGO mice exhibited peritoneal fibrosis with increased expression of mesenchymal markers, transforming growth factor-β1 (TGF-β1), and substantial deposition of extracellular matrix (ECM) proteins. Additionally, MGO mice exhibited peritoneal inflammation as indicated by elevated tumor necrosis factor-α expression and macrophage infiltration in peritoneal tissue. These effects were mitigated by pemafibrate treatment, which also restored peritoneal membrane function. Furthermore, pemafibrate promoted anti-inflammatory macrophage polarization in both mice and THP-1 cells. In human peritoneal mesothelial cells (HPMCs), pemafibrate effectively inhibited interferon-γ-induced production of TGF-β1 and ECM while suppressing the proinflammatory cytokines nuclear factor-κB (NF-κB) and activator protein 1. The NF-κB inhibitory effect of pemafibrate involved stabilization of the NF-κB inhibitory protein IkBα. Notably, pemafibrate hindered activation of the NLR family pyrin domain containing 3/caspase-1 axis in interferon-γ-stimulated THP-1 cells. These findings suggest that pemafibrate ameliorates peritoneal inflammation and fibrosis, making it a promising candidate for peritoneal fibrosis therapy.
Collapse
Affiliation(s)
- Yutaka Shinkai
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kensuke Sasaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Ryo Tamura
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Takeshi Ike
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Akira Takahashi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yosuke Osaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Naoki Ishiuchi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yujiro Maeoka
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| |
Collapse
|
18
|
Kang Y, Liu Y, Fu P, Ma L. Peritoneal fibrosis: from pathophysiological mechanism to medicine. Front Physiol 2024; 15:1438952. [PMID: 39301425 PMCID: PMC11411570 DOI: 10.3389/fphys.2024.1438952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
Peritoneal dialysis (PD) is currently one of the effective methods for treating end-stage renal disease (ESRD). However, long-term exposure to high concentration glucose in peritoneal dialysis environment could lead to peritoneal fibrosis (PF), impaired peritoneal filtration function, decreased peritoneal dialysis efficiency, and even withdrawal from peritoneal dialysis in patients. Considerable evidence suggests that peritoneal fibrosis after peritoneal dialysis is related to crucial factors such as mesothelial-to-mesenchymal transition (MMT), inflammatory response, and angiogenesis, etc. In our review, we summarize the pathophysiological mechanisms and further illustrate the future strategies against PF.
Collapse
Affiliation(s)
- Yingxi Kang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Yuan Liu
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ping Fu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Liang Ma
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Shi SS, Zhang YQ, Zhang LQ, Li YF, Zhou XS, Li RS. Expression and significance of SIRT6 in human peritoneal dialysis effluents and peritoneal mesothelial cells. Int Urol Nephrol 2024; 56:2659-2670. [PMID: 38483736 PMCID: PMC11266209 DOI: 10.1007/s11255-024-03970-5] [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: 08/30/2023] [Accepted: 02/02/2024] [Indexed: 07/25/2024]
Abstract
Sirtuin 6 (SIRT6) can inhibit the fibrosis of many organs. However, the relationship between SIRT6 and peritoneal fibrosis (PF) in peritoneal dialysis (PD) remains unclear. We collected 110 PD patients with a duration of PD for more than 3 months and studied the influence of PD duration and history of peritonitis on SIRT6 levels in PD effluents (PDEs). We also analyzed the relationship between SIRT6 levels in PDEs and transforming growth factor beta 1 (TGF-β1), IL-6, PD duration, peritoneal function, PD ultrafiltration (UF), and glucose exposure. We extracted human peritoneal mesothelial cells (HPMCs) from PDEs and measured the protein and gene expression levels of SIRT6, E-cadherin, vimentin, and TGF-β1 in these cells. Based on the clinical results, we used human peritoneal mesothelial cells lines (HMrSV5) to observe the changes in SIRT6 levels and mesothelial-to-mesenchymal transition (MMT) after intervention with PD fluid. By overexpressing and knocking down SIRT6 expression, we investigated the effect of SIRT6 expression on E-cadherin, vimentin, and TGF-β1 expression to elucidate the role of SIRT6 in mesothelial-to-epithelial transition in PMCs. Results: (1) With the extension of PD duration, the influence of infection on SIRT6 levels in PDEs increased. Patients with the PD duration of more than 5 years and a history of peritonitis had the lowest SIRT6 levels. (2) SIRT6 levels in PDEs were negatively correlated with PD duration, total glucose exposure, TGF-β1, IL-6 levels, and the dialysate-to-plasma ratio of creatinine (Cr4hD/P), but positively correlated with UF. This indicates that SIRT6 has a protective effect on the peritoneum. (3) The short-term group (PD ≤ 1 year) had higher SIRT6 and E-cadherin gene and protein levels than the mid-term group (1 year < PD ≤ 5 years) and long-term group (PD > 5 years) in PMCs, while vimentin and TGF-β1 levels were lower in the mid-term group and long-term group. Patients with a history of peritonitis had lower SIRT6 and E-cadherin levels than those without such a history. (4) After 4.25% PD fluid intervention for HPMCs, longer intervention time resulted in lower SIRT6 levels. (5) Overexpressing SIRT6 can lead to increased E-cadherin expression and decreased vimentin and TGF-β1 expression in HPMCs. Knocking down SIRT6 expression resulted in decreased E-cadherin expression and increased vimentin and TGF-β1 expression in HPMCs. This indicates that SIRT6 expression can inhibit MMT in HPMCs, alleviate PF associated with PD, and have a protective effect on the peritoneum.
Collapse
Affiliation(s)
- Shuai-Shuai Shi
- Department of Nephrology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, Shanxi, China
- Department of Nephrology, Heji Hospital of Changzhi Medical College, Changzhi, 046011, Shanxi, China
| | - Yi-Qiang Zhang
- Department of Biochemistry, Changzhi Medical College, Changzhi, 046000, Shanxi, China
| | - Lu-Qi Zhang
- First Clinical Department of Changzhi Medical College, Changzhi, 046000, Shanxi, China
| | - Yun-Feng Li
- First Clinical Department of Changzhi Medical College, Changzhi, 046000, Shanxi, China
| | - Xiao-Shuang Zhou
- Department of Nephrology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, Shanxi, China
| | - Rong-Shan Li
- Department of Nephrology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, Shanxi, China.
| |
Collapse
|
20
|
Kim JE, Han D, Kim KH, Seo A, Moon JJ, Jeong JS, Kim JH, Kang E, Bae E, Kim YC, Lee JW, Cha RH, Kim DK, Oh KH, Kim YS, Jung HY, Yang SH. Protective effect of Cyclo(His-Pro) on peritoneal fibrosis through regulation of HDAC3 expression. FASEB J 2024; 38:e23819. [PMID: 38984942 DOI: 10.1096/fj.202400854r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024]
Abstract
Peritoneal dialysis is a common treatment for end-stage renal disease, but complications often force its discontinuation. Preventive treatments for peritoneal inflammation and fibrosis are currently lacking. Cyclo(His-Pro) (CHP), a naturally occurring cyclic dipeptide, has demonstrated protective effects in various fibrotic diseases, yet its potential role in peritoneal fibrosis (PF) remains uncertain. In a mouse model of induced PF, CHP was administered, and quantitative proteomic analysis using liquid chromatography-tandem mass spectrometry was employed to identify PF-related protein signaling pathways. The results were further validated using human primary cultured mesothelial cells. This analysis revealed the involvement of histone deacetylase 3 (HDAC3) in the PF signaling pathway. CHP administration effectively mitigated PF in both peritoneal tissue and human primary cultured mesothelial cells, concurrently regulating fibrosis-related markers and HDAC3 expression. Moreover, CHP enhanced the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) while suppressing forkhead box protein M1 (FOXM1), known to inhibit Nrf2 transcription through its interaction with HDAC3. CHP also displayed an impact on spleen myeloid-derived suppressor cells, suggesting an immunomodulatory effect. Notably, CHP improved mitochondrial function in peritoneal tissue, resulting in increased mitochondrial membrane potential and adenosine triphosphate production. This study suggests that CHP can significantly prevent PF in peritoneal dialysis patients by modulating HDAC3 expression and associated signaling pathways, reducing fibrosis and inflammation markers, and improving mitochondrial function.
Collapse
Affiliation(s)
- Ji Eun Kim
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Dohyun Han
- Proteomics Core Facility, Seoul National University Hospital, Seoul, Korea
| | - Kyu Hong Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Areum Seo
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Jong Joo Moon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jin Seon Jeong
- Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | - Ji Hye Kim
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Korea
| | - Eunjeong Kang
- Transplantation Center, Seoul National University Hospital, Seoul, Korea
| | - Eunjin Bae
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Changwon, Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Wook Lee
- Nephrology Clinic, National Cancer Center, Goyang, Korea
| | - Ran-Hui Cha
- Department of Internal Medicine, National Medical Center, Seoul, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hoe-Yune Jung
- R&D Center, NovMetaPharma Co., Ltd, Pohang, Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Korea
| | - Seung Hee Yang
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Cheng XBJ, Bargman J. Complications of Peritoneal Dialysis Part II: Nonmechanical Complications. Clin J Am Soc Nephrol 2024; 19:791-799. [PMID: 38190143 PMCID: PMC11168822 DOI: 10.2215/cjn.0000000000000418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 12/21/2023] [Indexed: 01/09/2024]
Abstract
Peritoneal dialysis (PD) is a form of KRT that offers flexibility and autonomy to patients with ESKD. It is associated with lower costs compared with hemodialysis in many countries. Unlike mechanical complications that typical arise early in the course of treatment, noninfectious, nonmechanical complications often present late in patients who are established on PD. In this review, we first discuss abnormal-appearing drained dialysate, including hemoperitoneum, chyloperitoneum, and noninfectious cloudy dialysate. The underlying cause is frequently unrelated to PD. We then discuss encapsulating peritoneal sclerosis, a rare complication of PD. Finally, we review metabolic changes associated with PD and methods to mitigate its effects.
