Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Lab Invest ; 103(4): 100050, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36870292

RESUMEN

Long-term peritoneal dialysis (PD) is often associated with peritoneal dysfunction leading to withdrawal from PD. The characteristic pathologic features of peritoneal dysfunction are widely attributed to peritoneal fibrosis and angiogenesis. The detailed mechanisms remain unclear, and treatment targets in clinical settings have yet to be identified. We investigated transglutaminase 2 (TG2) as a possible novel therapeutic target for peritoneal injury. TG2 and fibrosis, inflammation, and angiogenesis were investigated in a chlorhexidine gluconate (CG)-induced model of peritoneal inflammation and fibrosis, representing a noninfectious model of PD-related peritonitis. Transforming growth factor (TGF)-ß type I receptor (TGFßR-I) inhibitor and TG2-knockout mice were used for TGF-ß and TG2 inhibition studies, respectively. Double immunostaining was performed to identify cells expressing TG2 and endothelial-mesenchymal transition (EndMT). In the rat CG model of peritoneal fibrosis, in situ TG2 activity and protein expression increased during the development of peritoneal fibrosis, as well as increases in peritoneal thickness and numbers of blood vessels and macrophages. TGFßR-I inhibitor suppressed TG2 activity and protein expression, as well as peritoneal fibrosis and angiogenesis. TGF-ß1 expression, peritoneal fibrosis, and angiogenesis were suppressed in TG2-knockout mice. TG2 activity was detected by α-smooth muscle actin-positive myofibroblasts, CD31-positive endothelial cells, and ED-1-positive macrophages. CD31-positive endothelial cells in the CG model were α-smooth muscle actin-positive, vimentin-positive, and vascular endothelial-cadherin-negative, suggesting EndMT. In the CG model, EndMT was suppressed in TG2-knockout mice. TG2 was involved in the interactive regulation of TGF-ß. As inhibition of TG2 reduced peritoneal fibrosis, angiogenesis, and inflammation associated with TGF-ß and vascular endothelial growth factor-A suppression, TG2 may provide a new therapeutic target for ameliorating peritoneal injuries in PD.


Asunto(s)
Fibrosis Peritoneal , Ratones , Ratas , Animales , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/prevención & control , Fibrosis Peritoneal/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Actinas/metabolismo , Clorhexidina/efectos adversos , Clorhexidina/metabolismo , Células Endoteliales/metabolismo , Peritoneo/patología , Factor de Crecimiento Transformador beta1/metabolismo , Fibrosis , Inflamación/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Ratones Noqueados
2.
Int J Mol Sci ; 24(22)2023 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-38003303

RESUMEN

In our preliminary experiment, peritoneal sclerosis likely induced by peritoneal dialysis was unexpectedly observed in the livers of rats given bleomycin and lansoprazole. We examined whether this peritoneal thickening around the liver was time-dependently induced by administration of both drugs. Male Wistar rats were injected with bleomycin and/or lansoprazole for 2 or 4 weeks. The 3YB-1 cell line derived from rat fibroblasts was treated by bleomycin and/or lansoprazole for 24 h. The administration of both drugs together, but not individually, thickened the peritoneal tissue around the liver. There was accumulation of collagen fibers, macrophages, and eosinophils under mesothelial cells. Expressions of Col1a1, Mcp1 and Mcp3 genes were increased in the peritoneal tissue around the liver and in 3YB-1 cells by the administration of both drugs together, and Opn genes had increased expressions in this tissue and 3YB-1 cells. Mesothelial cells indicated immunoreactivity against both cytokeratin, a mesothelial cell marker, and αSMA, a fibroblast marker, around the livers of rats given both drugs. Administration of both drugs induced the migration of macrophages and eosinophils and induced fibrosis associated with the possible activation of fibroblasts and the possible promotion of the mesothelial-mesenchymal transition. This might become a novel model of peritoneal sclerosis for peritoneal dialysis.


Asunto(s)
Fibrosis Peritoneal , Ratas , Masculino , Animales , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/genética , Bleomicina/efectos adversos , Ratas Wistar , Lansoprazol/efectos adversos , Lansoprazol/metabolismo , Células Epiteliales/metabolismo , Peritoneo/patología
3.
Mol Cell Biochem ; 477(7): 1959-1971, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35380292

RESUMEN

OBJECTIVE: Peritoneal fibrosis (PF) is commonly induced by bioincompatible dialysate exposure during peritoneal dialysis, but the underlying mechanisms remain elusive. This study aimed to investigate the roles of peroxisome proliferator-activated receptor gamma (PPARγ) in PF pathogenesis. METHODS: Rat and cellular PF models were established by high glucose dialysate and lipopolysaccharide treatments. Serum creatinine, urea nitrogen, and glucose contents were detected by ELISA. Histological evaluation was done through H&E and Masson staining. GLUT1, PPARγ, and other protein expression were measured by qRT-PCR, western blotting, and IHC. PPARγ and GLUT1 subcellular distribution were detected using confocal microscopy. Cell proliferation was assessed by MTT and Edu staining. RESULTS: Serum creatinine, urea nitrogen and glucose, and PPARγ and GLUT1 expression in rat PF model were reduced by PPARγ agonists Rosiglitazone or 15d-PGJ2 and elevated by antagonist GW9662. Rosiglitazone or 15d-PGJ2 repressed and GW9662 aggravated peritoneal fibrosis in rat PF model. PPARγ and GLUT1 were mainly localized in nucleus and cytosols of peritoneal mesothelial cells, respectively, which were reduced in cellular PF model, enhanced by Rosiglitazone or 15d-PGJ2, and repressed by GW9662. TGF-ß and a-SMA expression was elevated in cellular PF model, which was inhibited by Rosiglitazone or 15d-PGJ2 and promoted by GW9662. PPARγ silencing reduced GLUT1, elevated a-SMA and TGF-b expression, and promoted peritoneal mesothelial cell proliferation, which were oppositely changed by PPARγ overexpression. CONCLUSION: PPARγ inhibited high glucose-induced peritoneal fibrosis progression through elevating GLUT1 expression and repressing peritoneal mesothelial cell proliferation.


