Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 350
Filter
1.
Ren Fail ; 46(1): 2350235, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38721924

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 TGFß1 and α-SMA. Adding CAPE to PDF significantly inhibited PD-induced submesothelial thickening, reduced TGFß1 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.


Subject(s)
AMP-Activated Protein Kinases , Caffeic Acids , Peritoneal Dialysis , Peritoneal Fibrosis , Phenylethyl Alcohol , Rats, Sprague-Dawley , Sirtuin 1 , Animals , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/prevention & control , Sirtuin 1/metabolism , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Caffeic Acids/pharmacology , Caffeic Acids/therapeutic use , Rats , Male , AMP-Activated Protein Kinases/metabolism , Peritoneal Dialysis/adverse effects , Mitochondria/drug effects , Mitochondria/metabolism , Disease Models, Animal , Signal Transduction/drug effects , Peritoneum/pathology , Peritoneum/drug effects , Peritoneum/metabolism , Homeostasis/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/metabolism , Membrane Potential, Mitochondrial/drug effects , Dialysis Solutions
2.
Exp Clin Transplant ; 22(3): 239-241, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38695593

ABSTRACT

Encapsulating peritoneal sclerosis is a rare but highly morbid disease process in patients with end-stage kidney disease on peritoneal dialysis. Surgical management has been described in patients with encapsulation of bowel causing obstruction. Here, we describe a case of surgical management in a patient following kidney transplant with medically refractory ascites and lower extremity edema.


Subject(s)
Kidney Failure, Chronic , Kidney Transplantation , Peritoneal Fibrosis , Humans , Kidney Transplantation/adverse effects , Peritoneal Fibrosis/surgery , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/diagnosis , Peritoneal Fibrosis/diagnostic imaging , Kidney Failure, Chronic/surgery , Kidney Failure, Chronic/diagnosis , Treatment Outcome , Ascites/etiology , Ascites/surgery , Ascites/diagnosis , Edema/etiology , Edema/surgery , Male , Peritoneal Dialysis/adverse effects , Female , Middle Aged , Adult
3.
Front Endocrinol (Lausanne) ; 15: 1282925, 2024.
Article in English | MEDLINE | ID: mdl-38567303

ABSTRACT

Background: Encapsulating peritoneal sclerosis (EPS) is a rare complication of prolonged peritoneal dialysis (PD) exposure, characterised by peritoneal thickening, calcification, and fibrosis ultimately presenting with life-threatening bowel obstruction. The presence or role of peritoneal calcification in the pathogenesis of EPS is poorly characterised. We hypothesise that significantly aberrant bone mineral metabolism in patients on PD can cause peritoneal calcification which may trigger the development of EPS. We compared the temporal evolution of bone mineral markers during PD in EPS patients with non-EPS long-term PD controls. Methods: Linear mixed model and logistic regression analysis were used to compare four-monthly serum levels of calcium, phosphate, parathyroid hormone, and alkaline phosphatase (ALP) over the duration of PD exposure in 46 EPS and 46 controls (PD, non-EPS) patients. Results: EPS patients had higher mean calcium (2.51 vs. 2.41 mmol/L) and ALP (248.00 vs. 111.13 IU/L) levels compared with controls (p=0.01 and p<0.001 respectively, maximum likelihood estimation). Logistic regression analysis demonstrated that high serum calcium and phosphate levels during PD were associated with a 4.5 and 2.9 fold increase in the risk of developing EPS respectively. Conclusion: High levels of calcium and phosphate in patients on PD were identified to be risk factors for EPS development. Possible reasons for this may be an imbalance of pro-calcifying factors and calcification inhibitors promoting peritoneal calcification which increases peritoneal stiffness. Mechanical alterations may trigger, unregulated fibrosis and subsequent development of EPS. Improved management of secondary hyperparathyroidism during PD may ultimately diminish the EPS risk.


