Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 37(1): e22632, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36468785

RESUMEN

Peritoneal fibrosis (PF) is an irreversible complication of peritoneal dialysis (PD) that leads to loss of peritoneal membrane function. We investigated PD effluent and serum levels and the tissue expression of chemokine (C-C motif) ligand 8 (CCL8) in patients with PD. Additionally, we investigated their association with PF in a mouse model. Eighty-two end-stage renal disease (ESRD) patients with PD were examined. CCL8 levels were measured via enzyme-linked immunosorbent assays in PD effluents and serum and analyzed with peritoneal transport parameters. Human peritoneal mesothelial cells (hPMCs) were obtained from the PD effluents of 20 patients. Primary cultured hPMCs were treated with recombinant (r) transforming growth factor (TGF)-ß, and CCL8 expression was assessed via western blotting. As the duration of PD increased, the concentration of CCL8 in PD effluents significantly increased. Correlations between peritoneal transport parameters and dialysate CCL8 levels were observed. Western blotting analysis showed that CCL8 was upregulated via rTGF-ß treatment, accompanied by increases in markers of inflammation, fibrosis, senescence, and apoptosis in hPMCs after induction of fibrosis with rTGF-ß. Anti-CCL8 monoclonal antibody (mAb) treatment suppressed the rTGF-ß-induced increase in all analyzed markers. Immunohistochemical analysis revealed that CCL8 along with fibrosis- and inflammation-related markers were significantly increased in the PF mouse model. Functional blockade of CCL8 using a CCR8 inhibitor (R243) abrogated peritoneal inflammation and fibrosis in vivo. In conclusion, high CCL8 levels in PD effluents may be associated with an increased risk of PD failure, and the CCL8 pathway is associated with PF. CCL8 blockade can ameliorate peritoneal inflammation and fibrosis.


Asunto(s)
Fibrosis Peritoneal , Peritonitis , Animales , Ratones , Humanos , Fibrosis Peritoneal/prevención & control , Quimiocina CCL8 , Peritoneo , Quimiocinas , Ligandos , Inflamación , Modelos Animales de Enfermedad
2.
Ren Fail ; 46(1): 2350235, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38721924

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Ácidos Cafeicos , Diálisis Peritoneal , Fibrosis Peritoneal , Alcohol Feniletílico , Sirtuina 1 , Animales , Ratas , Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Ácidos Cafeicos/farmacología , Ácidos Cafeicos/uso terapéutico , Soluciones para Diálisis , Modelos Animales de Enfermedad , Homeostasis/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/etiología , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/prevención & control , Peritoneo/patología , Peritoneo/efectos de los fármacos , Peritoneo/metabolismo , Alcohol Feniletílico/análogos & derivados , Alcohol Feniletílico/farmacología , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Sirtuina 1/efectos de los fármacos , Sirtuina 1/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
3.
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
4.
Ren Fail ; 45(1): 2149411, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36724065

RESUMEN

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.


Asunto(s)
Diálisis Peritoneal , Fibrosis Peritoneal , Animales , Humanos , Ratas , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/farmacología , Transición Epitelial-Mesenquimal , Glucosa/farmacología , Glucosa/metabolismo , Lípidos , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/etiología , Fibrosis Peritoneal/prevención & control , Fibrosis Peritoneal/metabolismo , Peritoneo/metabolismo , Proteínas Klotho/metabolismo
5.
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
6.
Pak J Pharm Sci ; 35(6(Special)): 1767-1772, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36861241