Asunto(s)
Transportador de Glucosa de Tipo 1 , PPAR gamma , Fibrosis Peritoneal , Tiazolidinedionas , Animales , Proliferación Celular , Creatinina , Soluciones para Diálisis/farmacología , Glucosa/farmacología , Transportador de Glucosa de Tipo 1/metabolismo , Nitrógeno/metabolismo , Nitrógeno/farmacología , PPAR gamma/agonistas , PPAR gamma/genética , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/genética , Prostaglandina D2 , Ratas , Rosiglitazona/farmacología , Tiazolidinedionas/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Urea
4.
Med Mol Morphol ; 55(1): 27-40, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34622315

RESUMEN

Peritoneal fibrosis is a serious complication of long-term peritoneal dialysis, attributable to inflammation and mitochondrial dysfunction. Mitochonic acid-5 (MA-5), an indole-3-acetic acid derivative, improves mitochondrial dysfunction and has therapeutic potential against various diseases including kidney diseases. However, whether MA-5 is effective against peritoneal fibrosis remains unclear. Therefore, we investigated the effect of MA-5 using a peritoneal fibrosis mouse model. Peritoneal fibrosis was induced in C57BL/6 mice via intraperitoneal injection of chlorhexidine gluconate (CG) every other day for 3 weeks. MA-5 was administered daily by oral gavage. The mice were divided into control, MA-5, CG, and CG + MA-5 groups. Following treatment, immunohistochemical analyses were performed. Fibrotic thickening of the parietal peritoneum induced by CG was substantially attenuated by MA-5. The number of α-smooth muscle actin-positive myofibroblasts, transforming growth factor ß-positive cells, F4/80-positive macrophages, monocyte chemotactic protein 1-positive cells, and 4-hydroxy-2-nonenal-positive cells was considerably decreased. In addition, reduced ATP5a1-positive and uncoupling protein 2-positive cells in the CG group were notably increased by MA-5. MA-5 may ameliorate peritoneal fibrosis by suppressing macrophage infiltration and oxidative stress, thus restoring mitochondrial function. Overall, MA-5 has therapeutic potential against peritoneal fibrosis.


Asunto(s)
Fibrosis Peritoneal , Animales , Clorhexidina/análogos & derivados , Modelos Animales de Enfermedad , Ácidos Indolacéticos , Ratones , Ratones Endogámicos C57BL , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/tratamiento farmacológico , Fibrosis Peritoneal/prevención & control , Peritoneo/metabolismo , Peritoneo/patología , Fenilbutiratos/química
5.
Am J Physiol Renal Physiol ; 320(3): F273-F284, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33427062

RESUMEN

Peritoneal dialysis (PD)-related peritoneal fibrosis (PF) is characterized by progressive extracellular matrix (ECM) accumulation in peritoneal mesothelial cells (PMCs) during long-term use of high glucose (HG)-based dialysates. Activation of the renin-angiotensin system (RAS) has been shown to be associated with PF. The aim of this study was to explore the underlying mechanism of the RAS in HG-induced PF. We treated C57BL/6 mice and a human PMC line with HG to induce a PF model and to stimulate ECM accumulation, respectively. RAS activity was blocked using valsartan or angiotensin II (ANGII) type 1 receptor siRNA. The major findings were as follows. First, mice in the HG group exhibited increased collagen deposition and expression of ECM proteins, including α-smooth muscle actin (α-SMA) and collagen type I in the peritoneum. Consistent with the in vivo data, HG upregulated α-SMA expression in human peritoneal mesothelial cells (HPMCs) in a time- and dose-dependent manner. Second, HG stimulation led to RAS activation in HPMCs, and inactivation of RAS decreased the expression of ECM proteins in vivo and in vitro, even during HG stimulation. Finally, RAS-mediated ECM production was associated with lipid accumulation in HPMCs and depended on the dysregulation of the low-density lipoprotein receptor (LDLr) pathway. HG-stimulated HPMCs showed increased coexpression of LDLr and α-SMA, whereas blockade of RAS activity reversed the effect. Furthermore, inhibition of LDLr signaling decreased α-SMA and collagen type I expression in HPMCs when treated with HG and ANG II. In conclusion, increased intracellular RAS activity impaired lipid homeostasis and induced ECM accumulation in HPMCs by disrupting the LDLr pathway, which contributed to PF.