Collapse
Affiliation(s)
- Xin Bo Justin Cheng
- University Health Network, Toronto, Ontario, Canada
- Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Joanne Bargman
- University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Zhong W, Fu J, Liao J, Ouyang S, Yin W, Liang Y, Liu K. A protective role of nintedanib in peritoneal fibrosis through H19-EZH2-KLF2 axis via impeding mesothelial-to-mesenchymal transition. Int Urol Nephrol 2024; 56:1987-1999. [PMID: 38097887 DOI: 10.1007/s11255-023-03892-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 11/16/2023] [Indexed: 05/14/2024]
Abstract
BACKGROUND Peritoneal fibrosis (PF), a common complication of long-term peritoneal dialysis, accounts for peritoneal ultrafiltration failure to develop into increased mortality. Nintedanib has previously been shown to protect against multi-organ fibrosis, including PF. Unfortunately, the precise molecular mechanism underlying nintedanib in the pathogenesis of PF remains elusive. METHODS The mouse model of PF was generated by chlorhexidine gluconate (CG) injection with or without nintedanib administration, either with the simulation for the cell model of PF by constructing high-glucose (HG)-treated human peritoneal mesothelial cells (HPMCs). HE and Masson staining were applied to assess the histopathological changes of peritoneum and collagen deposition. FISH, RT-qPCR, western blot and immunofluorescence were employed to examine distribution or expression of targeted genes. Cell viability was detected using CCK-8 assay. Cell morphology was observed under a microscope. RNA immunoprecipitation (RIP) and chromatin immunoprecipitation (ChIP) assays were applied to validate the H19-EZH2-KLF2 regulatory axis. RESULTS Aberrantly overexpressed H19 was observed in both the mouse and cell model of PF, of which knockdown significantly blocked HG-induced mesothelial-to-mesenchymal transition (MMT) of HPMCs. Moreover, loss of H19 further strengthened nintedanib-mediated suppressive effects against MMT process in a mouse model of PF. Mechanistically, H19 could epigenetically repressed KLF2 via recruiting EZH2. Furthermore, TGF-β/Smad pathway was inactivated by nintedanib through mediating H19/KLF2 axis. CONCLUSION In summary, nintedanib disrupts MMT process through regulating H19/EZH2/KLF2 axis and TGF-β/Smad pathway, which laid the experimental foundation for nintedanib in the treatment of PF.
Collapse
Affiliation(s)
- Wei Zhong
- Department of Nephrology and Laboratory of Kidney Disease, Changsha Clinical Research Center for Kidney Disease, Hunan Clinical Research Center for Chronic Kidney Disease, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No.61, Jiefangxi Road, Changsha, 410002, Hunan, People's Republic of China
| | - Jia Fu
- Department of Oncology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410002, Hunan, People's Republic of China
| | - Jin Liao
- Department of Nephrology and Laboratory of Kidney Disease, Changsha Clinical Research Center for Kidney Disease, Hunan Clinical Research Center for Chronic Kidney Disease, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No.61, Jiefangxi Road, Changsha, 410002, Hunan, People's Republic of China
| | - Shaxi Ouyang
- Department of Nephrology and Laboratory of Kidney Disease, Changsha Clinical Research Center for Kidney Disease, Hunan Clinical Research Center for Chronic Kidney Disease, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No.61, Jiefangxi Road, Changsha, 410002, Hunan, People's Republic of China
| | - Wei Yin
- Department of Nephrology and Laboratory of Kidney Disease, Changsha Clinical Research Center for Kidney Disease, Hunan Clinical Research Center for Chronic Kidney Disease, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No.61, Jiefangxi Road, Changsha, 410002, Hunan, People's Republic of China
| | - Yumei Liang
- Department of Nephrology and Laboratory of Kidney Disease, Changsha Clinical Research Center for Kidney Disease, Hunan Clinical Research Center for Chronic Kidney Disease, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No.61, Jiefangxi Road, Changsha, 410002, Hunan, People's Republic of China
| | - Kanghan Liu
- Department of Nephrology and Laboratory of Kidney Disease, Changsha Clinical Research Center for Kidney Disease, Hunan Clinical Research Center for Chronic Kidney Disease, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No.61, Jiefangxi Road, Changsha, 410002, Hunan, People's Republic of China.
| |
Collapse
|
23
|
Huang Y, Chu C, Mai Y, Zhao Y, Cao L, Ji S, Zhu B, Shen Q. Treatment of peritoneal fibrosis: Therapeutic prospects of bioactive Agents from Astragalus membranaceus. Front Pharmacol 2024; 15:1347234. [PMID: 38835665 PMCID: PMC11148558 DOI: 10.3389/fphar.2024.1347234] [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: 12/08/2023] [Accepted: 05/02/2024] [Indexed: 06/06/2024] Open
Abstract
Peritoneal dialysis is one of the renal replacement treatments for patients with end-stage renal disease. Peritoneal dialysis-related peritoneal fibrosis is a pathological change in peritoneal tissue of peritoneal dialysis patients with progressive, non-suppurative inflammation accompanied by fibrous tissue hyperplasia, resulting in damage to the original structure and function, leading to peritoneal function failure. Currently, there is no specific drug in the clinic. Therefore, it is necessary to find a drug with good effects and few adverse reactions. Astragalus membranaceus (AMS) is the dried root of the Astragalus membranaceus (Fisch.) Bge. AMS and its active ingredients play a significant role in anti-inflammation, anti-fibrosis, regulation of immune function and regulation of blood pressure. Studies have shown that it can alleviate peritoneal fibrosis by reducing inflammatory response, inhibiting oxidative stress, degrading extracellular matrix deposition, regulating apoptosis, and regulating Transforming Growth Factor-β. The author summarized the relationship between AMS and its active ingredients by referring to relevant literature at home and abroad, in order to provide some theoretical basis for further clinical research.
Collapse
Affiliation(s)
- Ying Huang
- School of Public Health, Hangzhou Medical College, Hangzhou, China
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Chenling Chu
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Department of Clinical Medicine and Stomatology, School of Hangzhou Normal University, Hangzhou, China
| | - Yuanyuan Mai
- Basic Medical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Yue Zhao
- School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Luxi Cao
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Shuiyu Ji
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Bin Zhu
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Quanquan Shen
- Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Department of Nephrology, Zhejiang Provincial People’s Hospital Bijie Hospital, Bijie, China
| |
Collapse
|
24
|
Yin Y, Yang Y, Zhang Y, Shang Y, Li Q, Yuan J. MiR-132-3p suppresses peritoneal fibrosis induced by peritoneal dialysis via targeting TGF-β1/Smad2/3 signaling pathway. PLoS One 2024; 19:e0301540. [PMID: 38603722 PMCID: PMC11008817 DOI: 10.1371/journal.pone.0301540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Peritoneal fibrosis (PF) is the main complication of peritoneal dialysis (PD) and the most common cause of cessation from PD. There is still no effective therapeutic approach to reserve PF. We aimed to investigate the role of miR-132-3p and underlying potential mechanisms in PF. METHODS A total of 18 Sprague-Dawley (SD) rats were divided randomly into three groups (n = 6): (i)Control group (ii)PF group (iii)PF+Losartan group; Rats in the PF group and PF+Losartan group received daily intraperitoneal injections of 3 mg/kg chlorhexidine for 14 days, and rats in the PF+Losartan group simultaneously received daily intraperitoneal injections of 2 mg/kg losartan for 14 days. The control group was injected with saline in the same volume. Met-5A cells were treated for 24h with TGF-β1 dissolved in recombinant buffered saline at a concentration of 10 ng/ml, meanwhile, PBS solution as a negative control. The human peritoneal solution was collected for the detection of miR-132-3p. RESULTS In vivo, SD rats were infused with chlorhexidine to establish PF model, and we found that miR-132-3p significantly decreased and the expressions of transforming growth factor-β1 (TGF-β1), and Smad2/3 were up-regulated in PF. In vitro, miR-132-3p mimics suppressed TGF-β1/Smad2/3 activity, whereas miR-132-3p inhibition activated the pathway. In human peritoneal solution, we found that the expression of miR-132-3p decreased in a time-dependent model and its effect became more pronounced with longer PD duration. CONCLUSION MiR-132-3p ameliorated PF by suppressing TGF-β1/Smad2/3 activity, suggesting that miR-132-3p represented a potential therapeutic approach for PF.
Collapse
Affiliation(s)
- Yangyang Yin
- School of Medicine, Guizhou University, Guiyang, Guizhou, China
- Department of Nephrology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Yuqi Yang
- Department of Nephrology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Yongqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yu Shang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Qian Li
- Department of Nephrology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Jing Yuan
- School of Medicine, Guizhou University, Guiyang, Guizhou, China
- Department of Nephrology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| |
Collapse
|
25
|
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
|
26
|
Ozaki T, Fu HY, Onishi K, Yokoyama S, Fujita T, Tobiume A, Sofue T, Akimitsu K, Minamino T. Partial replacement of d-glucose with d-allose ameliorates peritoneal injury and hyperglycaemia induced by peritoneal dialysis fluid in rats. Perit Dial Int 2024; 44:125-132. [PMID: 37525525 DOI: 10.1177/08968608231184354] [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: 08/02/2023] Open
Abstract
BACKGROUND Peritoneal dialysis (PD) is a crucial dialysis method for treating end-stage kidney disease. However, its use is restricted due to high glucose-induced peritoneal injury and hyperglycaemia, particularly in patients with diabetes mellitus. In this study, we investigated whether partially replacing d-glucose with the rare sugar d-allose could ameliorate peritoneal injury and hyperglycaemia induced by peritoneal dialysis fluid (PDF). METHODS Rat peritoneal mesothelial cells (RPMCs) were exposed to a medium containing d-glucose or d-glucose partially replaced with different concentrations of d-allose. Cell viability, oxidative stress and cytokine production were evaluated. Sprague-Dawley (SD) rats were administrated saline, a PDF containing 4% d-glucose (PDF-G4.0%) or a PDF containing 3.6% d-glucose and 0.4% d-allose (PDF-G3.6%/A0.4%) once a day for 4 weeks. Peritoneal injury and PD efficiency were assessed using immuno-histological staining and peritoneal equilibration test, respectively. Blood glucose levels were measured over 120 min following a single injection of saline or PDFs to 24-h fasted SD rats. RESULTS In RPMCs, the partial replacement of d-glucose with d-allose increased cell viability and decreased oxidative stress and cytokine production compared to d-glucose alone. Despite the PDF-G3.6%/A0.4% having a lower d-glucose concentration compared to PDF-G4.0%, there were no significant changes in osmolality. When administered to SD rats, the PDF-G3.6%/A0.4% suppressed the elevation of peritoneal thickness and blood d-glucose levels induced by PDF-G4.0%, without impacting PD efficiency. CONCLUSIONS Partial replacement of d-glucose with d-allose ameliorated peritoneal injury and hyperglycaemia induced by high concentration of d-glucose in PDF, indicating that d-allose could be a potential treatment option in PD.