RESUMEN

The current study set out to elucidate the function of epigallocatechin gallate (EGCG) against peritoneal fibrosis in peritoneal dialysis (PD) patients. Firstly, human peritoneal mesothelial cells (HPMCs) were pretreated with 0, 12.5, 25, 50 or 100µmol/L EGCG. Epithelial-mesenchymal transition (EMT) models were induced by advanced glycation end products (AGEs). Untreated-cells were regarded as the blank control group. Changes in proliferation and migration were analyzed by MTT assay and scratch test and levels of HPMC epithelial and interstitial molecular marker proteins were measured by Western blot assay and immunofluorescence, while trans-endothelial resistance was assessed using an epithelial trans membrane cell resistance meter. Inhibition rates of HPMCs, migration numbers and the levels of Snail, E-cadherin, CK and ZO-1 were all decreased, while the levels of α-SMA and FSP1 and trans cellular resistance values were increased in treatment groups (P<0.05). With the increase of EGCG concentrations, HPMCs growth inhibition rates and migration numbers, the levels of α-SMA and FSP1 and TER values were decreased and the levels of Snail, E-cadherin, CK and ZO-1 were enhanced (P<0.05). Overall, the current study highlights that EGCG effectively inhibits the proliferation and migration of HPMCs, increases permeability, suppresses EMT and ultimately delays peritoneal fibrosis.


Asunto(s)
Catequina , Fibrosis Peritoneal , Humanos , Transición Epitelial-Mesenquimal , Fibrosis Peritoneal/prevención & control , Catequina/farmacología , Cadherinas
7.
J Pathol ; 250(1): 79-94, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31579944

RESUMEN

Dysregulation of histone methyltransferase enhancer of zeste homolog 2 (EZH2) has been implicated in the pathogenesis of many cancers. However, the role of EZH2 in peritoneal fibrosis remains unknown. We investigated EZH2 expression in peritoneal dialysis (PD) patients and assessed its role in peritoneal fibrosis in cultured human peritoneal mesothelial cells (HPMCs) and murine models of peritoneal fibrosis induced by chlorhexidine gluconate (CG) or high glucose peritoneal dialysis fluid (PDF) by using 3-deazaneplanocin A (3-DZNeP), and EZH2 conditional knockout mice. An abundance of EZH2 was detected in the peritoneum of patients with PD associated peritonitis and the dialysis effluent of long-term PD patients, which was positively correlated with expression of TGF-ß1, vascular endothelial growth factor, and IL-6. EZH2 was found highly expressed in the peritoneum of mice following injury by CG or PDF. In both mouse models, treatment with 3-DZNeP attenuated peritoneal fibrosis and inhibited activation of several profibrotic signaling pathways, including TGF-ß1/Smad3, Notch1, epidermal growth factor receptor and Src. EZH2 inhibition also inhibited STAT3 and nuclear factor-κB phosphorylation, and reduced lymphocyte and macrophage infiltration and angiogenesis in the injured peritoneum. 3-DZNeP effectively improved high glucose PDF-associated peritoneal dysfunction by decreasing the dialysate-to-plasma ratio of blood urea nitrogen and increasing the ratio of dialysate glucose at 2 h after PDF injection to initial dialysate glucose. Moreover, delayed administration of 3-DZNeP inhibited peritoneal fibrosis progression, reversed established peritoneal fibrosis and reduced expression of tissue inhibitor of metalloproteinase 2, and matrix metalloproteinase-2 and -9. Finally, EZH2-KO mice exhibited less peritoneal fibrosis than EZH2-WT mice. In HPMCs, treatment with EZH2 siRNA or 3-DZNeP suppressed TGF-ß1-induced upregulation of α-SMA and Collagen I and preserved E-cadherin. These results indicate that EZH2 is a key epigenetic regulator that promotes peritoneal fibrosis. Targeting EZH2 may have the potential to prevent and treat peritoneal fibrosis. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Adenosina/análogos & derivados , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Fibrosis Peritoneal/prevención & control , Peritoneo/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/genética , Adenosina/farmacología , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica , Fibrosis Peritoneal/genética , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Peritoneo/metabolismo , Peritoneo/patología , ARN Interferente Pequeño/metabolismo , Transducción de Señal
8.
Ren Fail ; 43(1): 869-877, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33993842