Asunto(s)
Matriz Extracelular/metabolismo , Fibrosis Peritoneal/metabolismo , Peritoneo/metabolismo , Receptores de LDL/metabolismo , Sistema Renina-Angiotensina , Actinas/metabolismo , Animales , Línea Celular , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Matriz Extracelular/patología , Glucosa , Humanos , Masculino , Ratones Endogámicos C57BL , Oxidación-Reducción , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/genética , Fibrosis Peritoneal/patología , Peritoneo/patología , Receptores de LDL/genética , Sistema Renina-Angiotensina/genética , Transducción de Señal
6.
J Pathol ; 250(1): 55-66, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31579932

RESUMEN

Peritoneal fibrosis remains a problem in kidney failure patients treated with peritoneal dialysis. Severe peritoneal fibrosis with encapsulation or encapsulating peritoneal sclerosis is devastating and life-threatening. Although submesothelial fibroblasts as the major precursor of scar-producing myofibroblasts in animal models and M2 macrophage (Mϕ)-derived chemokines in peritoneal effluents of patients before diagnosis of encapsulating peritoneal sclerosis have been identified, attenuation of peritoneal fibrosis is an unmet medical need partly because the mechanism for cross talk between Mϕs and fibroblasts remains unclear. We use a sodium hypochlorite-induced mouse model akin to clinical encapsulated peritoneal sclerosis to study how the peritoneal Mϕs activate fibroblasts and fibrosis. Sodium hypochlorite induces the disappearance of CD11bhigh F4/80high resident Mϕs but accumulation of CD11bint F4/80int inflammatory Mϕs (InfMϕs) through recruiting blood monocytes and activating local cell proliferation. InfMϕs switch to express chemokine (C-C motif) ligand 17 (CCL17), CCL22, and arginase-1 from day 2 after hypochlorite injury. More than 75% of InfMϕs undergo genetic recombination by Csf1r-driven Cre recombinase, providing the possibility to reduce myofibroblasts and fibrosis by diphtheria toxin-induced Mϕ ablation from day 2 after injury. Furthermore, administration of antibody against CCL17 can reduce Mϕs, myofibroblasts, fibrosis, and improve peritoneal function after injury. Mechanistically, CCL17 stimulates migration and collagen production of submesothelial fibroblasts in culture. By breeding mice that are induced to express red fluorescent protein in Mϕs and green fluorescence protein (GFP) in Col1a1-expressing cells, we confirmed that Mϕs do not produce collagen in peritoneum before and after injury. However, small numbers of fibrocytes are found in fibrotic peritoneum of chimeric mice with bone marrow from Col1a1-GFP reporter mice, but they do not contribute to myofibroblasts. These data demonstrate that InfMϕs switch to pro-fibrotic phenotype and activate peritoneal fibroblasts through CCL17 after injury. CCL17 blockade in patients with peritoneal fibrosis may provide a novel therapy. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Quimiocina CCL17/metabolismo , Fibroblastos/metabolismo , Mediadores de Inflamación/metabolismo , Activación de Macrófagos , Macrófagos Peritoneales/metabolismo , Comunicación Paracrina , Fibrosis Peritoneal/metabolismo , Peritoneo/metabolismo , Animales , Proliferación Celular , Quimiocina CCL17/genética , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Modelos Animales de Enfermedad , Fibroblastos/patología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Macrófagos Peritoneales/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/genética , Fibrosis Peritoneal/patología , Peritoneo/patología , Fenotipo , Regiones Promotoras Genéticas , Transducción de Señal , Hipoclorito de Sodio
7.
Clin Exp Nephrol ; 25(9): 935-943, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33909175

RESUMEN

BACKGROUND: Peritoneal dialysis (PD) is essential for patients with end-stage renal disease. Peritoneal fibrosis (PF) is a complex inflammatory, fibrogenic process. No effective treatments are available to prevent these processes. Hepatocyte growth factor (HGF) possesses anti-inflammatory and anti-fibrotic properties. The aim of this study was to analyze whether HGF suppresses MGO-induced peritoneal inflammation and fibrosis in a mouse model. METHODS: PF was induced by intraperitoneal (IP) injections of MGO for 14 days. C57/BL/6 mice were divided into three groups: Sham group (only vehicle); Sham + MGO group (PF induced by MGO); and HGF + MGO group (PF mice treated with recombinant human-HGF). PF was assessed from tissue samples by Masson's trichrome staining. Inflammation and fibrosis-associated factors were assessed by immunohistochemistry and quantitative real-time PCR. RESULTS: MGO-injected mice showed significant thickening of the submesothelial compact zone with PF. Treatment with HGF significantly reduced PM thickness and suppressed the expression of collagen I and III and α-SMA. Expression of profibrotic and proinflammatory cytokines (TGF-ß, TNF-α, IL-1ß) was reduced by HGF treatment. The number of macrophages, and M1 and M2 macrophage-related markers, such as CD86, CD206, and CD163, was reduced in HGF + MGO mice. CONCLUSION: HGF attenuates MGO-induced PF in mice. Furthermore, HGF treatment reduces myofibroblast and macrophage infiltration, and attenuates the upregulated expression of proinflammatory and profibrotic genes in peritoneal tissues. HGF might be an effective approach to prevent the development of PF in patients undergoing PD.


Asunto(s)
Factor de Crecimiento de Hepatocito/uso terapéutico , Fibrosis Peritoneal/tratamiento farmacológico , Fibrosis Peritoneal/metabolismo , Peritonitis/tratamiento farmacológico , Peritonitis/metabolismo , Actinas/metabolismo , Animales , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Interleucina-1beta/genética , Macrófagos , Masculino , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Miofibroblastos , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/patología , Peritonitis/inducido químicamente , Peritonitis/patología , Piruvaldehído , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Factor de Crecimiento Transformador beta/genética , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba/efectos de los fármacos
8.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33947038