Collapse
Affiliation(s)
- Taro Ozaki
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Miki, Japan
- Department of Nephrology, Sakaide City Hospital, Kagawa, Japan
- These authors contributed equally
| | - Hai Ying Fu
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Miki, Japan
- These authors contributed equally
| | - Keisuke Onishi
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Miki, Japan
| | - Shota Yokoyama
- Department of Cardiology, Mizushima Central Hospital, Kurashiki, Okayama, Japan
| | - Takuro Fujita
- Department of Nephrology, Kaifu Hospital, Takamatsu, Kagawa, Japan
| | - Atsushi Tobiume
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Miki, Japan
| | - Tadashi Sofue
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Miki, Japan
| | - Kazuya Akimitsu
- International Institute of Rare Sugar Research and Education & Faculty of Agriculture, Kagawa University, Miki, Japan
| | - Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Miki, Japan
| |
Collapse
|
27
|
Nakamata J, Morimoto H, Baba R, Kokubu K, Miyamoto T. Glucose Induces ER Stress Response-Mediated Peritoneal Mesothelial Cell Death. Acta Histochem Cytochem 2024; 57:7-14. [PMID: 38463207 PMCID: PMC10918429 DOI: 10.1267/ahc.23-00050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/27/2023] [Indexed: 03/12/2024] Open
Abstract
Peritoneal dialysis (PD) fluid, which contains a high concentration of glucose, is involved in peritoneal damage after long-term use. The mechanisms through which glucose induces damage to the mesothelium have not been clearly elucidated. Although, endoplasmic reticulum (ER) stress response is associated with several diseases, the involvement of ER stress in peritoneal damage has not yet been demonstrated. Primary-cultured rat peritoneal mesothelial cells (RPMCs) and rat PD model were used to investigate the influence of glucose on the peritoneum. Cells treated with glucose were examined for cytotoxicity, induction of apoptosis, and activation of the ER stress pathway. Glucose treatment of RPMCs induced cell death at concentrations higher than 3%. Annexin V positive, that is a feature of apoptosis, occurred in dead cells. Treatment with glucose led to the activation of protein kinase R-like ER kinase (PERK) and eukaryotic translation initiation factor-2α (eIF-2α). Glucose also induced the expression and nuclear translocation of homologous protein C/EBP. Cell death was rescued by the integrated stress response inhibitor, ISRIB, which suppresses the integrated stress response pathway, including ER stress. Glucose in PD fluid induces PERK/eIF-2α-mediated ER stress in RPMCs, resulting in apoptosis. This cellular stress may cause peritoneal damage in patients receiving PD.
Collapse
Affiliation(s)
- Junichi Nakamata
- Second Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807-8555, Japan
- Present affiliation: Ashiya Central Hospital, 283-7, Yamaga, Ashiya, Onga, Fukuoka 807-0141, Japan
| | - Hiroyuki Morimoto
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807-8555, Japan
| | - Ryoko Baba
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807-8555, Japan
| | - Keiji Kokubu
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807-8555, Japan
| | - Tetsu Miyamoto
- Second Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807-8555, Japan
| |
Collapse
|
28
|
Zhang J, Li H, Zhong H, Chen X, Hu ZX. Omega-3 polyunsaturated fatty acids protect peritoneal mesothelial cells from hyperglycolysis and mesothelial-mesenchymal transition through the FFAR4/CaMKKβ/AMPK/mTOR signaling pathway. Int Immunopharmacol 2024; 128:111561. [PMID: 38262160 DOI: 10.1016/j.intimp.2024.111561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/25/2024]
Abstract
Peritoneal fibrosis is a severe clinical complication associated with peritoneal dialysis (PD) and impacts its efficacy and patient outcomes. The process of mesothelial-mesenchymal transition (MMT) in peritoneal mesothelial cells plays a pivotal role in fibrogenesis, whereas metabolic reprogramming, characterized by excessive glycolysis, is essential in MMT development. No reliable therapies are available despite substantial progress made in understanding the mechanisms underlying peritoneal fibrosis. Protective effect of omega-3 polyunsaturated fatty acids (ω3 PUFAs) has been described in PD-induced peritoneal fibrosis, although the detailed mechanisms remain unknown. It is known that ω3 PUFAs bind to and activate the free fatty acid receptor 4 (FFAR4). However, the expression and role of FFAR4 in the peritoneum have not been investigated. Thus, we hypothesized that ω3 PUFAs would alleviate peritoneal fibrosis by inhibiting hyperglycolysis and MMT through FFAR4 activation. First, we determined FFAR4 expression in peritoneal mesothelium in humans and mice. FFAR4 expression was abnormally decreased in patients on PD and mice and HMrSV5 mesothelial cells exposed to PD fluid (PDF); this change was restored by the ω3 PUFAs (EPA and DHA). ω3 PUFAs significantly inhibited peritoneal hyperglycolysis, MMT, and fibrosis in PDF-treated mice and HMrSV5 mesothelial cells; these changes induced by ω3 PUFAs were blunted by treatment with the FFAR4 antagonist AH7614 and FFAR4 siRNA. Additionally, ω3 PUFAs induced FFAR4, Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ), and AMPK and suppressed mTOR, leading to the inhibition of hyperglycolysis, demonstrating that the ω3 PUFAs-mediated FFAR4 activation ameliorated peritoneal fibrosis by inhibiting hyperglycolysis and MMT via CaMKKβ/AMPK/mTOR signaling. As natural FFAR4 agonists, ω3 PUFAs may be considered for the treatment of PD-associated peritoneal fibrosis.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Li
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Zhong
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhang-Xue Hu
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China; National Clinical Research Center for Geriatrics and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
29
|
Sheng L, Shan Y, Dai H, Yu M, Sun J, Huang L, Wang F, Sheng M. Intercellular communication in peritoneal dialysis. Front Physiol 2024; 15:1331976. [PMID: 38390449 PMCID: PMC10882094 DOI: 10.3389/fphys.2024.1331976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Long-term peritoneal dialysis (PD) causes structural and functional alterations of the peritoneal membrane. Peritoneal deterioration and fibrosis are multicellular and multimolecular processes. Under stimulation by deleterious factors such as non-biocompatibility of PD solution, various cells in the abdominal cavity show differing characteristics, such as the secretion of different cytokines, varying protein expression levels, and transdifferentiation into other cells. In this review, we discuss the role of various cells in the abdominal cavity and their interactions in the pathogenesis of PD. An in-depth understanding of intercellular communication and inter-organ communication in PD will lead to a better understanding of the pathogenesis of this disease, enabling the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Li Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Shan
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Huibo Dai
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Manshu Yu
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinyi Sun
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liyan Huang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Funing Wang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meixiao Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
30
|
Ruan H, Li X, Zhou L, Zheng Z, Hua R, Wang X, Wang Y, Fan Y, Guo S, Wang L, Ur Rahman S, Wang Z, Wei Y, Yu S, Zhang R, Cheng Q, Sheng J, Li X, Liu X, Yuan R, Zhang X, Chen L, Xu G, Guan Y, Nie J, Qin H, Zheng F. Melatonin decreases GSDME mediated mesothelial cell pyroptosis and prevents peritoneal fibrosis and ultrafiltration failure. SCIENCE CHINA. LIFE SCIENCES 2024; 67:360-378. [PMID: 37815699 DOI: 10.1007/s11427-022-2365-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/12/2023] [Indexed: 10/11/2023]
Abstract
Peritoneal fibrosis together with increased capillaries is the primary cause of peritoneal dialysis failure. Mesothelial cell loss is an initiating event for peritoneal fibrosis. We find that the elevated glucose concentrations in peritoneal dialysate drive mesothelial cell pyroptosis in a manner dependent on caspase-3 and Gasdermin E, driving downstream inflammatory responses, including the activation of macrophages. Moreover, pyroptosis is associated with elevated vascular endothelial growth factor A and C, two key factors in vascular angiogenesis and lymphatic vessel formation. GSDME deficiency mice are protected from high glucose induced peritoneal fibrosis and ultrafiltration failure. Application of melatonin abrogates mesothelial cell pyroptosis through a MT1R-mediated action, and successfully reduces peritoneal fibrosis and angiogenesis in an animal model while preserving dialysis efficacy. Mechanistically, melatonin treatment maintains mitochondrial integrity in mesothelial cells, meanwhile activating mTOR signaling through an increase in the glycolysis product dihydroxyacetone phosphate. These effects together with quenching free radicals by melatonin help mesothelial cells maintain a relatively stable internal environment in the face of high-glucose stress. Thus, Melatonin treatment holds some promise in preserving mesothelium integrity and in decreasing angiogenesis to protect peritoneum function in patients undergoing peritoneal dialysis.
Collapse
Affiliation(s)
- Hongxia Ruan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Xuejuan Li
- Department of Nephrology, Second Hospital, Dalian Medical University, Dalian, 116023, China.
- Wuhu Hospital and Health Science Center, East China Normal University, Shanghai, 200241, China.
| | - Lina Zhou
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Zihan Zheng
- Chongqing International Institute for Immunology, Chongqing, 401320, China
| | - Rulin Hua
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Xu Wang
- Department of Nephrology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518101, China
| | - Yuan Wang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Yujie Fan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Shuwen Guo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Lihua Wang
- Department of Nephrology, Second Hospital, Dalian Medical University, Dalian, 116023, China
| | - Shafiq Ur Rahman
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ziwei Wang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Yuyuan Wei
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Shuangyan Yu
- Department of Nephrology, Second Hospital, Dalian Medical University, Dalian, 116023, China
| | - Rongzhi Zhang
- Department of Nephrology, Second Hospital, Dalian Medical University, Dalian, 116023, China
| | - Qian Cheng
- Department of Nephrology, Second Hospital, Dalian Medical University, Dalian, 116023, China
| | - Jie Sheng
- Department of Nephrology, Second Hospital, Dalian Medical University, Dalian, 116023, China
| | - Xue Li
- Department of Nephrology, Second Hospital, Dalian Medical University, Dalian, 116023, China
| | - Xiaoyan Liu
- Department of Nephrology, Second Hospital, Dalian Medical University, Dalian, 116023, China
| | - Ruqiang Yuan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Xiaoyan Zhang
- Wuhu Hospital and Health Science Center, East China Normal University, Shanghai, 200241, China
| | - Lihong Chen
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
- Wuhu Hospital and Health Science Center, East China Normal University, Shanghai, 200241, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
- Wuhu Hospital and Health Science Center, East China Normal University, Shanghai, 200241, China
| | - Jing Nie
- Peking University First Hospital, Peking University, Beijing, 100034, China
| | - Hongqiang Qin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
| | - Feng Zheng
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
- Department of Nephrology, Second Hospital, Dalian Medical University, Dalian, 116023, China.