RESUMEN

OBJECTIVE: Peritoneal fibrosis (PF) ultimately causes ultrafiltration failure and peritoneal dialysis (PD) termination, but there are few effective therapies for it. Core fucosylation, which is catalyzed by α1,6-fucosyltransferase (Fut8) in mammals, may play a crucial role in PF development. This study aims to assess the effects of inhibiting core fucosylation of epidermal growth factor (EGF) receptor on PF rats. METHODS: PF rats (established by 4.25% glucose dialysate) were treated with either an adenovirus-Fut8 short hairpin RNA (Fut8shRNA) or adenovirus-control. Masson's staining and net ultrafiltration were performed at week six. Fut8 level and core fucosylation of EGF receptor and collagen I in the peritoneal membrane were assessed, and EGF signaling was detected, including signal transducer and activator of transcription 3 (STAT3), nuclear factor kappa B (NF-κB) and their phosphorylation. Monocyte chemoattractant protein-1 (MCP-1) in peritoneal effluent was examined. RESULTS: Fut8 was upregulated in PF rats but decreased after Fut8shRNA treatment. EGF and EGF receptor expression was upregulated in PF rats, while core fucosylation of EGF receptor decreased after Fut8shRNA treatment. Masson's staining results showed an increase in peritoneal thickness in PF rats but a decrease after Fut8shRNA treatment. Fut8shRNA treatment increased net ultrafiltration, reduced the expression of collagen I and MCP-1 compared to PF rats. Fut8shRNA treatment suppressed phosphorylation of STAT3 and NF-κB in the peritoneal membrane of PF rats. CONCLUSIONS: Fut8shRNA treatment ameliorated the fibrotic changes in PF rats. A potential mechanism may be that Fut8shRNA treatment inactivated EGF signaling pathway by suppressing the phosphorylation of STAT3 and NF-κB.


Asunto(s)
Receptores ErbB/metabolismo , Fucosiltransferasas/farmacología , Glicosilación/efectos de los fármacos , Diálisis Peritoneal/métodos , Fibrosis Peritoneal/prevención & control , Peritoneo/metabolismo , Animales , Quimiocina CCL2/metabolismo , Soluciones para Diálisis , Modelos Animales de Enfermedad , Receptores ErbB/efectos de los fármacos , Fucosiltransferasas/genética , Masculino , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Peritoneo/efectos de los fármacos , Peritoneo/patología , Fosforilación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
9.
Am J Physiol Renal Physiol ; 318(2): F457-F467, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31760768

RESUMEN

As an electrophilic nitroalkene fatty acid, nitro-oleic acid (OA-NO2) exerts multiple biological effects that contribute to anti-inflammation, anti-oxidative stress, and antiapoptosis. However, little is known about the role of OA-NO2 in peritoneal fibrosis. Thus, in the present study, we examined the effects of OA-NO2 on the high glucose (HG)-induced epithelial-mesenchymal transition (EMT) in human peritoneal mesothelial cells (HPMCs) and evaluated the morphological and immunohistochemical changes in a rat model of peritoneal dialysis-related peritoneal fibrosis. In in vitro experiments, we found that HG reduced the expression level of E-cadherin and increased Snail, N-cadherin, and α-smooth muscle actin expression levels in HPMCs. The above-mentioned changes were attenuated by pretreatment with OA-NO2. Additionally, OA-NO2 also inhibited HG-induced activation of the transforming growth factor-ß1/Smad signaling pathway and NF-κB signaling pathway. Meanwhile, OA-NO2 inhibited HG-induced phosphorylation of Erk and JNK. The results from the in vivo experiments showed that OA-NO2 notably relieved peritoneal fibrosis by decreasing the thickness of the peritoneum; it also inhibited expression of transforming growth factor-ß1, α-smooth muscle actin, N-cadherin, and vimentin and enhanced expression of E-cadherin in the peritoneum. Collectively, these results suggest that OA-NO2 inhibits the HG-induced epithelial-mesenchymal transition in HPMCs and attenuates peritoneal dialysis-related peritoneal fibrosis.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Glucosa , Ácidos Oléicos/farmacología , Diálisis Peritoneal , Fibrosis Peritoneal/prevención & control , Peritoneo/efectos de los fármacos , Actinas/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Peritoneo/metabolismo , Peritoneo/patología , Ratas Wistar , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Vimentina/metabolismo
10.
Nephrology (Carlton) ; 24(10): 1081-1089, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30887626