Subject(s)
Calcinosis , Hyperparathyroidism , Peritoneal Fibrosis , Humans , Peritoneal Fibrosis/etiology , Calcium , Risk Factors , Calcinosis/etiology , Minerals , Phosphates
4.
Sci Rep ; 14(1): 7412, 2024 03 28.
Article in English | MEDLINE | ID: mdl-38548914

ABSTRACT

Peritoneal membrane dysfunction in peritoneal dialysis (PD) is primarily attributed to angiogenesis; however, the integrity of vascular endothelial cells can affect peritoneal permeability. Hyaluronan, a component of the endothelial glycocalyx, is reportedly involved in preventing proteinuria in the normal glomerulus. One hypothesis suggests that development of encapsulating peritoneal sclerosis (EPS) is triggered by protein leakage due to vascular endothelial injury. We therefore investigated the effect of hyaluronan in the glycocalyx on peritoneal permeability and disease conditions. After hyaluronidase-mediated degradation of hyaluronan on the endothelial cells of mice, macromolecules, including albumin and ß2 microglobulin, leaked into the dialysate. However, peritoneal transport of small solute molecules was not affected. Pathologically, hyaluronan expression was diminished; however, expression of vascular endothelial cadherin and heparan sulfate, a core protein of the glycocalyx, was preserved. Hyaluronan expression on endothelial cells was studied using 254 human peritoneal membrane samples. Hyaluronan expression decreased in patients undergoing long-term PD treatment and EPS patients treated with conventional solutions. Furthermore, the extent of hyaluronan loss correlated with the severity of vasculopathy. Hyaluronan on endothelial cells is involved in the peritoneal transport of macromolecules. Treatment strategies that preserve hyaluronan in the glycocalyx could prevent the leakage of macromolecules and subsequent related complications.


Subject(s)
Peritoneal Dialysis , Peritoneal Fibrosis , Humans , Animals , Mice , Hyaluronic Acid/metabolism , Endothelial Cells , Peritoneal Dialysis/adverse effects , Peritoneum/metabolism , Biological Transport , Dialysis Solutions/metabolism , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/metabolism
6.
Sci China Life Sci ; 67(2): 360-378, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37815699

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.


Subject(s)
Melatonin , Peritoneal Fibrosis , Humans , Animals , Mice , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/prevention & control , Peritoneal Fibrosis/pathology , Melatonin/pharmacology , Melatonin/therapeutic use , Vascular Endothelial Growth Factor A , Pyroptosis , Ultrafiltration , Epithelial Cells , Glucose/pharmacology , Fibrosis
7.
J Formos Med Assoc ; 123(1): 98-105, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37365098

ABSTRACT

BACKGROUND/PURPOSE: Encapsulating peritoneal sclerosis (EPS) is a rare and potential lethal complication of peritoneal dialysis characterized by bowel obstruction. Surgical enterolysis is the only curative therapy. Currently, there are no tools for predicting postsurgical prognosis. This study aimed to identify a computed tomography (CT) scoring system that could predict mortality after surgery in patients with severe EPS. METHODS: This retrospective study enrolled patients with severe EPS who underwent surgical enterolysis in a tertiary referral medical center. The association of CT score with surgical outcomes including mortality, blood loss, and bowel perforation was analyzed. RESULTS: Thirty-four patients who underwent 37 procedures were recruited and divided into a survivor and non-survivor group. The survivor group had higher body mass indices (BMIs, 18.1 vs. 16.7 kg/m2, p = 0.035) and lower CT scores (11 vs. 17, p < 0.001) than the non-survivor group. The receiver operating characteristic curve revealed that a CT score of ≥15 could be considered a cutoff point to predict surgical mortality, with an area under the curve of 0.93, sensitivity of 88.9%, and specificity of 82.1%. Compared with the group with CT scores of <15, the group with CT scores of ≥15 had a lower BMI (19.7 vs. 16.2 kg/m2, p = 0.004), higher mortality (4.2% vs. 61.5%, p < 0.001), greater blood loss (50 vs. 400 mL, p = 0.007), and higher incidence of bowel perforation (12.5% vs. 61.5%, p = 0.006). CONCLUSION: The CT scoring system could be useful in predicting surgical risk in patients with severe EPS receiving enterolysis.