RESUMEN

We investigated the effectiveness of the transforming growth factor beta-1 (TGF-ß) receptor inhibitor GW788388 on the epithelial to mesenchymal transition (EMT) using human peritoneal mesothelial cells (HPMCs) and examined the effectiveness of GW788388 on the peritoneal membrane using a peritoneal fibrosis mouse model. HPMCs were treated with TGF-ß with or without GW788388. Animal experiments were conducted on male C57/BL6 mice. Peritoneal fibrosis was induced by intraperitoneal injection of chlorhexidine gluconate. GW788388 was administered by once-daily oral gavage. The morphological change, cell migration, and invasion resulted from TGF-ß treatment, but these changes were attenuated by cotreatment with GW788388. TGF-ß-treated HPMCs decreased the level of the epithelial cell marker and increased the levels of the mesenchymal cell markers. Cotreatment with GW788388 reversed these changes. Phosphorylated Smad2 and Smad3 protein levels were stimulated with TGF-ß and the change was attenuated by cotreatment with GW788388. For the peritoneal fibrosis mice, thickness and collagen deposition of parietal peritoneum was increased, but this change was attenuated by cotreatment with GW788388. GW788388, an orally available potent TGF-ß receptor type 1 inhibitor, effectively attenuated TGF-ß-induced EMT in HPMCs. Cotreatment with GW788388 improved peritoneal thickness and fibrosis, and recovered peritoneal membrane function in a peritoneal fibrosis mouse model.


Asunto(s)
Benzamidas/farmacología , Células Epiteliales/efectos de los fármacos , Fibrosis Peritoneal/patología , Peritoneo/citología , Pirazoles/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Clorhexidina/análogos & derivados , Clorhexidina/toxicidad , Colágeno/metabolismo , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Peritoneal/inducido químicamente , Peritoneo/efectos de los fármacos , Fosforilación , Procesamiento Proteico-Postraduccional , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores
9.
J Cell Physiol ; 235(1): 364-379, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31236971

RESUMEN

BACKGROUND: Transforming growth factor-ß1 (TGF-ß1) is a profibrotic cytokine which induces mesothelial cell mesothelial-to-mesenchymal transition (MMT) and peritoneal fibrosis in patients receiving treatment of peritoneal dialysis. Because thrombospondin-1 (TSP-1) is able to activate latent TGF-ß1 in vivo, we investigated whether blockade of TSP-1 could modulate mesothelial cell MMT and ameliorate peritoneal fibrosis. METHODS: Human pleural mesothelial cells (Met-5A cells) were treated with TSP-1 and addition of TGF-ß1 neutralizing antibody to assess the effect of TSP-1 on MMT. Furthermore, TSP-1 blocking peptide Leu-Ser-Lys-Leu (LSKL) was applied to Met-5A cells treated with 4.25% d-glucose to determine its function in high glucose-induced MMT. Consequently, a uremic dialysate injection rat model was set up to confirm the results in vivo. RESULTS: Exposure of Met-5A cells to TSP-1 increased TGF-ß1 secretion, expression and bioactivity, triggered Smad3 phosphorylation, upregulated the expression of mesenchymal molecules including fibronectin, collagen type III, α-smooth muscle actin, Snail, and decreased calretinin expression. The effect was partially attenuated by TGF-ß1 neutralizing antibody. TSP-1 expression in Met-5A cells was increased by 4.25% d-glucose, followed by increased secretion and bioactivity of TGF-ß1, the onset of Smad3 phosphorylation and induction of MMT. LSKL significantly attenuated high glucose-mediated mesothelial cell MMT and ameliorated peritoneal fibrosis in uremic rats receiving dextrose dialysate injection. CONCLUSIONS: Taken together, these data demonstrated that TSP-1 contributes to mesothelial cell MMT by activating TGF-ß1/Smad3 signaling pathway and blockade of TSP-1 attenuates high glucose-mediated mesothelial cell MMT and peritoneal fibrosis.


Asunto(s)
Fibrosis Peritoneal/patología , Proteína smad3/metabolismo , Trombospondina 1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Diferenciación Celular/fisiología , Línea Celular , Regulación hacia Abajo/genética , Células Epiteliales/citología , Glucosa , Humanos , Masculino , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
10.
J Cell Physiol ; 234(5): 5915-5925, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30515805

RESUMEN

Peritoneal fibrosis remains to be one of the most severe causes of failure in continuous peritoneal dialysis. The current study cultured peritoneal mesothelial cells in high glucose to stimulate the environment of peritoneal fibrosis model in rats, and investigate whether microRNA-21 (miR-21) targeting Sprouty-1 affects high glucose-induced fibrosis in peritoneal mesothelial cells via the rennin angiotensin system (Ras)-mitogen-activated protein kinase (MAPK) signaling pathway, as well as potential mechanisms. Peritoneal tissues in fibrosis rats were collected to extract peritoneal mesothelial cells, which, after in vitro culture, were transfected with a series of mimic or inhibitor of miR-21, or the small interfering RNA (siRNA) against Sprouty-1. Reverse-transcription quantitative polymerase chain reaction and western blot analyses were performed to determine the levels of related genes or proteins. MTT assay and flow cytometry were conducted to observe the cell viability and cell apoptosis of peritoneal mesothelial cells. Dual-luciferase reporter gene assay revealed that Sprouty-1 is the target gene of miR-21. Peritoneal fibrosis manifested with elevated miR-21, extracellular-signal-regulated kinase (ERK), c-Jun NH2-terminal protein kinase (JNK), RAS and p38MAPK but reduced Sprouty-1. Cells transfected with miR-21 mimic exhibited decreased Sprouty-1 expressions, but increased levels of ERK, JNK, RAS, and p38MAPK. As for cellular process, miR-21 mimic or siRNA against Sprouty-1 exposure reduced cell viability, which resulted in more cells arrested at the G1 stage, and induced apoptosis. In contrast, miR-21 inhibitor exposure was observed to have induced effects on peritoneal mesothelial cells. These key findings provide evidence that miR-21 inhibits Sprouty-1 to promote the progression of fibrosis in peritoneal mesothelial cells by activating the Ras-MAPK signaling pathway.