RESUMEN

AIM: This study was conducted to investigate the chronic injury of peritoneal glucose injection on the peritoneum and intestine and the protective effects of omega-3 polyunsaturated fatty acid (ω-3PUFA) during peritoneal dialysis (PD). METHODS: Peritoneal dialysis animal models were established by intraperitoneal injection of 4.25% glucose for 28 days. Protein expression in ileum and peritoneum was measured by immunofloresence and immunohistochemistry. Protein expression in macrophages was measured by Western blot. Fibrosis was analyzed by Masson staining. RESULTS: Peritoneal dialysis significantly increased the structural injury and decreased junction-related protein ZO-1 and occludin expression in ileum, the expression of proteins relating to the activation of M2 (Erg2, IRF4), but not M1 (CD38, IRF5) macrophages. PD significantly increased the expression of TGF-ß1, VEGF and ALK5 protein in peritoneal tissues. PD significantly increased fibrosis (Masson staining) and the expression of fibroblast marker α-SMA in peritoneal tissues. Injection of macrophage clean reagent and ω-3PUFA significantly inhibited M2 activation, and decreased Masson staining, α-SMA, TGF-ß1, VEGF and ALK5 protein expression in peritoneal tissues in PD treated rats. ω-3PUFA injection significantly decreased PD-induced injury in ileum and normalized the expression of ZO-1 and occludin in the ileum of PD rats. CONCLUSION: Omega-3 fatty acids can provide a protective role on PD-induced peritoneal fibrosis and injury of the intestine.


Asunto(s)
Ácidos Grasos Omega-3/farmacología , Glucosa , Íleon , Macrófagos , Fibrosis Peritoneal , Peritoneo , Animales , Soluciones para Diálisis/química , Glucosa/administración & dosificación , Glucosa/efectos adversos , Íleon/efectos de los fármacos , Íleon/metabolismo , Inyecciones Intraperitoneales , Activación de Macrófagos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Diálisis Peritoneal/efectos adversos , Diálisis Peritoneal/métodos , Fibrosis Peritoneal/etiología , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/prevención & control , Peritoneo/efectos de los fármacos , Peritoneo/metabolismo , Sustancias Protectoras/farmacología , Ratas , Edulcorantes/administración & dosificación , Edulcorantes/efectos adversos , Resultado del Tratamiento
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.
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
14.
Am J Physiol Renal Physiol ; 314(2): F167-F180, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28978530

RESUMEN

The characteristic features of chronic peritoneal injury with peritoneal dialysis (PD) are submesothelial fibrosis and neoangiogenesis. Transforming growth factor (TGF)ß and vascular endothelial growth factor (VEGF)-A are the main mediators of fibrosis and neoangiogenesis, respectively; however, the effect of the interaction between them on the peritoneum is not well known. In this study, we investigated the relationship between TGF-ß1 and VEGF-A in inducing peritoneal fibrosis by use of human tissues and dialysate, cultured cells, and animal models. The VEGF-A concentration correlated with the dialysate-to-plasma ratio of creatinine (D/P Cr) ( P < 0.001) and TGF-ß1 ( P < 0.001) in human PD effluent. VEGF-A mRNA levels increased significantly in the peritoneal tissues of human ultrafiltration failure (UFF) patients and correlated with number of vessels ( P < 0.01) and peritoneal thickness ( P < 0.001). TGF-ß1 increased VEGF-A production in human mesothelial cell lines and fibroblast cell lines, and TGF-ß1-induced VEGF-A was suppressed by TGF-ß receptor I (TGFßR-I) inhibitor. Incremental peak values of VEGF-A mRNA stimulated by TGF-ß1 in human cultured mesothelial cells derived from PD patients with a range of peritoneal membrane functions correlated with D/P Cr ( P < 0.05). To evaluate the regulatory mechanisms of VEGF-A and neoangiogenesis in vivo, we administered TGFßR-I inhibitor intraperitoneally in a rat chlorhexidine-induced peritoneal injury (CG) model. TGFßR-I inhibitor administration in the CG model decreased peritoneal thickness ( P < 0.001), the number of vessels ( P < 0.001), and VEGF-A levels ( P < 0.05). These results suggest that neoangiogenesis is associated with fibrosis through the TGF-ß1-VEGF-A pathway in mesothelial cells and fibroblasts. These findings are important when considering the strategy for management of UFF in PD patients.