Subject(s)
Intestinal Perforation , Peritoneal Fibrosis , Humans , Peritoneal Fibrosis/diagnostic imaging , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/surgery , Retrospective Studies , Intestinal Perforation/diagnostic imaging , Intestinal Perforation/etiology , Intestinal Perforation/surgery , Tomography, X-Ray Computed , Treatment Outcome , Sclerosis/complications
9.
Intern Med ; 63(5): 659-663, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37468244

ABSTRACT

We herein report a case of encapsulating peritoneal sclerosis (EPS) in a patient without chronic kidney disease after gastrectomy. A 69-year-old man underwent distal gastrectomy for early gastric cancer at 25 years old. After 43 years, he developed bowel obstruction and underwent enterolysis of the encapsulated small intestine. A pathological examination of the capsular membranes revealed inflammation, foam, and giant cells that destroyed foreign substances. The patient was discharged 1.5 months later. Foreign body reactions to surgical instruments used in gastrectomy are considered a cause of EPS. EPS due to foreign body reactions to surgical instruments should also be considered in such cases.


Subject(s)
Foreign Bodies , Peritoneal Fibrosis , Stomach Neoplasms , Aged , Humans , Male , Foreign Bodies/complications , Gastrectomy/adverse effects , Peritoneal Fibrosis/diagnostic imaging , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/surgery , Peritoneum , Sclerosis , Stomach Neoplasms/pathology
10.
Perit Dial Int ; 44(2): 141-144, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37905306

ABSTRACT

Encapsulating peritoneal sclerosis (EPS) is a rare and potentially fatal complication of long-term peritoneal dialysis (PD). EPS-induced large volume and recurrent ascites represents a challenging condition. We report a 51-year-old man with kidney failure treated with PD for 13 years who eventually developed early stage of EPS accompanied with poor intake and recurrent ascites. After management including discontinuing PD and switching to haemodialysis, as well as oral steroids and tamoxifen administration, the patient had refractory ascites. An intervention of weekly intraperitoneal steroid infusion with methylprednisolone was implemented for a year. Gradually, we observed a reduction in ascites drainage, an improvement of clinical symptoms and the patient's nutritional status. The PD catheter was successfully removed as there was no recurrence of ascites. Intraperitoneal corticosteroid administration represents a new intervention for patients with early stage of EPS and recurrent ascites after PD cessation.


Subject(s)
Peritoneal Dialysis , Peritoneal Fibrosis , Male , Humans , Middle Aged , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/diagnosis , Peritoneal Fibrosis/drug therapy , Peritoneal Fibrosis/etiology , Ascites/drug therapy , Ascites/etiology , Renal Dialysis/adverse effects , Steroids , Sclerosis/complications
11.
Int Immunopharmacol ; 126: 111300, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38016346

ABSTRACT

Encapsulating peritoneal sclerosis (EPS) is a severe complication of peritoneal dialysis (PD). Surgery is a therapeutic strategy for the treatment of complete intestinal obstruction. However, complete intestinal obstruction in long-term PD results in high mortality and morbidity rates after surgery. Immunopathogenesis participates in EPS formation: CD8, Th1, and Th17 cell numbers increased during the formation of EPS. The anti-inflammatory and immunomodulatory effects of melatonin may have beneficial effects on this EPS. In the present study, we determined that melatonin treatment significantly decreases the Th1 and Th17 cell populations in mice with EPS, decreases the production of IL-1ß, TNF-α, IL-6, and IFN-γ, and increases the production of IL-10. The suppression of Th1 and Th17 cell differentiation by melatonin occurs through the inhibition of dendritic cell (DC) activation by affecting the initiation of the NF-κB signaling pathway in DCs. Our study suggests that melatonin has preventive potential against the formation of EPS in patients with PD.