Asunto(s)
Glucosa , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fibrosis Peritoneal/enzimología , Peritoneo/enzimología , Proteínas ras/metabolismo , Animales , Apoptosis , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Puntos de Control de la Fase G1 del Ciclo Celular , Masculino , MicroARNs/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas del Tejido Nervioso/genética , Diálisis Peritoneal , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/genética , Fibrosis Peritoneal/patología , Peritoneo/patología , Ratas Sprague-Dawley , Transducción de Señal , Proteínas ras/genética
11.
BMC Nephrol ; 20(1): 411, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31727005

RESUMEN

BACKGROUND: Peritoneal fibrosis is the most common complication of peritoneal dialysis, but there is currently no effective treatment. We previously reported that suramin pretreatment prevents the development of peritoneal fibrosis in a rat model of peritoneal fibrosis induced by chlorhexidine gluconate (CG). Here, we further examined the effectiveness of delayed administration of suramin on peritoneal fibrosis and the mechanism (s) involved in this process. METHODS: In the rat model of peritoneal fibrosis induced by CG, suramin or saline was administered at day 21 and 28. All rats were then sacrificed to collect peritoneal tissues for Western blot analysis and histological staining at day 35. RESULTS: Our results demonstrated that delayed administration of suramin starting at 21 days following CG injection can ameliorate peritoneal damage, with greater efficacy after two injections. Suramin also reduced the expression of α-smooth muscle actin, Collagen 1, and Fibronectin and suppressed phosphorylation of Smad-3, epidermal growth factor receptor (EGFR), signal transducers, activator of transcription 3 (STAT3) as well as extracellular signal-regulated kinases 1/2 (ERK 1/2) in the peritoneum injured with CG. Moreover, delayed administration of suramin inhibited overproduction of transforming growth factor-ß1(TGF-ß1) and expression of several pro-inflammatory cytokines, including monocyte chemoattractant protein-1, tumor necrosis factor-α, interleukin-1, and interleukin-6. CONCLUSIONS: Our results indicated that suramin can attenuate progression of peritoneal fibrosis by a mechanism involving inhibition of the TGF-ß1/Smad3 and EGFR signaling pathways as well as suppression of multiple proinflammatory cytokines. Thus, suramin may have the potential to offer an effective treatment for peritoneal fibrosis.


Asunto(s)
Antineoplásicos/administración & dosificación , Fibrosis Peritoneal/prevención & control , Suramina/administración & dosificación , Actinas/metabolismo , Animales , Quimiocina CCL2/metabolismo , Clorhexidina/análogos & derivados , Colágeno Tipo I/metabolismo , Receptores ErbB/metabolismo , Fibronectinas/metabolismo , Interleucina-1/metabolismo , Interleucina-6/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/metabolismo , Peritoneo/efectos de los fármacos , Peritoneo/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Proteína smad3/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
12.
Ren Fail ; 41(1): 267-277, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30982374

RESUMEN

OBJECTIVE: To compare the anti-peritoneal fibrotic effects between a mammalian target of rapamycin complex 1-specific blocker and a phosphatidyl-inositol 3-kinase/mammalian target of rapamycin dual-blocker. METHODS: A total of 40 male Sprague-Dawley rats were randomly divided into five groups with eight animals per group. The normal group (N group) did not receive any intervention. The normal saline group (NS group) received an intraperitoneal injection of normal saline at 1 ml/100 g daily. The model group (3 W group), rapamycin (RAPA) group and BEZ235 (PI3K/mTOR dual-blocker) group all received an intraperitoneal injection of 0.1% chlorhexidine gluconate at 1 ml/100g daily. And the RAPA and BEZ235 groups also received a 0.5 mg/d RAPA or 2.5 mg/d BEZ235 gavage every day, respectively. Rats in each group were sacrificed after 3 weeks. RESULTS: Immunohistochemistry, real-time PCR and western blotting analysis of fibrosis-related indicators (FN, Col 1, and α-SMA) confirmed that RAPA and BEZ235 significantly inhibited peritoneal fibrosis and that these two drugs had similar effects. The p-Akt, p-mTOR, p-p70S6K expression levels were significantly up-regulated in the 3 W group compared to the NS group, confirming that the mTOR pathway was significantly activated during peritoneal fibrosis. RAPA significantly inhibited the phosphorylation of mTOR and p70S6K but did not have significant effects on p-Akt upstream of mTOR. BEZ235 had significant inhibitory effects on all signaling molecules (p-Akt, p-mTOR, and p-p70S6K) in the mTOR pathway. CONCLUSION: RAPA did not up-regulate p-Akt in a negative feedback fashion. Both drugs effectively inhibited peritoneal fibrosis.


Asunto(s)
Imidazoles/farmacología , Fallo Renal Crónico/terapia , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/prevención & control , Quinolinas/farmacología , Sirolimus/farmacología , Animales , Clorhexidina/administración & dosificación , Clorhexidina/análogos & derivados , Clorhexidina/toxicidad , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Imidazoles/uso terapéutico , Inyecciones Intraperitoneales , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinolinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
13.
J Cell Mol Med ; 22(7): 3452-3463, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29654659