- Wuhu Hospital and Health Science Center, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
31
|
Wang L, Fan J, Yang T, Shen J, Wang L, Ge W. Investigating the therapeutic effects and mechanisms of Roxadustat on peritoneal fibrosis Based on the TGF-β/Smad pathway. Biochem Biophys Res Commun 2024; 693:149387. [PMID: 38145606 DOI: 10.1016/j.bbrc.2023.149387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/27/2023]
Abstract
Peritoneal fibrosis (PF) is particularly common in individuals undergoing peritoneal dialysis (PD). Fibrosis of the parenchymal tissue typically progresses slowly. Therefore, preventing and reducing the advancement of fibrosis is crucial for effective patient treatment. Roxadustat is a hypoxia-inducible factor prolyl hydroxylase inhibitor (HIF-PHI), primarily used to treat and improve renal anemia. Recent studies have found that HIF-1α possesses antioxidant activity and exerts a certain protective effect in ischemic heart disease and spinal cord injury, while it can also delay the progression of pulmonary and renal fibrosis. This study establishes the mice model through intraperitoneal injection of 4.25 % peritoneal dialysate fluid (PDF) and explores the therapeutic effects of Roxadustat by inducing TGF-β1-mediated epithelial-mesenchymal transition (EMT) in Met-5A cells. The aim is to investigate the protective role and mechanisms of Roxadustat against PD-related PF. We observed thicker peritoneal tissue and reduced permeability in animals with PD-related PF samples. This was accompanied by heightened inflammation, which Roxadustat alleviated by lowering the levels of inflammatory cytokines (IL-6, TNF-α). Furthermore, Roxadustat inhibited EMT in PF mice and TGF-β1-induced Met-5A cells, as evidenced by decreased expression of fibrotic markers, such as fibronectin, collagen I, and α-SMA, alongside an elevation in the expression of the epithelial marker, E-cadherin. Roxadustat also significantly decreased the expression of TGF-β1 and the phosphorylation of p-Smad2 and p-Smad3. In conclusion, Roxadustat ameliorates peritoneal fibrosis through the TGF-β/Smad pathway.
Collapse
Affiliation(s)
- Lingyun Wang
- Department of Pharmacy, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, China
| | - Jiangqing Fan
- Department of Pharmacy, China Pharmaceutical University Nanjing Drum Tower Hospital, China
| | - Ting Yang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, China
| | - Jizhong Shen
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, China.
| | - Lulu Wang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, China; Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, 410219, China.
| | - Weihong Ge
- Department of Pharmacy, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, China.
| |
Collapse
|
32
|
Dai H, Shan Y, Yu M, Wang F, Zhou Z, Sun J, Sheng L, Huang L, Sheng M. Network pharmacology, molecular docking and experimental verification of the mechanism of huangqi-jixuecao herb pair in treatment of peritoneal fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116874. [PMID: 37437794 DOI: 10.1016/j.jep.2023.116874] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/30/2023] [Accepted: 07/01/2023] [Indexed: 07/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Huangqi-Jixuecao herb pair (HQJXCHP) is a traditional herbal formula composed of two widely applied TCM prescriptions, Huangqi (Astragalus membranaceus (Fisch.) Bunge) and Jixuecao (Centella asiatica (L.) Urb.), used for hundreds of years to replenish qi and clear away heat. However, the therapeutic effects of HQJXCHP against peritoneal fibrosis (PF) and potential targets are currently unclear. AIMS OF THE STUDY The main objective of this study was preliminary prediction and validation of the effects and molecular mechanisms of action of HQJXCHP against PF based on network pharmacology analysis and experimental verification. MATERIALS AND METHODS The ingredients of HQJXCHP were analyzed via HPLC-Q-TOF/MS. Bioactive compounds of HQJXCHP used for network pharmacology analysis were obtained from the TCMSP database. HQJXCHP-related therapeutic targets in PF were obtained from the GeneCards, OMIM, Therapeutic Targets and PharmGkb databases. Therapeutic target-related signaling pathways were predicted via GO and KEGG pathway enrichment analyses. The targets of HQJXCHO were further validated in a PDS-induced PF mouse model in vivo and PMCs MMT model in vitro. RESULTS A total of 23 bioactive compounds of HQJXCHP related 188 target genes were retrieved. The HQJXCHP compound-target and PF-related target networks identified 131 common target genes. Subsequent protein-protein interaction (PPI) network analysis results disclosed Akt1, TP53, TNF, VEGFA and CASP3 as the top five key targets of HQJXCHP. Further molecular docking data revealed strong affinity of the two key compounds of HQJXCHP, quercetin and kaempferol, for these key targets. GO and KEGG pathway enrichment analyses further showed that PI3K/Akt, IL-17, TNF and TLR pathways contribute to the therapeutic effects of HQJXCHP on PF. An in vivo PDS-induced PF mouse model and in vitro PMCs mesothelial-to-mesenchymal transition (MMT) model with or without HQJXCHP intervention were used to confirm the effects and mechanisms of action of HQJXCHP. Western blot and qRT-PCR results showed that HQ, JXC and HQJXCHP reduced PDS-induced inflammatory cell aggregation and peritoneal thickening through suppressing the MMT process, among which HQJXCHP exerted the greatest therapeutic effect. Moreover, HQJXCHP inhibited activation of the PI3K/Akt, IL-17, TNF and TLR signaling pathways induced by PDS. CONCLUSIONS This is the first study to employ network pharmacology and molecular docking analyses to predict the targets of HQJXCHP with therapeutic effects on PDS-related PF. Data from in vivo and in vitro validation experiments collectively showed that HQJXCHP delays the PF process through inhibiting PI3K/Akt, IL-17, TNF and TLR signaling pathways. Overall, our findings highlight the successful application of network pharmacology theory to provide a scientific basis for clinical utility of HQJXCHP against PF.
Collapse
Affiliation(s)
- Huibo Dai
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Shan
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Manshu Yu
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Funing Wang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziren Zhou
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinyi Sun
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liyan Huang
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinic Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Meixiao Sheng
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
33
|
Shakour N, Karami S, Iranshahi M, Butler AE, Sahebkar A. Antifibrotic effects of sodium-glucose cotransporter-2 inhibitors: A comprehensive review. Diabetes Metab Syndr 2024; 18:102934. [PMID: 38154403 DOI: 10.1016/j.dsx.2023.102934] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/25/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND AND AIMS Scar tissue accumulation in organs is the underlying cause of many fibrotic diseases. Due to the extensive array of organs affected, the long-term nature of fibrotic processes and the large number of people who suffer from the negative impact of these diseases, they constitute a serious health problem for modern medicine and a huge economic burden on society. Sodium-glucose cotransporter-2 inhibitors (SGLT2is) are a relatively new class of anti-diabetic pharmaceuticals that offer additional benefits over and above their glucose-lowering properties; these medications modulate a variety of diseases, including fibrosis. Herein, we have collated and analyzed all available research on SGLT2is and their effects on organ fibrosis, together with providing a proposed explanation as to the underlying mechanisms. METHODS PubMed, ScienceDirect, Google Scholar and Scopus were searched spanning the period from 2012 until April 2023 to find relevant articles describing the antifibrotic effects of SGLT2is. RESULTS The majority of reports have shown that SGLT2is are protective against lung, liver, heart and kidney fibrosis as well as arterial stiffness. According to the results of clinical trials and animal studies, many SGLT2 inhibitors are promising candidates for the treatment of fibrosis. Recent studies have demonstrated that SGLT2is affect an array of cellular processes, including hypoxia, inflammation, oxidative stress, the renin-angiotensin system and metabolic activities, all of which have been linked to fibrosis. CONCLUSION Extensive evidence indicates that SGLT2is are promising treatments for fibrosis, demonstrating protective effects in various organs and influencing key cellular processes linked to fibrosis.
Collapse
Affiliation(s)
- Neda Shakour
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shima Karami
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Adliya, Bahrain
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
34
|
Si Z, Su W, Zhou Z, Li J, Su C, Zhang Y, Hu Z, Huang Z, Zhou H, Cong A, Zhou Z, Cao W. Hyperglycolysis in endothelial cells drives endothelial injury and microvascular alterations in peritoneal dialysis. Clin Transl Med 2023; 13:e1498. [PMID: 38037461 PMCID: PMC10689974 DOI: 10.1002/ctm2.1498] [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: 05/29/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Endothelial cell (EC) dysfunction leading to microvascular alterations is a hallmark of technique failure in peritoneal dialysis (PD). However, the mechanisms underlying EC dysfunction in PD are poorly defined. METHODS We combined RNA sequencing with metabolite set analysis to characterize the metabolic profile of peritoneal ECs from a mouse model of PD. This was combined with EC-selective blockade of glycolysis by genetic or pharmacological inhibition of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) in vivo and in vitro. We also investigated the association between peritoneal EC glycolysis and microvascular alterations in human peritoneal samples from patients with end-stage kidney disease (ESKD). RESULTS In a mouse model of PD, peritoneal ECs had a hyperglycolytic metabolism that shunts intermediates into nucleotide synthesis. Hyperglycolytic mouse peritoneal ECs displayed a unique active phenotype with increased proliferation, permeability and inflammation. The active phenotype of mouse peritoneal ECs can be recapitulated in human umbilical venous ECs and primary human peritoneal ECs by vascular endothelial growth factor that was released from high glucose-treated mesothelial cells. Importantly, reduction of peritoneal EC glycolysis, via endothelial deficiency of the glycolytic activator PFKFB3, inhibited PD fluid-induced increases in peritoneal capillary density, vascular permeability and monocyte extravasation, thereby protecting the peritoneum from the development of structural and functional damages. Mechanistically, endothelial PFKFB3 deficiency induced the protective effects in part by inhibiting cell proliferation, VE-cadherin endocytosis and monocyte-adhesion molecule expression. Pharmacological PFKFB3 blockade induced a similar therapeutic benefit in this PD model. Human peritoneal tissue from patients with ESKD also demonstrated evidence of increased EC PFKFB3 expression associated with microvascular alterations and peritoneal dysfunction. CONCLUSIONS These findings reveal a critical role of glycolysis in ECs in mediating the deterioration of peritoneal function and suggest that strategies targeting glycolysis in peritoneal ECs may be of therapeutic benefit for patients undergoing PD.