Asunto(s)
Neovascularización Patológica , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/metabolismo , Peritoneo/irrigación sanguínea , Peritoneo/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Anciano , Anciano de 80 o más Años , Aminoácidos/farmacología , Inhibidores de la Angiogénesis/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Masculino , Persona de Mediana Edad , Fibrosis Peritoneal/etiología , Fibrosis Peritoneal/patología , Fibrosis Peritoneal/prevención & control , Peritoneo/efectos de los fármacos , Peritoneo/patología , Ratas Sprague-Dawley , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Xantenos/farmacología
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.
Cell Physiol Biochem ; 51(6): 2794-2813, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30562743

RESUMEN

BACKGROUND/AIMS: The epithelial-to-mesenchymal transition (EMT) of peritoneal mesothelial cells (PMCs) is a crucial event in the induction of peritoneal fibrosis (PF), in which canonical Wnt/ß-catenin signaling participates. Smads signaling is reported to interact with ß-catenin and synergistically regulates EMT. This study was aimed to reveal the effect of Astragalus on ß-catenin in EMT of PMCs. METHODS: To obtain the role of ß-catenin in EMT, gene transfer into HMrSV5 cell line and rats has been achieved. After Astragalus treatment, EMT markers and signaling pathway-related indicators were detected by western blotting, immunofluorescence, immunohistochemistry, immunoprecipitation and real time-PCR. RESULTS: ß-catenin knockdown suppressed EMT of HMrSV5 cells. Astragalus alleviated EMT of PMCs characterized by increased E-cadherin and decreased α-SMA and Vimentin. In rat model of peritoneal dialysis (PD), Astragalus attenuated peritoneal thickening and fibrosis. Astragalus down-regulated ß-catenin by stabilizing the Glycogen synthase kinase-3ß (GSK-3ß)/ß-catenin complex and further inhibited the nuclear translocation of ß-catenin. Meanwhile, Astragalus down-regulated ß-catenin by enhancing Smad7 expression. Silencing Smad7 antagonized the EMT-inhibitory effect of Astragalus. CONCLUSION: Astragalus inhibits EMT of PMCs by down-regulating ß-catenin. The modulation of ß-catenin in peritoneum can be a novel tool to prevent PF.


Asunto(s)
Planta del Astrágalo , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Peritoneo/efectos de los fármacos , Extractos Vegetales/farmacología , beta Catenina/genética , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Planta del Astrágalo/química , Línea Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Epitelio/efectos de los fármacos , Humanos , Masculino , Fibrosis Peritoneal/genética , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Fibrosis Peritoneal/prevención & control , Peritoneo/citología , Peritoneo/metabolismo , Peritoneo/patología , Extractos Vegetales/química , Extractos Vegetales/uso terapéutico , Ratas Sprague-Dawley , beta Catenina/metabolismo
18.
Nephrol Dial Transplant ; 33(6): 943-953, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29165602