RESUMEN

MiRNAs contribute greatly to epithelial to mesenchymal transition (EMT) of peritoneal mesothelial cells (PMCs), which is a crucial step in peritoneal fibrosis (PF). In this study, we tried to profile whether miRNA expression differences exist after human umbilical cord mesenchymal stem cells (hUCMSCs) treatment in PF rats and investigate the possible role of miR-153-3p involved in anti-EMT process. We randomly assigned 34 rats into three groups: control group (Group Control), MGO-induced PF rats (Group MGO) and hUCMSCs-treated rats (Group MGO + hUCMSCs). MiRNA microarrays and real-time PCR analyses were conducted in three groups. α-SMA, Snail1 and E-cadherin expression were detected by Western blot. Luciferase reporter assays were used to detect the effects of miR-153-3p overexpression on Snai1 in rat peritoneal mesothelial cells (RPMCs). We identified differentially expressed miRNAs related to EMT, in which miR-153-3p demonstrated the greatest increase in Group MGO + hUCMSCs. Transient cotransfection of miR-153-3p mimics with luciferase expression plasmids resulted in a significant repression of Snai1 3'-untranslated region luciferase activity in RPMCs. These studies suggest that miR-153-3p is a critical molecule in anti-EMT effects of hUCMSCs in MGO-induced PF rats. MiR-153-3p might exert its beneficial effect through directly targeting Snai1.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Fibrosis Peritoneal/genética , Fibrosis Peritoneal/terapia , Regiones no Traducidas 3' , Animales , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Transición Epitelial-Mesenquimal/genética , Regulación de la Expresión Génica , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/patología , Piruvaldehído , Ratas Wistar , Factores de Transcripción de la Familia Snail/genética , Factor de Crecimiento Transformador beta1/farmacología , Cordón Umbilical/citología , Regulación hacia Arriba
14.
Am J Physiol Renal Physiol ; 315(6): F1732-F1746, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30280601

RESUMEN

In a previous study of fungal peritoneal injury in peritoneal dialysis patients, complement (C)-dependent pathological changes were developed in zymosan (Zy)-induced peritonitis by peritoneal scraping. However, the injuries were limited to the parietal peritoneum and did not show any fibrous encapsulation of the visceral peritoneum, which differs from human encapsular peritoneal sclerosis (EPS). We investigated peritoneal injury in a rat model of Zy-induced peritonitis pretreated with methylglyoxal (MGO) instead of scraping (Zy/MGO peritonitis) to clarify the role of C in the process of fibrous encapsulation of the visceral peritoneum. Therapeutic effects of an anti-C5a complementary peptide, AcPepA, on peritonitis were also studied. In Zy/MGO peritonitis, peritoneal thickness, fibrin exudation, accumulation of inflammatory cells, and deposition of C3b and C5b-9 with loss of membrane C regulators were increased along the peritoneum until day 5. On day 14, fibrous encapsulation of the visceral peritoneum was observed, resembling human EPS. Peritoneal injuries and fibrous changes were significantly improved with AcPepA treatment, even when AcPepA was administered following injection of Zy in Zy/MGO peritonitis. The data show that C5a might play a role in the development of encapsulation-like changes in the visceral peritoneum in Zy/MGO peritonitis. AcPepA might have therapeutic effects in fungal infection-induced peritoneal injury by preventing subsequent development of peritoneal encapsulation.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Complemento C5a/antagonistas & inhibidores , Inactivadores del Complemento/farmacología , Fibrosis Peritoneal/prevención & control , Peritoneo/efectos de los fármacos , Piruvaldehído , Zimosan , Animales , Complemento C5a/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Masculino , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/inmunología , Fibrosis Peritoneal/patología , Peritoneo/inmunología , Peritoneo/patología , Peritonitis/inducido químicamente , Peritonitis/inmunología , Peritonitis/metabolismo , Peritonitis/patología , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
15.
Kidney Int ; 93(6): 1384-1396, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29571940

RESUMEN

Ultrafiltration failure is a major complication of long-term peritoneal dialysis, resulting in dialysis failure. Peritoneal fibrosis induced by continuous exposure to high glucose dialysate is the major contributor of ultrafiltration failure, for which there is no effective treatment. Overactivation of several signaling pathways, including transforming growth factor-ß1 (TGF-ß1) and platelet-derived growth factor (PDGF) pathways, contribute to the development of peritoneal fibrosis. Therefore, simultaneously blocking multiple signaling pathways might be a potential novel method of treating peritoneal fibrosis. Previously, we showed that core fucosylation, an important posttranslational modification of the TGF-ß1 receptors, can regulate the activation of TGF-ß1 signaling in renal interstitial fibrosis. However, it remains unclear whether core fucosylation affects the progression of peritoneal fibrosis. Herein, we show that core fucosylation was enriched in the peritoneal membrane of rats accompanied by peritoneal fibrosis induced by a high glucose dialysate. Blocking core fucosylation dramatically attenuated peritoneal fibrosis in the rat model achieved by simultaneously inactivating the TGF-ß1 and PDGF signaling pathways. Next the protective effects of blocking core fucosylation and imatinib (a selective PDGF receptor inhibitor) on peritoneal fibrosis were compared and found to exhibit a greater inhibitory effect over imatinib alone, suggesting that blocking activation of multiple signaling pathways may have superior inhibitory effects on the development of peritoneal fibrosis. Thus, core fucosylation is essential for the development of peritoneal fibrosis by regulating the activation of multiple signaling pathways. This may be a potential novel target for drug development to treat peritoneal fibrosis.