Collapse
Affiliation(s)
- Zekun Si
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Wenyan Su
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Zhuoyu Zhou
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Jinjin Li
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Cailing Su
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Ying Zhang
- Division of NephrologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouP. R. China
| | - Zuoyu Hu
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Zhijie Huang
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Hong Zhou
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Ansheng Cong
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Zhanmei Zhou
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| | - Wei Cao
- Division of NephrologyState Key Laboratory of Organ Failure ResearchGuangdong Provincial Key Laboratory of NephrologyGuangdong Provincial Clinical Research Center for Kidney DiseaseNanfang HospitalSouthern Medical UniversityGuangzhouP. R. China
| |
Collapse
|
35
|
Wang Q, Sun J, Wang R, Sun J. Inhibition of EZH2 mitigates peritoneal fibrosis and lipid precipitation in peritoneal mesothelial cells mediated by klotho. Ren Fail 2023; 45:2149411. [PMID: 36724065 PMCID: PMC9897791 DOI: 10.1080/0886022x.2022.2149411] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Peritoneal fibrosis caused by long-term peritoneal dialysis (PD) is the main reason why patients withdraw from PD treatment. Lipid accumulation in the peritoneum was shown to participate in fibrosis, and klotho is a molecule involved in lipid metabolism. GSK343 (enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) inhibitor) has been verified to inhibit epithelial mesenchymal transdifferentiation (EMT) and peritoneal fibrosis, but its related mechanism remains unclear. This study aimed to investigate whether lipid accumulation was involved in the effect of GSK343 and its related mechanism. MATERIALS AND METHODS First, the expression of EZH2, klotho and EMT indices in human peritoneal mesothelial cells (HMrSV5) incubated with high glucose (HG) levels was detected. After EZH2 was inhibited by GSK343, Western blot (WB), wound healing and Transwell assays were used to explore the effect of GSK343. EZH2 and klotho expression was also detected. Oil red O and Nile red staining and triglyceride (TG) detection kits were used to detect lipid accumulation. A rescue experiment with small interfering RNA specific for klotho (si-klotho) on the basis of GSK343 was also conducted to verify that GSK343 exerted its effect via klotho. In in vivo experiments, rats were administered GSK343, and the related index was assessed. RESULTS In our study, we revealed that the expression of EZH2 was significantly upregulated and klotho was significantly downregulated in HMrSV5 cells induced by high glucose. With the aid of GSK343, we found that lipid deposition caused by HG was significantly decreased. In addition, EMT and fibrosis were also significantly alleviated. Moreover, GSK343 could also restore the downregulation of klotho. To further verify whether klotho mediated the effect of EZH2, a rescue experiment with si-klotho was also conducted. The results showed that si-klotho could counteract the protective effect of GSK343 on high glucose-induced lipid accumulation and fibrosis. In vivo experiments also revealed that GSK343 could relieve peritoneal fibrosis, lipid deposition and EMT by mitigating EZH2 and restoring klotho expression. CONCLUSIONS Combining these findings, we found that EZH2 regulated lipid deposition, peritoneal fibrosis, and EMT mediated by klotho. To our knowledge, this is the first study to demonstrate the effect of the EZH2-klotho interaction on peritoneal fibrosis. Hence, EZH2 and klotho could act as potential targets for the treatment of peritoneal fibrosis.
Collapse
Affiliation(s)
- Qinglian Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China,Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Jingshu Sun
- Department of Nephrology, Weifang People’s Hospital, Weifang, Shandong, China
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China,Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Jing Sun
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China,Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China,CONTACT Jing Sun Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong250021, China
| |
Collapse
|
36
|
Wang J, Xin lv, Aniwan A, Liu H, Lin Y, Shao X, Zhou S, Yu P. O-GlcNAcylation regulates HIF-1α and induces mesothelial-mesenchymal transition and fibrosis of human peritoneal mesothelial cells. Heliyon 2023; 9:e22916. [PMID: 38144265 PMCID: PMC10746441 DOI: 10.1016/j.heliyon.2023.e22916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 12/26/2023] Open
Abstract
O-GlcNAcylation is a post-translational modification of proteins that regulates various biological processes. However, its involvement in peritoneal dialysis fibrosis remains unclear. This study aimed to investigate the impact of O-GlcNAcylation on human peritoneal mesothelial cells (HPMCs) cultured in control and high-glucose medium. To manipulate cellular conditions, we employed knockdown techniques targeting HIF-1α and OGT, along with the administration of pharmacological agents (PUGNAc, OSMI-1, MG-132, FG-4592, and HIF-1α inhibitor). Our findings revealed that elevated glucose levels increased global O-GlcNAcylation and the abundance of HIF-1α, α-SMA, fibronectin, and COL1A2. Conversely, the expression of E-Cadherin was decreased. Significantly, a positive correlation was observed between O-GlcNAcylation, HIF-1α, mesothelial-to-mesenchymal transition (MMT), and fibrosis in HPMCs. Notably, O-GlcNAcylation was found to regulate HIF-1α, thereby promoting MMT and fibrosis under high glucose conditions. Furthermore, we discovered that high glucose levels induced O-GlcNAcylation of HIF-1α, preventing its ubiquitination and proteasomal degradation. In summary, our study demonstrates the critical role of O-GlcNAcylation-mediated regulation of HIF-1α in MMT and fibrosis during peritoneal dialysis.
Collapse
Affiliation(s)
- Jian Wang
- Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Xin lv
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Ashanjiang Aniwan
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Hongyan Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Yao Lin
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Xian Shao
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Saijun Zhou
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Pei Yu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| |
Collapse
|
37
|
Tian J, Zhang R, Zhu N, Gu L, Guo Y, Yuan W. Association of serum thymosin β4 with malnutrition-inflammation-atherosclerosis syndrome in peritoneal dialysis patients: a cross-sectional study. Ren Fail 2023; 45:2202761. [PMID: 37133832 PMCID: PMC10158543 DOI: 10.1080/0886022x.2023.2202761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND Malnutrition-inflammation-atherosclerosis (MIA) syndrome may worsen the prognosis of peritoneal dialysis (PD) patients. Serum thymosin β4 (sTβ4) protects against inflammation, fibrosis and cardiac dysfunction. OBJECTIVES The present study aimed to characterize the association between sTβ4 and MIA syndrome as well as to investigate the potential of regulating sTβ4 to improve the prognosis of PD patients. METHODS We performed a cross-sectional, single-center pilot study involving 76 PD patients. Demographic characteristics, clinical characteristics, nutritional profiles, inflammatory mediators, atherosclerosis-related factors and sTβ4 levels were collected and subjected to association analysis for sTβ4 and MIA syndrome. RESULTS sTβ4 levels did not significantly vary with sex or primary disease in PD patients. Ages and PD features did not vary between patients with different levels of sTβ4. PD patients with higher levels of sTβ4 had significantly higher levels of nutritional indicators, including subjective global nutritional assessment (SGA) (p < 0.001) and serum albumin (ALB) (p < 0.001) but lower levels of inflammatory and atherosclerotic indicators, including serum C reaction protein (CRP) (p = 0.009), the right common carotid artery (RCCA) intimal thickness (p < 0.001) and the left common carotid artery (LCCA) intimal thickness (p = 0.02). Correlation analysis showed that sTβ4 was positively associated with SGA (p < 0.001) and serum ALB (p < 0.001) but negatively associated with CRP (p = 0.020), RCCA intimal thickness (p < 0.001) and LCCA intimal thickness (p = 0.033). In multiple adjusted models, the prevalence of MIA syndrome was significantly decreased in PD patients with increased levels of sTβ4 when patients without MIA syndrome were compared to those with all indicators of MIA syndrome (OR = 0.996, 95% CI 0.993-0.999, p = 0.003) or those with at least one indicator of MIA syndrome (OR = 0.997, 95% CI 0.995-0.998, p < 0.001). CONCLUSIONS The sTβ4 level decreases in PD patients with MIA syndrome. The prevalence of MIA syndrome decreases significantly as the level of sTβ4 increases in PD patients.
Collapse
Affiliation(s)
- Jiakun Tian
- Department of Nephrology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Zhang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Zhu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijie Gu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunshan Guo
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weijie Yuan
- Department of Nephrology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
38
|
Marchant V, Trionfetti F, Tejedor-Santamaria L, Rayego-Mateos S, Rotili D, Bontempi G, Domenici A, Menè P, Mai A, Martín-Cleary C, Ortiz A, Ramos AM, Strippoli R, Ruiz-Ortega M. BET Protein Inhibitor JQ1 Ameliorates Experimental Peritoneal Damage by Inhibition of Inflammation and Oxidative Stress. Antioxidants (Basel) 2023; 12:2055. [PMID: 38136175 PMCID: PMC10740563 DOI: 10.3390/antiox12122055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Peritoneal dialysis (PD) is a current replacement therapy for end-stage kidney diseases (ESKDs). However, long-term exposure to PD fluids may lead to damage of the peritoneal membrane (PM) through mechanisms involving the activation of the inflammatory response and mesothelial-to-mesenchymal transition (MMT), leading to filtration failure. Peritoneal damage depends on a complex interaction among external stimuli, intrinsic properties of the PM, and subsequent activities of the local innate-adaptive immune system. Epigenetic drugs targeting bromodomain and extra-terminal domain (BET) proteins have shown beneficial effects on different experimental preclinical diseases, mainly by inhibiting proliferative and inflammatory responses. However the effect of BET inhibition on peritoneal damage has not been studied. To this aim, we have evaluated the effects of treatment with the BET inhibitor JQ1 in a mouse model of peritoneal damage induced by chlorhexidine gluconate (CHX). We found that JQ1 ameliorated the CHX-induced PM thickness and inflammatory cell infiltration. Moreover, JQ1 decreased gene overexpression of proinflammatory and profibrotic markers, together with an inhibition of the nuclear factor-κB (NF-κB) pathway. Additionally, JQ1 blocked the activation of nuclear factor erythroid 2-related factor 2 (NRF2) and restored changes in the mRNA expression levels of NADPH oxidases (NOX1 and NOX4) and NRF2/target antioxidant response genes. To corroborate the in vivo findings, we evaluated the effects of the BET inhibitor JQ1 on PD patients' effluent-derived primary mesothelial cells and on the MeT-5A cell line. JQ1 inhibited tumor necrosis factor-α (TNF-α)-induced proinflammatory gene upregulation and restored MMT phenotype changes, together with the downmodulation of oxidative stress. Taken together, these results suggest that BET inhibitors may be a potential therapeutic option to ameliorate peritoneal damage.
Collapse
Affiliation(s)
- Vanessa Marchant
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (V.M.); (L.T.-S.); (S.R.-M.)