RESUMEN

Background: Connective tissue growth factor (CTGF/CCN2) regulates the signalling of other growth factors and promotes fibrosis. CTGF is increased in mice and humans with peritoneal fibrosis. Inhibition of CTGF has not been examined as a potential therapeutic target for peritoneal fibrosis because systemic CTGF knockout mice die at the perinatal stage. Methods: To study the role of CTGF in peritoneal fibrosis of adult mice, we generated CTGF conditional knockout (cKO) mice by crossing CTGF floxed mice with RosaCreERT2 mice. We administered tamoxifen to Rosa-CTGF cKO mice to delete the CTGF gene throughout the body. We induced peritoneal fibrosis by intraperitoneal injection of chlorhexidine gluconate (CG) in wild-type and Rosa-CTGF cKO mice. Results: Induction of peritoneal fibrosis in wild-type mice increased CTGF expression and produced severe thickening of the peritoneum. In contrast, CG-treated Rosa-CTGF cKO mice exhibited reduced thickening of the peritoneum. Peritoneal equilibration test revealed that the excessive peritoneal small-solute transport in CG-treated wild-type mice was normalized by CTGF deletion. CG-treated Rosa-CTGF cKO mice exhibited a reduced number of αSMA-, Ki67-, CD31- and MAC-2-positive cells in the peritoneum. Analyses of peritoneal mRNA showed that CG-treated Rosa-CTGF cKO mice exhibited reduced expression of Cd68, Acta2 (αSMA), Pecam1 (CD31) and Vegfa. Conclusions: These results indicate that a deficiency of CTGF can reduce peritoneal thickening and help to maintain peritoneal function by reducing angiogenesis and inflammation in peritoneal fibrosis. These results suggest that CTGF plays an important role in the progression of peritoneal fibrosis.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/antagonistas & inhibidores , Inflamación/prevención & control , Neovascularización Patológica/prevención & control , Fibrosis Peritoneal/prevención & control , Inhibidores de la Angiogénesis/farmacología , Animales , Factor de Crecimiento del Tejido Conjuntivo/fisiología , Humanos , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/etiología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Fibrosis Peritoneal/etiología , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Transducción de Señal/efectos de los fármacos
19.
J Am Soc Nephrol ; 27(9): 2631-44, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26677863

RESUMEN

Inhibitors of EGF receptor (EGFR) have antifibrotic effects in several organs, but the effect of these inhibitors on the development of peritoneal fibrosis is unknown. Here, we explored the therapeutic effect of gefitinib, a specific inhibitor of EGFR, on the development and progression of peritoneal fibrosis in a rat model. Daily intraperitoneal injections of chlorhexidine gluconate induced peritoneal fibrosis, indicated by thickening of the submesothelial area with an accumulation of collagen fibrils and activation of myofibroblasts, accompanied by time-dependent phosphorylation of EGFR. Administration of gefitinib immediately after injury prevented the onset of peritoneal fibrosis and delayed administration after the onset of peritoneal fibrosis halted fibrosis progression. Gefitinib treatment abrogated the increased phosphorylation of EGFR, Smad3, signal transducer and activator of transcription 3, and NF-κB during peritoneal fibrosis; it also inhibited the accompanying overproduction of TGF-ß1 and proinflammatory cytokines and the infiltration of macrophages to the injured peritoneum. Moreover, gefitinib significantly reduced the peritoneal increase of CD31-positive blood vessels and vascular EGF-positive cells after injury. Finally, gefitinib also attenuated high glucose-induced peritoneal fibrosis in rats and abrogated TGF-ß1-induced phosphorylation of Smad3 and the epithelial-to-mesenchymal transition of cultured human peritoneal mesothelial cells. These results demonstrate that EGFR contributes to peritoneal fibrosis, inflammation, and angiogenesis, suggesting that EGFR inhibitors may have therapeutic potential in attenuating peritoneal fibrosis.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Fibrosis Peritoneal/prevención & control , Quinazolinas/uso terapéutico , Animales , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Gefitinib , Masculino , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Factor de Crecimiento Transformador beta1
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