Asunto(s)
Soluciones para Diálisis , Fucosa/metabolismo , Fucosiltransferasas/metabolismo , Glucosa , Diálisis Peritoneal/métodos , Fibrosis Peritoneal/prevención & control , Peritoneo/metabolismo , Interferencia de ARN , Animales , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fucosiltransferasas/genética , Mesilato de Imatinib/farmacología , Masculino , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Peritoneo/efectos de los fármacos , Peritoneo/patología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas Sprague-Dawley , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo
16.
Kidney Int ; 94(2): 346-362, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29861057

RESUMEN

Peritoneal membrane failure due to fibrosis limits the use of peritoneal dialysis (PD). Peritoneal fibrosis may potentially be induced by sterile inflammation caused by ongoing cellular stress due to prolonged exposure to PD solutions (PDS). Effective therapies to prevent this process remain to be developed. Toll-like receptors (TLRs) mediate sterile inflammation by recognizing damage-associated molecular patterns (DAMPs) released by cellular stress. We evaluated the involvement of TLRs and DAMPs in PDS-induced fibrosis models and the therapeutic potential of TLR-DAMP targeting for preventing fibrosis. A range of PDS elicited pro-inflammatory and fibrotic responses from PD patient peritoneal leukocytes, mesothelial cells and mouse peritoneal leukocytes. TLR2/4 blockade of human peritoneal cells or TLR2/4 knockouts inhibited these effects. PDS did not induce rapid ERK phosphorylation or IκB-α degradation, suggesting that they do not contain components capable of direct TLR activation. However, PDS increased the release of Hsp70 and hyaluronan, both TLR2/4 DAMP ligands, by human and mouse peritoneal cells, and their blockade decreased PDS-driven inflammation. Soluble TLR2, a TLR inhibitor, reduced PDS-induced pro-inflammatory and fibrotic cytokine release ex vivo. Daily catheter infusion of PDS in mice caused peritoneal fibrosis, but co-administration of soluble TLR2 prevented fibrosis, suppressed pro-fibrotic gene expression and pro-inflammatory cytokine production, reduced leukocyte/neutrophil recruitment, recovered Treg cell levels and increased the Treg:Th17 ratio. Thus, TLR2/4, Hsp70 and hyaluronan showed major roles in PDS-induced peritoneal inflammation and fibrosis. The study demonstrates the therapeutic potential of a TLR-DAMP targeting strategy to prevent PDS-induced fibrosis.


Asunto(s)
Soluciones para Diálisis/toxicidad , Inflamación/prevención & control , Fibrosis Peritoneal/prevención & control , Receptor Toll-Like 2/administración & dosificación , Receptores Toll-Like/antagonistas & inhibidores , Alarminas/antagonistas & inhibidores , Alarminas/inmunología , Alarminas/metabolismo , Animales , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Voluntarios Sanos , Humanos , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Fallo Renal Crónico/terapia , Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Diálisis Peritoneal/efectos adversos , Diálisis Peritoneal/métodos , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/inmunología , Fibrosis Peritoneal/patología , Peritoneo/citología , Peritoneo/patología , Cultivo Primario de Células , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Receptor Toll-Like 2/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
17.
Am J Nephrol ; 48(6): 456-464, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30472702

RESUMEN

BACKGROUND: Peritoneal fibrosis is a devastating complication of peritoneal dialysis. However, its precise mechanism is unclear, and specific treatments have not yet been established. Recent evidence suggests that the sonic hedgehog (SHH) signaling pathway is involved in tissue fibrogenesis. Drugs that inhibit this pathway are emerging in the field of anti-fibrosis therapy. Itraconazole, an anti-fungal agent, was also recently recognized as an inhibitor of the SHH signaling pathway. In this study, we used a mouse model to investigate whether the SHH signaling pathway is involved in the development of peritoneal fibrosis and the effects of itraconazole on peritoneal fibrosis. METHODS: Peritoneal fibrosis was induced by intraperitoneal (IP) injection of 0.1% chlorhexidine gluconate (CG) solution every other day for 4 weeks, with or without itraconazole treatment (20 mg/kg, IP injection on a daily basis). Male C57BL/6 mice were divided into 4 groups: saline group, saline plus itraconazole group, CG group, and CG plus itraconazole group. Isotonic saline was administered intraperitoneally to the control group. The peritoneal tissues were evaluated for histological changes, expression of fibrosis markers, and the main components of the SHH signaling pathway. RESULTS: Peritoneal thickening was evident in the CG group and was significantly decreased by itraconazole administration (80.4 ± 7.7 vs. 28.2 ± 3.8 µm, p < 0.001). The expression of the following SHH signaling pathway components was upregulated in the CG group and suppressed by itraconazole treatment: SHH, patched, smoothened, and glioma-associated oncogene transcription factor 1. The IP injection of CG solution increased the expression of fibrosis markers such as α-smooth muscle actin and transforming growth factor-ß1 in the peritoneal tissues. Itraconazole treatment significantly decreased the expression of these markers. CONCLUSION: Our study provides the first evidence that the SHH signaling pathway may be implicated in peritoneal fibrosis. It also demonstrates that itraconazole treatment has protective effects on peritoneal fibrosis through the regulation of the SHH signaling pathway. These findings suggest that blockage of the SHH signaling pathway is a potential therapeutic strategy for peritoneal fibrosis.


Asunto(s)
Proteínas Hedgehog/metabolismo , Itraconazol/farmacología , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Clorhexidina/administración & dosificación , Clorhexidina/análogos & derivados , Clorhexidina/toxicidad , Modelos Animales de Enfermedad , Humanos , Inyecciones Intraperitoneales , Itraconazol/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/patología , Peritoneo/efectos de los fármacos , Peritoneo/patología , Resultado del Tratamiento
18.
Exp Cell Res ; 360(2): 375-383, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28942023