- RICORS2040, 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Flavia Trionfetti
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, 00149 Rome, Italy; (F.T.); (G.B.); (R.S.)
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Lucia Tejedor-Santamaria
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (V.M.); (L.T.-S.); (S.R.-M.)
- RICORS2040, 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Sandra Rayego-Mateos
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (V.M.); (L.T.-S.); (S.R.-M.)
- RICORS2040, 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (D.R.); (A.M.)
| | - Giulio Bontempi
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, 00149 Rome, Italy; (F.T.); (G.B.); (R.S.)
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessandro Domenici
- Renal Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy; (A.D.); (P.M.)
| | - Paolo Menè
- Renal Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Rome, Italy; (A.D.); (P.M.)
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (D.R.); (A.M.)
| | - Catalina Martín-Cleary
- Laboratory of Nephrology, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain;
| | - Alberto Ortiz
- RICORS2040, 28029 Madrid, Spain; (A.O.); (A.M.R.)
- Laboratory of Nephrology, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain;
| | - Adrian M. Ramos
- RICORS2040, 28029 Madrid, Spain; (A.O.); (A.M.R.)
- Laboratory of Nephrology, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain;
| | - Raffaele Strippoli
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, 00149 Rome, Italy; (F.T.); (G.B.); (R.S.)
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Marta Ruiz-Ortega
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, IIS-Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (V.M.); (L.T.-S.); (S.R.-M.)
- RICORS2040, 28029 Madrid, Spain; (A.O.); (A.M.R.)
| |
Collapse
|
39
|
Liang J, Cheng L, Feng J, Han Z, Huang C, Xie F, Li Y, Luo X, Wang Q, He J, Chen H. Molecular mechanism of Danshenol C in reversing peritoneal fibrosis: novel network pharmacological analysis and biological validation. BMC Complement Med Ther 2023; 23:361. [PMID: 37833759 PMCID: PMC10571429 DOI: 10.1186/s12906-023-04170-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023] Open
Abstract
OBJECTIVE The primary objective of this study is to elucidate the molecular mechanism underlying the reversal of peritoneal fibrosis (PF) by Danshenol C, a natural compound derived from the traditional Chinese medicine Salvia miltiorrhiza. By comprehensively investigating the intricate interactions and signaling pathways involved in Danshenol C's therapeutic effects on PF, we aim to unveil novel insights into its pharmacological actions. This investigation holds the potential to revolutionize the clinical application of Salvia miltiorrhiza in traditional Chinese medicine, offering promising new avenues for the treatment of PF and paving the way for evidence-based therapeutic interventions. METHODS Firstly, we utilized the YaTCM database to retrieve the structural formula of Danshenol C, while the SwissTargetPrediction platform facilitated the prediction of its potential drug targets. To gain insights into the genetic basis of PF, we acquired the GSE92453 dataset and GPL6480-9577 expression profile from the GEO database, followed by obtaining disease-related genes of PF from major disease databases. R software was then employed to screen for DEG associated with PF. To explore the intricate interactions between Danshenol C's active component targets, we utilized the String database and Cytoscape3.7.2 software to construct a PPI network. Further analysis in Cytoscape3.7.2 enabled the identification of core modules within the PPI network, elucidating key targets and molecular pathways critical to Danshenol C's therapeutic actions. Subsequently, we employed R to perform GO and KEGG pathway enrichment analyses, providing valuable insights into the functional implications and potential biological mechanisms of Danshenol C in the context of PF. To investigate the binding interactions between the core active components and key targets, we conducted docking studies using Chem3D, autoDock1.5.6, SYBYL2.0, and PYMOL2.4 software. We applied in vivo and in vitro experiments to prove that Danshenol C can improve PF. In order to verify the potential gene and molecular mechanism of Danshenol C to reverse PF, we used quantitative PCR, western blot, and apoptosis, ensuring robust and reliable verification of the results. RESULTS ① Wogonin, sitosterol, and Signal Transducer and Activator of Transcription 5 (STAT5) emerged as the most significant constituents among the small-molecule active compounds and gene targets investigated. ②38 targets intersected with the disease, among which MAPK14, CASP3, MAPK8 and STAT3 may be the key targets; The results of GO and KEGG analysis showed that there was a correlation between inflammatory pathway and Apoptosis. ④Real-time PCR showed that the mRNA expressions of MAPK8 (JNK1), MAPK14 (P38) and STAT3 were significantly decreased after Danshenol C treatment (P < 0.05), while the mRNA expression of CASP3 was significantly increased (P < 0.05)⑤Western blot showed that protein expressions of CASP3 and MAPK14 were significantly increased (P < 0.05), while the expression of STAT3 and MAPK8 was decreased after Danshenol C treatment (P < 0.05). ⑥There was no significant difference in flow analysis of apoptosis among groups. CONCLUSION The findings suggest that Danshenol C may modulate crucial molecular pathways, including the MAPK, Apoptosis, Calcium signaling, JAK-STAT signaling, and TNF signaling pathways. This regulation is mediated through the modulation of core targets such as STAT3, MAPK14, MAPK8, CASP3, and others. By targeting these key molecular players, Danshenol C exhibits the potential to regulate cellular responses to chemical stress and inflammatory stimuli. The identification of these molecular targets and pathways represents a significant step forward in understanding the molecular basis of Danshenol C's therapeutic effects in PF. This preliminary exploration provides novel avenues for the development of anti-PF treatment strategies and the discovery of potential therapeutic agents. By targeting specific core targets and pathways, Danshenol C opens up new possibilities for the development of more effective and targeted drugs to combat PF. These findings have the potential to transform the landscape of PF treatment and offer valuable insights for future research and drug development endeavors.
Collapse
Affiliation(s)
- Jiabin Liang
- Guangzhou Panyu Central Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lulu Cheng
- Guangzhou Panyu Central Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jie Feng
- Radiology Department of Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zeping Han
- Guangzhou Panyu Central Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chen Huang
- Guangzhou Panyu Central Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
- Medical Imaging Institute of Panyu, Guangzhou, China
| | - Fangmei Xie
- Guangzhou Panyu Central Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongsheng Li
- Guangzhou Municipality Tianhe Nuoya Bio-Engineering Co., Ltd, Guangzhou, China
| | - Xun Luo
- Kerry Rehabilitation Medicine Research Institute, Shenzhen, China
| | - Qingmei Wang
- Stroke Biological Recovery Laboratory, Teaching Affiliate of Harvard Medical School, Spaulding Rehabilitation Hospital, Charlestown, MA, USA
| | - Jinhua He
- Guangzhou Panyu Central Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Hanwei Chen
- Guangzhou Panyu Central Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
- Medical Imaging Institute of Panyu, Guangzhou, China.
- Panyu Health Management Center (Panyu Rehabilitation Hospital), Guangzhou, China.
| |
Collapse
|
40
|
Bai Y, Wang L, TingYang, Wang L, Ge W. Silymarin ameliorates peritoneal fibrosis by inhibiting the TGF-β/Smad signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2379-2391. [PMID: 37052642 DOI: 10.1007/s00210-023-02450-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 02/24/2023] [Indexed: 04/14/2023]
Abstract
Peritoneal dialysis (PD) is the mainstay of treatment for renal failure replacement therapy. Although PD has greatly improved the quality of life of end-stage renal disease (ESRD) patients, long-term PD can lead to ultrafiltration failure, which in turn causes peritoneal fibrosis (PF). Silymarin (SM) is a polyphenolic flavonoid isolated from the milk thistle (Silybum marianum) species that has a variety of pharmacological actions, including antioxidant, anti-inflammatory, antiviral, and anti-fibrotic pharmacological activities. However, the effect of SM on PF and its potential mechanisms have not been clarified. The aim of this study was to investigate the preventive effect of SM on PF in vitro and in vivo as well as elucidate the underlying mechanisms. We established PF mouse models and human pleural mesothelial cell fibrosis in vitro by intraperitoneal injection of high-glucose peritoneal dialysis solution (PDS) or transforming growth factor-β1 (TGF-β1), and evaluated the effect of SM on peritoneal fibrosis in vivo and in vitro. We found that SM alleviated peritoneal dysfunction. Meanwhile, SM inhibited the expression of fibrotic markers (TGF-β1, collagen I, fibronectin) and restored the expression of E-cadherin, BMP-7 in PF mice and TGF-β1-treated Met-5A cells. Furthermore, SM markedly down-regulated the expression of TGF-β1, p-Smad2, and p-Smad3 and up-regulated the expression of smad7. In conclusion, these findings suggested that SM may be an efficient and novel therapy for the prevention of PF through inhibition of TGF-β/Smad signaling.
Collapse
Affiliation(s)
- Yingwen Bai
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Lulu Wang
- Nanjing Drum Tower Hospital, Nanjing, 210008, Jiangsu Province, China
| | - TingYang
- Nanjing Drum Tower Hospital, Nanjing, 210008, Jiangsu Province, China
| | - Lingyun Wang
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Weihong Ge
- Nanjing Drum Tower Hospital, Nanjing, 210008, Jiangsu Province, China.
| |
Collapse
|
41
|
Li S, Luo C, Chen S, Zhuang Y, Ji Y, Zeng Y, Zeng Y, He X, Xiao J, Wang H, Chen X, Long H, Peng F. Brahma-related gene 1 acts as a profibrotic mediator and targeting it by micheliolide ameliorates peritoneal fibrosis. J Transl Med 2023; 21:639. [PMID: 37726857 PMCID: PMC10510267 DOI: 10.1186/s12967-023-04469-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/21/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Progressive peritoneal fibrosis is a worldwide public health concern impacting patients undergoing peritoneal dialysis (PD), yet there is no effective treatment. Our previous study revealed that a novel compound, micheliolide (MCL) inhibited peritoneal fibrosis in mice. However, its mechanism remains unclear. Brahma-related gene 1 (BRG1) is a key contributor to organ fibrosis, but its potential function in PD-related peritoneal fibrosis and the relationship between MCL and BRG1 remain unknown. METHODS The effects of MCL on BRG1-induced fibrotic responses and TGF-β1-Smads pathway were examined in a mouse PD model and in vitro peritoneal mesothelial cells. To investigate the targeting mechanism of MCL on BRG1, coimmunoprecipitation, MCL-biotin pulldown, molecular docking and cellular thermal shift assay were performed. RESULTS BRG1 was markedly elevated in a mouse PD model and in peritoneal mesothelial cells cultured in TGF-β1 or PD fluid condition. BRG1 overexpression in vitro augmented fibrotic responses and promoted TGF-β1-increased-phosphorylation of Smad2 and Smad3. Meanwhile, knockdown of BRG1 diminished TGF-β1-induced fibrotic responses and blocked TGF-β1-Smad2/3 pathway. MCL ameliorated BRG1 overexpression-induced peritoneal fibrosis and impeded TGF-β1-Smad2/3 signaling pathway both in a mouse PD model and in vitro. Mechanically, MCL impeded BRG1 from recognizing and attaching to histone H3 lysine 14 acetylation by binding to the asparagine (N1540) of BRG1, in thus restraining fibrotic responses and TGF-β1-Smad2/3 signaling pathway. After the mutation of N1540 to alanine (N1540A), MCL was unable to bind to BRG1 and thus, unsuccessful in suppressing BRG1-induced fibrotic responses and TGF-β1-Smad2/3 signaling pathway. CONCLUSION Our research indicates that BRG1 may be a crucial mediator in peritoneal fibrosis and MCL targeting N1540 residue of BRG1 may be a novel therapeutic strategy to combat PD-related peritoneal fibrosis.
Collapse
Affiliation(s)
- Shuting Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Congwei Luo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Sijia Chen
- Department of Nephrology and Rheumatology, The First Hospital of Changsha, Changsha, China
| | - Yiyi Zhuang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yue Ji
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yiqun Zeng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yao Zeng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaoyang He
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jing Xiao
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Huizhen Wang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaowen Chen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Haibo Long
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Fenfen Peng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
42
|
Zhao H, Zhang HL, Jia L. High glucose dialysate-induced peritoneal fibrosis: Pathophysiology, underlying mechanisms and potential therapeutic strategies. Biomed Pharmacother 2023; 165:115246. [PMID: 37523983 DOI: 10.1016/j.biopha.2023.115246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023] Open
Abstract
Peritoneal dialysis is an efficient renal replacement therapy for patients with end-stage kidney disease. However, continuous exposure of the peritoneal membrane to dialysate frequently leads to peritoneal fibrosis, which alters the function of the peritoneal membrane and results in withdrawal from peritoneal dialysis in patients. Among others, high glucose dialysate is considered as a predisposing factor for peritoneal fibrosis in patients on peritoneal dialysis. Glucose-induced inflammation, metabolism disturbance, activation of the renin-angiotensin-aldosterone system, angiogenesis and noninflammation-induced reactive oxygen species are implicated in the pathogenesis of high glucose dialysate-induced peritoneal fibrosis. Specifically, high glucose causes chronic inflammation and recurrent peritonitis, which could cause migration and polarization of inflammatory cells, as well as release of cytokines and fibrosis. High glucose also interferes with lipid metabolism and glycolysis by activating the sterol-regulatory element-binding protein-2/cleavage-activating protein pathway and increasing hypoxia inducible factor-1α expression, leading to angiogenesis and peritoneal fibrosis. Activation of the renin-angiotensin-aldosterone system and Ras-mitogen activated protein kinase signaling pathway is another contributing factor in high glucose dialysate-induced fibrosis. Ultimately, activation of the transforming growth factor-β1/Smad pathway is involved in mesothelial-mesenchymal transition or epithelial-mesenchymal transition, which leads to the development of fibrosis. Although possible intervention strategies for peritoneal dialysate-induced fibrosis by targeting the transforming growth factor-β1/Smad pathway have occasionally been proposed, lack of laboratory evidence renders clinical decision-making difficult. We therefore aim to revisit the upstream pathways of transforming growth factor-beta1/Smad and propose potential therapeutic targets for high glucose-induced peritoneal fibrosis.
Collapse
Affiliation(s)
- Hanxue Zhao
- First Clinical Medical College, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Beijing 100053, China
| | - Hong-Liang Zhang
- Department of Life Sciences, National Natural Science Foundation of China, No. 83 Shuangqing Road, Beijing 100085, China.
| | - Linpei Jia
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Beijing 100053, China.
| |
Collapse
|
43
|
Mo M, Zeng Y, Zeng Y, Li S, He X, Chen X, Luo Q, Liu M, Luo C, Dou X, Peng F, Long H. N-methylpiperazine-diepoxyovatodiolide ameliorates peritoneal fibrosis via suppressing TGF-β/Smad and JAK/STAT signaling pathway. Chem Biol Interact 2023; 382:110589. [PMID: 37268199 DOI: 10.1016/j.cbi.2023.110589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/04/2023]
Abstract
Peritoneal fibrosis (PF) is the main cause of peritoneal ultrafiltration failure in patients undergoing long-term peritoneal dialysis (PD). Epithelial-mesenchymal transition (EMT) is the key pathogenesis of PF. However, currently, no specific treatments are available to suppress PF. N-methylpiperazine-diepoxyovatodiolide (NMPDOva) is a newly synthesized compound that involves a chemical modification of ovatodiolide. In this study, we aimed to explore the antifibrotic effects of NMPDOva in PD-related PF and underlying mechanisms. A mouse model of PD-related PF was established via daily intraperitoneal injection of 4.25% glucose PD fluid. In vitro studies were performed using the transforming growth factor-beta1 (TGF-β1)-stimulated HMrSV5 cell line. Pathological changes were observed, and fibrotic markers were significantly elevated in the peritoneal membrane in mice model of PD-related PF. However, NMPDOva treatment significantly alleviated PD-related PF by decreasing the extracellular matrix accumulation. NMPDOva treatment decreased the expression of fibronectin, collagen Ⅰ, and alpha-smooth muscle actin (α-SMA) in mice with PD-related PF. Moreover, NMPDOva could alleviate TGF-β1-induced EMT in HMrSV5 cells, inhibited phosphorylation and nuclear translocation of Smad2/3, and increased the expression of Smad7. Meanwhile, NMPDOva inhibited phosphorylation of JAK2 and STAT3. Collectively, these results indicated that NMPDOva prevents PD-related PF by inhibiting the TGF-β1/Smad and JAK/STAT signaling pathway. Therefore, because of these antifibrotic effects, NMPDOva may be a promising therapeutic agent for PD-related PF.
Collapse
Affiliation(s)
- Min Mo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, 528308, China
| | - Yao Zeng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yiqun Zeng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Shuting Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaoyang He
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaowen Chen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Qimei Luo
- Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, 528308, China
| | - Mi Liu
- Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, 528308, China
| | - Congwei Luo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xianrui Dou
- Department of Nephrology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, 528308, China.
| | - Fenfen Peng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Haibo Long
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
44
|
Lu J, Gao J, Sun J, Wang H, Sun H, Huang Q, Zhang Y, Zhong S. Apolipoprotein A-I attenuates peritoneal fibrosis associated with peritoneal dialysis by inhibiting oxidative stress and inflammation. Front Pharmacol 2023; 14:1106339. [PMID: 37576813 PMCID: PMC10422021 DOI: 10.3389/fphar.2023.1106339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Apolipoprotein A-I (apoA-I), 90% of which is present in high-density lipoprotein (HDL), is the main constituent of HDL, has anti-inflammatory and anti-oxidant properties, and has received extensive attention in anti-atherosclerosis. Yet little is known about apoA-I 's role in peritoneal dialysis. In this study, by analyzing PD patients (n = 81), we found that decreased apoA/HDL-C ratio is significantly associated with rapid decline in peritoneal function. Further studies were performed in animal experiments to determine the ascendancy of apolipoprotein A-I mimetic peptide (D-4F) on peritoneum, we found that D-4F administration reduced peritoneal fibrosis and peritoneal endothelial mesenchymal transformation (EMT) induced by high glucose peritoneal dialysate, such as N-cadherin, Fibronectin, Vimentin, and α-smooth muscle actin (α-SMA) expression decreased. In mechanism, D-4F can significantly inhibit Smad2/3 phosphorylation, which is the major pathway leading to fibrosis. Furthermore, D-4F treatment inhibited NADPH oxidase and thiobarbituric acid reactive substances (TBARS) expression, increased the activity of certain enzymes, such as superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). Finally, treatment with D-4F inhibits the expression of interleukins-6(IL-6), Interleukin-1β(IL-1β), and tumor necrosis factor-α(TNF-α). Taken together, based on the above research evidence, apoA-I and its peptide mimic may regulate the oxidative stress, TGF- β1/Smads signaling pathway and inflammatory response to reduce peritoneal fibrosis due to peritoneal dialysis.
Collapse
Affiliation(s)
- Jing Lu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jie Gao
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jing Sun
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Haiping Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Huijuan Sun
- Jinzhou First People’s Hospital, Dalian, China
| | - Qian Huang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yao Zhang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shuo Zhong
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
45
|
Suryantoro SD, Thaha M, Sutanto H, Firdausa S. Current Insights into Cellular Determinants of Peritoneal Fibrosis in Peritoneal Dialysis: A Narrative Review. J Clin Med 2023; 12:4401. [PMID: 37445436 DOI: 10.3390/jcm12134401] [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: 05/16/2023] [Revised: 06/16/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Peritoneal fibrosis is the final process of progressive changes in the peritoneal membrane due to chronic inflammation and infection. It is one of the main causes of discontinuation of peritoneal dialysis (PD), apart from peritonitis and cardiovascular complications. Over time, morphological changes occur in the peritoneal membranes of patients who use PD. Of those are mesothelial-to-mesenchymal transition (MMT), neoangiogenesis, sub-mesothelial fibrosis, and hyalinizing vasculopathy. Several key molecules are involved in the complex pathophysiology of peritoneal fibrosis, including advanced glycosylation end products (AGEs), transforming growth factor beta (TGF-β), and vascular endothelial growth factor (VEGF). This narrative review will first discuss the physiology of the peritoneum and PD. Next, the multifaceted pathophysiology of peritoneal fibrosis, including the effects of hyperglycemia and diabetes mellitus on the peritoneal membrane, and the promising biomarkers of peritoneal fibrosis will be reviewed. Finally, the current and future management of peritoneal fibrosis will be discussed, including the potential benefits of new-generation glucose-lowering medications to prevent or slow down the progression of peritoneal fibrosis.
Collapse
Affiliation(s)
- Satriyo Dwi Suryantoro
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Universitas Airlangga Hospital, Surabaya 60115, Indonesia
| | - Mochammad Thaha
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Universitas Airlangga Hospital, Surabaya 60115, Indonesia
| | - Henry Sutanto
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Sarah Firdausa
- Department of Internal Medicine, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh 23111, Indonesia
| |
Collapse
|
46
|
Abstract
Peritoneal fibrosis is an important cause of peritoneal dialysis (PD) discontinuation worldwide and is associated with high morbidity and mortality rate. Although the era of metagenomics has provided new insights into the interactions between the gut microbiota and fibrosis in various organs and tissues, its role in peritoneal fibrosis has rarely been discussed. This review provides a scientific rationale and points out the potential role of gut microbiota in peritoneal fibrosis. In addition, the interaction between the gut, circulatory, and peritoneal microbiota is highlighted, with an emphasis on the relationship to PD outcomes. More research is needed to elucidate the mechanisms underlying the role of gut microbiota in peritoneal fibrosis and potentially unveil new target options for the management of PD technique failure.
Collapse
Affiliation(s)
- Natalia Stepanova
- Department of Nephrology and Dialysis, State Institution “Institute of Nephrology of the National Academy of Medical Sciences of Ukraine”, Kyiv, Ukraine
| |
Collapse
|
47
|
Lu X, Wu K, Jiang S, Li Y, Wang Y, Li H, Li G, Liu Q, Zhou Y, Chen W, Mao H. Therapeutic mechanism of baicalein in peritoneal dialysis-associated peritoneal fibrosis based on network pharmacology and experimental validation. Front Pharmacol 2023; 14:1153503. [PMID: 37266145 PMCID: PMC10229821 DOI: 10.3389/fphar.2023.1153503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023] Open
Abstract
Baicalein (5,6,7-trihydroxyflavone) is a traditional Chinese medicine with multiple pharmacological and biological activities including anti-inflammatory and anti-fibrotic effects. However, whether baicalein has a therapeutic impact on peritoneal fibrosis has not been reported yet. In the present study, network pharmacology and molecular docking approaches were performed to evaluate the role and the potential mechanisms of baicalein in attenuating peritoneal dialysis-associated peritoneal fibrosis. The results were validated in both animal models and the cultured human mesothelial cell line. Nine intersection genes among baicalein targets and the human peritoneum RNA-seq dataset including four encapsulating peritoneal sclerosis samples and four controls were predicted by network analysis. Among them, MMP2, BAX, ADORA3, HIF1A, PIM1, CA12, and ALOX5 exhibited higher expression in the peritoneum with encapsulating peritoneal sclerosis compared with those in the control, which might be crucial targets of baicalein against peritoneal fibrosis. Furthermore, KEGG and GO enrichment analyses suggested that baicalein played an anti-peritoneal fibrosis role through the regulating cell proliferation, inflammatory response, and AGE-RAGE signaling pathway. Moreover, molecular docking analysis revealed a strong potential binding between baicalein and MMP2, which was consistent with the predictive results. Importantly, using a mouse model of peritoneal fibrosis by intraperitoneally injecting 4.25% glucose dialysate, we found that baicalein treatment significantly attenuated peritoneal fibrosis, as evident by decreased collagen deposition, protein expression of α-SMA and fibronectin, and peritoneal thickness, at least, by reducing the expression of MMP2, suggesting that baicalein may have therapeutic potential in suppressing peritoneal dialysis-related fibrosis.
Collapse
Affiliation(s)
- Xiaohui Lu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology, Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, China
| | - Kefei Wu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology, Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, China
| | - Simin Jiang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology, Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, China
| | - Yi Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology, Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, China
| | - Yating Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology, Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, China
| | - Hongyu Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology, Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, China
| | - Guanglan Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology, Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, China
| | - Qinghua Liu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology, Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, China
| | - Yi Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology, Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology, Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology, Guangdong Provincial Key Laboratory of Nephrology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
48
|
Su HY, Yang JJ, Zou R, An N, Chen XC, Yang C, Yang HJ, Yao CW, Liu HF. Autophagy in peritoneal fibrosis. Front Physiol 2023; 14:1187207. [PMID: 37256065 PMCID: PMC10226653 DOI: 10.3389/fphys.2023.1187207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/03/2023] [Indexed: 06/01/2023] Open
Abstract
Peritoneal dialysis (PD) is a widely accepted renal replacement therapy for patients with end-stage renal disease (ESRD). Morphological and functional changes occur in the peritoneal membranes (PMs) of patients undergoing long-term PD. Peritoneal fibrosis (PF) is a common PD-related complication that ultimately leads to PM injury and peritoneal ultrafiltration failure. Autophagy is a cellular process of "self-eating" wherein damaged organelles, protein aggregates, and pathogenic microbes are degraded to maintain intracellular environment homeostasis and cell survival. Growing evidence shows that autophagy is involved in fibrosis progression, including renal fibrosis and hepatic fibrosis, in various organs. Multiple risk factors, including high-glucose peritoneal dialysis solution (HGPDS), stimulate the activation of autophagy, which participates in PF progression, in human peritoneal mesothelial cells (HPMCs). Nevertheless, the underlying roles and mechanisms of autophagy in PF progression remain unclear. In this review, we discuss the key roles and potential mechanisms of autophagy in PF to offer novel perspectives on future therapy strategies for PF and their limitations.
Collapse
|
49
|
Bora F, Aslan B, Sarı F, Yılmaz F, Ersoy FF, Köksoy S, Özdem S, Küçükçetin İÖ, Sipahioğlu M, Karakaya İ, Koç Y, Ulu MS. C allele in transforming growth factor-β1 rs1800471 gene polymorphisms might indicate a protective feature in encapsulating peritoneal sclerosis development. Ther Apher Dial 2023; 27:353-360. [PMID: 36054236 DOI: 10.1111/1744-9987.13913] [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: 01/18/2022] [Revised: 07/20/2022] [Accepted: 07/28/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Peritoneal fibrosis may progress in peritoneal dialysis (PD) patients to a fatal clinical condition called encapsulating peritoneal sclerosis (EPS). Transforming growth factor (TGF)-β plays a pivotal role in the pathogenesis of peritoneal fibrosis. We aimed to investigate the association among polymorphisms in the gene encoding TGF-β1, which were -509C/T (rs1800469), +869T/C (rs1982073), and +915G/C (rs1800471) in EPS patients. METHODS A total of 16 PD patients who were clinically and radiologically diagnosed with EPS were enrolled and 22 age- and gender-matched PD patients were selected as the non-EPS group. RESULTS G allele frequency at the rs1800471 gene polymorphism was significantly higher in the EPS group than non-EPS group (p = 0.005). Interestingly, the non-EPS group patients had CC or CG polymorphisms. CONCLUSION C allele in TGF-β1 rs1800471 gene polymorphisms might indicate a protective feature in EPS development. Knowing the presence of polymorphism may be effective in selecting renal replacement therapy in patients.
Collapse
Affiliation(s)
- Feyza Bora
- Department of Internal Medicine, Division of Nephrology, Akdeniz University Medical School, Antalya, Turkey
| | - Bengisu Aslan
- Department of Internal Medicine, Division of Nephrology, Akdeniz University Medical School, Antalya, Turkey
| | - Funda Sarı
- Department of Internal Medicine, Division of Nephrology, Akdeniz University Medical School, Antalya, Turkey
| | - Fatih Yılmaz
- Department of Nephrology, Antalya Atatürk State Hospital, Antalya, Turkey
| | - Fettah Fevzi Ersoy
- Department of Internal Medicine, Division of Nephrology, Akdeniz University Medical School, Antalya, Turkey
| | - Sadi Köksoy
- Department of Medical Microbiology, Akdeniz University Medical School, Antalya, Turkey
| | - Sebahat Özdem
- Department of Basic Medical Sciences, Medical Biochemistry, Akdeniz University Medical School, Antalya, Turkey
| | - İkbal Özen Küçükçetin
- Department of Basic Medical Sciences, Medical Biochemistry, Akdeniz University Medical School, Antalya, Turkey
| | - Murat Sipahioğlu
- Department of Internal Medicine, Division of Nephrology, Erciyes University Medical School, Kayseri, Turkey
| | - İbrahim Karakaya
- Department of Internal Medicine, Division of Nephrology, Çukurova University Medical School, Adana, Turkey
| | - Yener Koç
- Department of Internal Medicine, Division of Nephrology, Cumhuriyet University Medical School, Sivas, Turkey
| | - Memnune Sena Ulu
- Department of Internal Medicine, Division of Nephrology Bahçeşehir University Medical School, Istanbul, Turkey
| |
Collapse
|
50
|
Trionfetti F, Marchant V, González-Mateo GT, Kawka E, Márquez-Expósito L, Ortiz A, López-Cabrera M, Ruiz-Ortega M, Strippoli R. Novel Aspects of the Immune Response Involved in the Peritoneal Damage in Chronic Kidney Disease Patients under Dialysis. Int J Mol Sci 2023; 24:5763. [PMID: 36982834 PMCID: PMC10059714 DOI: 10.3390/ijms24065763] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
Chronic kidney disease (CKD) incidence is growing worldwide, with a significant percentage of CKD patients reaching end-stage renal disease (ESRD) and requiring kidney replacement therapies (KRT). Peritoneal dialysis (PD) is a convenient KRT presenting benefices as home therapy. In PD patients, the peritoneum is chronically exposed to PD fluids containing supraphysiologic concentrations of glucose or other osmotic agents, leading to the activation of cellular and molecular processes of damage, including inflammation and fibrosis. Importantly, peritonitis episodes enhance peritoneum inflammation status and accelerate peritoneal injury. Here, we review the role of immune cells in the damage of the peritoneal membrane (PM) by repeated exposure to PD fluids during KRT as well as by bacterial or viral infections. We also discuss the anti-inflammatory properties of current clinical treatments of CKD patients in KRT and their potential effect on preserving PM integrity. Finally, given the current importance of coronavirus disease 2019 (COVID-19) disease, we also analyze here the implications of this disease in CKD and KRT.
Collapse
Affiliation(s)
- Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149 Rome, Italy
| | - Vanessa Marchant
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
- REDINREN/RICORS2040, 28029 Madrid, Spain
| | - Guadalupe T. González-Mateo
- Cell-Cell Communication & Inflammation Unit, Centre for Molecular Biology “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain
- Premium Research, S.L., 19005 Guadalajara, Spain
| | - Edyta Kawka
- Department of Pathophysiology, Poznan University of Medical Sciences, 10 Fredry St., 61-701 Poznan, Poland
| | - Laura Márquez-Expósito
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
- REDINREN/RICORS2040, 28029 Madrid, Spain
| | - Alberto Ortiz
- IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Manuel López-Cabrera
- Cell-Cell Communication & Inflammation Unit, Centre for Molecular Biology “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
- REDINREN/RICORS2040, 28029 Madrid, Spain
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149 Rome, Italy
| |
Collapse
|