Subject(s)
Intestinal Obstruction , Melatonin , Peritoneal Fibrosis , Humans , Animals , Mice , Peritoneal Fibrosis/etiology , NF-kappa B/metabolism , Melatonin/pharmacology , Melatonin/therapeutic use , Cell Differentiation , Signal Transduction , Dendritic Cells/metabolism , Intestinal Obstruction/complications , Intestinal Obstruction/pathology
12.
Int Urol Nephrol ; 56(6): 1987-1999, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38097887

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-ß/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-ß/Smad pathway, which laid the experimental foundation for nintedanib in the treatment of PF.


Subject(s)
Enhancer of Zeste Homolog 2 Protein , Epithelial-Mesenchymal Transition , Indoles , Kruppel-Like Transcription Factors , Peritoneal Fibrosis , Peritoneal Fibrosis/prevention & control , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/etiology , Animals , Mice , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Epithelial-Mesenchymal Transition/drug effects , Indoles/pharmacology , Enhancer of Zeste Homolog 2 Protein/metabolism , Cells, Cultured , Mice, Inbred C57BL , Disease Models, Animal , Humans , Male
13.
Stem Cell Res Ther ; 14(1): 293, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37817212

ABSTRACT

Peritoneal fibrosis (PF) is a pathophysiological condition caused by a variety of pathogenic factors. The most important features of PF are mesothelial-mesenchymal transition and accumulation of activated (myo-)fibroblasts, which hinder effective treatment; thus, it is critical to identify other practical approaches. Recently, stem cell (SC) therapy has been indicated to be a potential strategy for this disease. Increasing evidence suggests that many kinds of SCs alleviate PF mainly by differentiating into mesothelial cells; secreting cytokines and extracellular vesicles; or modulating immune cells, particularly macrophages. However, there are relatively few articles summarizing research in this direction. In this review, we summarize the risk factors for PF and discuss the therapeutic roles of SCs from different sources. In addition, we outline effective approaches and potential mechanisms of SC therapy for PF. We hope that our review of articles in this area will provide further inspiration for research on the use of SCs in PF treatment.


Subject(s)
Peritoneal Fibrosis , Humans , Peritoneal Fibrosis/etiology , Epithelial-Mesenchymal Transition/physiology , Epithelium , Fibroblasts/pathology , Cell- and Tissue-Based Therapy/adverse effects , Peritoneum/pathology , Fibrosis
14.
Sci Rep ; 13(1): 16340, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37770630

ABSTRACT

Peritoneal calcification is a prominent feature of the later stage of encapsulating peritoneal sclerosis (EPS) in patients undergoing long-term peritoneal dialysis (PD). However, the pathogenesis and preventive strategy for peritoneal calcification remain unclear. Peritoneum samples from EPS patients were examined histologically. Peritoneal calcification was induced in mice by feeding with an adenine-containing diet combined with intraperitoneal administration of lipopolysaccharide and a calcifying solution containing high calcium and phosphate. Excised mouse peritoneum, human mesothelial cells (MeT5A), and mouse embryonic fibroblasts (MEFs) were cultured in calcifying medium. Immunohistochemistry confirmed the appearance of osteoblastic differentiation-marker-positive cells in the visceral peritoneum from EPS patients. Intraperitoneal administration of magnesium suppressed peritoneal fibrosis and calcification in mice. Calcifying medium increased the calcification of cultured mouse peritoneum, which was prevented by magnesium. Calcification of the extracellular matrix was accelerated in Met5A cells and MEFs treated with calcification medium. Calcifying medium also upregulated osteoblastic differentiation markers in MeT5A cells and induced apoptosis in MEFs. Conversely, magnesium supplementation mitigated extracellular matrix calcification and phenotypic transdifferentiation and apoptosis caused by calcifying conditions in cultured MeT5A cells and MEFs. Phosphate loading contributes to the progression of EPS through peritoneal calcification and fibrosis, which can be prevented by magnesium supplementation.


Subject(s)
Calcinosis , Peritoneal Dialysis , Peritoneal Fibrosis , Humans , Animals , Mice , Peritoneum/pathology , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/prevention & control , Peritoneal Fibrosis/pathology , Magnesium/pharmacology , Fibroblasts/pathology , Peritoneal Dialysis/adverse effects , Calcinosis/pathology
16.
Intern Med ; 62(21): 3203-3207, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37438140

ABSTRACT

Encapsulating peritoneal sclerosis (EPS) is a fatal complication of peritoneal dialysis. A 68-year-old man undergoing peritoneal dialysis for 10 years started receiving daily 50 mg of glucocorticoids for idiopathic pulmonary sclerosis. At the transition to hemodialysis, a peritoneal biopsy was performed, which demonstrated mild histological changes, including no fibrin formation and mild T lymphocyte infiltration at the time of 6.5 mg glucocorticoids. However, five months later, he developed EPS when receiving 2.5 mg glucocorticoids. Afterward, over 5 mg daily glucocorticoids were required to avoid the recurrence of EPS. These findings suggest that glucocorticoids may conceal peritoneal inflammation, a main contributor to EPS.


Subject(s)
Peritoneal Dialysis , Peritoneal Fibrosis , Male , Humans , Aged , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/pathology , Glucocorticoids/adverse effects , Sclerosis , Peritoneal Dialysis/adverse effects , Peritoneum
18.
Clin Exp Nephrol ; 27(9): 717-727, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37278945

ABSTRACT

Encapsulating peritoneal sclerosis (EPS), a condition with a high mortality rate, is a serious complication of peritoneal dialysis (PD). In Japan, EPS became a central issue in the clinical setting during the mid-90s and the beginning of this century. However, following the introduction of biocompatible neutral PD solutions containing lower levels of glucose degradation products, the incidence and clinical severity of EPS has been greatly lessened. During the past three decades, the etiology of EPS has been elucidated by findings obtained by peritoneal biopsy, laparoscopy, and surgical intervention. Accumulating findings suggest the need for a paradigm change on the nature of EPS pathophysiology; notably, EPS appears not to reflect peritoneal sclerosis per se, but rather the formation of a neo-membrane as a biological reaction to peritoneal injury. This narrative review looks back on the history of EPS in Japan, and discusses EPS pathophysiology, the impact of neutral PD solution on peritoneal protection, and a future novel diagnostic approach, ultra-fine endoscope, for the identification of patients at high risk of EPS.


Subject(s)
Peritoneal Dialysis , Peritoneal Fibrosis , Humans , Peritoneal Fibrosis/diagnosis , Peritoneal Fibrosis/etiology , Japan/epidemiology , Peritoneal Dialysis/adverse effects , Peritoneum/pathology , Dialysis Solutions/adverse effects , Sclerosis/complications , Sclerosis/pathology
19.
Chem Biol Interact ; 382: 110589, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37268199

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-ß1)-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-ß1-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-ß1/Smad and JAK/STAT signaling pathway. Therefore, because of these antifibrotic effects, NMPDOva may be a promising therapeutic agent for PD-related PF.


Subject(s)
Peritoneal Fibrosis , Mice , Animals , Peritoneal Fibrosis/drug therapy , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/pathology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta/metabolism , Signal Transduction , Peritoneum/metabolism , Peritoneum/pathology , Epithelial-Mesenchymal Transition , Fibrosis
20.
Naunyn Schmiedebergs Arch Pharmacol ; 396(10): 2379-2391, 2023 10.
Article in English | MEDLINE | ID: mdl-37052642

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-ß1 (TGF-ß1), 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-ß1, collagen I, fibronectin) and restored the expression of E-cadherin, BMP-7 in PF mice and TGF-ß1-treated Met-5A cells. Furthermore, SM markedly down-regulated the expression of TGF-ß1, 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-ß/Smad signaling.


Subject(s)
Peritoneal Fibrosis , Silymarin , Humans , Mice , Animals , Peritoneal Fibrosis/drug therapy , Peritoneal Fibrosis/prevention & control , Peritoneal Fibrosis/etiology , Transforming Growth Factor beta1/metabolism , Silymarin/pharmacology , Silymarin/therapeutic use , Quality of Life , Signal Transduction , Fibrosis , Smad Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...