RESUMEN

Epithelial-mesenchymal transition(EMT) is the main reason for peritoneal fibrosis and the mechanism underlying peritoneal EMT were extensively studied in recent years. Recent researches showed that miRNAs were so important in the development of organ EMT and fibrosis, the role of microRNAs on peritoneal dialysis have also been studied. In the current study, we investigated microRNA-23(miR-23) expression in high glucose(HG) induced EMT in human mesotheial peritoneal cells(HPMCs). We found that HG promoted EMT, which was characterized by the upregulation of mesenchymal markers α-SMA and FN and downregulation of epithelial marker E-cadherin. The expression miR-23 showed a significant upregulation when treated with HG. Enhanced expression of miR-23 could aggravate HG induced EMT by targeting VDR, inhibition of miR-23 in HPMCs could reverse HG induced EMT by targeting VDR. Furthermore, VDRshRNA exacerbated the EMT process and reversed miR-23 inhibitor-attenuated EMT process in HPMCs. These data manifested that miR-23 played a key role in HG-induced EMT of HPMCs by targeting VDR.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Epitelio/efectos de los fármacos , Glucosa/efectos adversos , MicroARNs/fisiología , Peritoneo/efectos de los fármacos , Receptores de Calcitriol/genética , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Epitelio/metabolismo , Epitelio/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/etiología , Fibrosis Peritoneal/genética , Peritoneo/metabolismo , Peritoneo/patología
19.
J Recept Signal Transduct Res ; 37(4): 409-415, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28292218

RESUMEN

OBJECTIVE: It has been proved that lactate-4.25% dialysate could result in peritoneal fibrosis by inducing alternative activation of macrophages in our previous study, but the mechanism of high glucose-induced alternative activation has not been elucidated. This study was, therefore, to investigate the mechanism by high glucose stimuli. METHODS: In this study, Raw264.7 (murine macrophage cell line) cells were cultured and stimulated by 4.25% glucose medium, and mannitol medium was used as osmotic pressure control. Cells were harvested at 0 h, 4 h, 8 h, and 12 h to examine the expression of Arg-1, CD206, and p-Akt. After blocking PI3K by LY294002, the expression of Arg-1, CD206, and p-Akt was examined again. RESULTS: The expression of Arg-1 and CD206 was increased in a time-dependent manner induced by high glucose medium. On the contrary, there was mainly no Agr-1 or CD206 expressed in cells cultured in the mannitol medium with the same osmotic pressure. What's more, Akt was phosphorylated at the eighth hour stimulated by high glucose medium, and LY294002 inhibited the expression of Arg-1 and CD206 by blocking the phosphorylation of Akt. CONCLUSIONS: Our study indicated that high glucose rather than high osmotic pressure induced M2 phenotype via PI3K/Akt signaling pathway.


Asunto(s)
Glucosa/toxicidad , Macrófagos/efectos de los fármacos , Fibrosis Peritoneal/genética , Proteínas Proto-Oncogénicas c-akt/genética , Animales , Arginasa/genética , Cromonas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Lectinas Tipo C/genética , Macrófagos/patología , Receptor de Manosa , Lectinas de Unión a Manosa/genética , Ratones , Morfolinas/farmacología , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosforilación/efectos de los fármacos , Células RAW 264.7/efectos de los fármacos , Células RAW 264.7/metabolismo , Receptores de Superficie Celular/genética , Transducción de Señal/efectos de los fármacos
20.
Am J Physiol Renal Physiol ; 310(5): F342-50, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26661649

RESUMEN

HL156A is a novel AMP-activated protein kinase (AMPK) activator. We aimed to investigate the protective mechanism of HL156A against peritoneal fibrosis (PF) in in vivo and in vitro models. The rat PF model was induced by daily intraperitoneally injection of chlorhexidine (CHX) solution containing 0.1% CHX gluconate and 15% ethanol for 4 wk. The rats in the treatment group were treated with HL156A (1 mg·kg(-1)·day(-1)). Control rats were injected with vehicle alone. In vitro, cultured rat peritoneal mesothelial cells (RPMCs) were treated with either high glucose (HG; 50 mM), normal glucose (NG; 5 mM), NG+HL156A, or HG+HL156A. HL156A in supplemented rats ameliorated peritoneal calcification, cocoon formation, bowel obstruction, and PF. Immunohistochemistry showed reduced fibronectin accumulation in the peritoneum of HL156A-treated rats compared with those injected with CHX alone. HL156A treatment of RPMCs inhibited HG-induced myofibroblast transdifferentiation and markers of epithelial-mesenchymal transition (EMT). Moreover, HL156A ameliorated HG-induced transforming growth factor-ß1, Smad3, Snail, and fibronectin expression in the RPMCs via AMPK upregulation. These results suggest that HL156A exhibits a protective effect in PF progression. Further research is warranted to seek the therapeutic potential of HL156A as an antifibrotic agent in peritoneal dialysis patients.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Activadores de Enzimas/farmacología , Guanidinas/farmacología , Fibrosis Peritoneal/prevención & control , Peritoneo/efectos de los fármacos , Pirrolidinas/farmacología , Proteínas Quinasas Activadas por AMP/genética , Animales , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Clorhexidina/análogos & derivados , Citoprotección , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Activación Enzimática , Activadores de Enzimas/síntesis química , Transición Epitelial-Mesenquimal/efectos de los fármacos , Etanol , Fibronectinas/genética , Fibronectinas/metabolismo , Glucosa/metabolismo , Guanidinas/síntesis química , Masculino , Miofibroblastos/efectos de los fármacos , Miofibroblastos/enzimología , Miofibroblastos/patología , Fibrosis Peritoneal/inducido químicamente , Fibrosis Peritoneal/enzimología , Fibrosis Peritoneal/genética , Fibrosis Peritoneal/patología , Peritoneo/enzimología , Peritoneo/patología , Pirrolidinas/síntesis química , Interferencia de ARN , Ratas Wistar , Proteína smad3/genética , Proteína smad3/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA