Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
Am J Physiol Renal Physiol ; 322(1): F105-F119, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34866403

RESUMEN

15-Lipoxygenase (15-LO) is a nonheme iron-containing dioxygenase that has both pro- and anti-inflammatory roles in many tissues and disease states. 15-LO is thought to influence macrophage phenotype, and silencing 15-LO reduces fibrosis after acute inflammatory triggers. The goal of the present study was to determine whether altering 15-LO expression influences inflammation and fibrogenesis in a murine model of unilateral ureteral obstruction (UUO). C57BL/6J mice, 15-LO knockout (Alox15-/-) mice, and 15-LO transgenic overexpressing (15LOTG) mice were subjected UUO, and kidneys were analyzed at 3, 10, and 14 days postinjury. Histology for fibrosis, inflammation, cytokine quantification, flow cytometry, and metabolomics were performed on injured tissues and controls. PD146176, a specific 15-LO inhibitor, was used to complement experiments involving knockout animals. Compared with wild-type animals undergoing UUO, Alox15-/- mouse kidneys had less proinflammatory, profibrotic message along with less fibrosis and macrophage infiltration. PD146176 inhibited 15-LO and resulted in reduced fibrosis and macrophage infiltration similar to Alox15-/- mice. Flow cytometry revealed that Alox15-/- UUO-injured kidneys had a dynamic change in macrophage phenotype, with an early blunting of CD11bHiLy6CHi "M1" macrophages and an increase in anti-inflammatory CD11bHiLy6CInt "M2c" macrophages and reduced expression of the fractalkine receptor chemokine (C-X3-C motif) receptor 1. Many of these findings were reversed when UUO was performed on 15LOTG mice. Metabolomics analysis revealed that wild-type kidneys developed a glycolytic shift postinjury, while Alox15-/- kidneys exhibited increased oxidative phosphorylation. In conclusion, 15-LO manipulation by genetic or pharmacological means induces dynamic changes in the inflammatory microenvironment in the UUO model and appears to be critical in the progression of UUO-induced fibrosis.NEW & NOTEWORTHY 15-Lipoxygenase (15-LO) has both pro- and anti-inflammatory functions in leukocytes, and its role in kidney injury and repair is unexplored. Our study showed that 15-LO worsens inflammation and fibrosis in a rodent model of chronic kidney disease using genetic and pharmacological manipulation. Silencing 15-LO promotes an increase in M2c-like wound-healing macrophages in the kidney and alters kidney metabolism globally, protecting against anaerobic glycolysis after injury.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/metabolismo , Citocinas/metabolismo , Metabolismo Energético , Mediadores de Inflamación/metabolismo , Riñón/enzimología , Metaboloma , Nefritis/etiología , Obstrucción Ureteral/complicaciones , Animales , Araquidonato 12-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/genética , Microambiente Celular , Citocinas/genética , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Fibrosis , Riñón/efectos de los fármacos , Riñón/patología , Leucocitos/enzimología , Inhibidores de la Lipooxigenasa/farmacología , Macrófagos/enzimología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Nefritis/enzimología , Nefritis/patología , Nefritis/prevención & control , Fenotipo , Obstrucción Ureteral/tratamiento farmacológico , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/patología
2.
Int J Med Sci ; 19(2): 321-330, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35165517

RESUMEN

Aim: Endoplasmic reticulum-associated degradation (ERAD), which involves degradation of improperly folded proteins retained in the ER, is implicated in various diseases including chronic kidney disease. This study is aimed to determine the role of ERAD in Klotho deficiency of mice and human kidney tubular epithelial cells (HK-2) with renal interstitial fibrosis (RIF). Methods: Following establishment of a mouse RIF model by unilateral ureteral obstruction (UUO), a specific ERAD inhibitor, Eeyarestatin I (EerI), was administered to experimental animals by intraperitoneal injection. Serum and kidney samples were collected for analysis 10 days after operation. Soluble Klotho levels were measured by enzyme-linked immunosorbent assay, while the degree of kidney injury was assessed by renal histopathology. Renal Klotho expression was determined by quantitative real-time PCR, immunohistochemical and western blotting analyses. ERAD and unfolded protein response (UPR) were evaluated by detecting associated components such as Derlin-1, glucose-regulated protein 78 (GRP78), activating transcription factor 4 (ATF4) and protein disulfide isomerase (PDI). HK-2 cells were exposed to transforming growth factor (TGF)-ß1 with or without EerI, and expressions of related proteins including Klotho, Derlin-1, GRP78, ATF4 and PDI were determined by western blotting analyses. Results: UUO induced severe kidney injuries and RIF. Klotho expression in both serum and kidney tissue was obviously downregulated, while Derlin-1 was notably upregulated, indicating that ERAD was activated to potentially degrade improperly folded Klotho protein in this model. Intriguingly, treatment with EerI led to significantly increased Klotho expression, especially soluble (functional) Klotho. Furthermore, specific inhibition of ERAD increased expression of GRP78, ATF4 and PDI compared with the UUO group. The consistent results in vitro were also obtained in TGF-ß1-treated HK-2 cells exposed to EerI. These observations suggest that UPR was remarkably enhanced in the presence of ERAD inhibition and compensated for excess improperly folded proteins, subsequently contributing to the additional production of mature Klotho protein. Conclusion: ERAD is involved in Klotho deficiency in RIF and its specific inhibition significantly promoted Klotho expression, possibly through enhanced UPR. This may represent a novel regulatory mechanism and new therapeutic target for reversing Klotho deficiency.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico/genética , Riñón/patología , Proteínas Klotho/deficiencia , Nefritis Intersticial/enzimología , Obstrucción Ureteral/enzimología , Animales , Modelos Animales de Enfermedad , Fibrosis , Humanos , Hidrazonas/administración & dosificación , Hidroxiurea/administración & dosificación , Hidroxiurea/análogos & derivados , Inyecciones Intraperitoneales , Túbulos Renales/citología , Proteínas Klotho/efectos de los fármacos , Ratones
3.
J Am Soc Nephrol ; 31(5): 983-995, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32209589

RESUMEN

BACKGROUND: Expression of SerpinB2, a regulator of inflammatory processes, has been described in the context of macrophage activation and cellular senescence. Given that mechanisms for these processes interact and can shape kidney disease, it seems plausible that SerpinB2 might play a role in renal aging, injury, and repair. METHODS: We subjected SerpinB2 knockout mice to ischemia-reperfusion injury or unilateral ureteral obstruction. We performed phagocyte depletion to study SerpinB2's role beyond the effects of macrophages and transplanted bone marrow from knockout mice to wild-type mice and vice versa to dissect cell type-dependent effects. Primary tubular cells and macrophages from SerpinB2 knockout and wild-type mice were used for functional studies and transcriptional profiling. RESULTS: Cultured senescent tubular cells, kidneys of aged mice, and renal stress models exhibited upregulation of SerpinB2 expression. Functionally, lack of SerpinB2 in aged knockout mice had no effect on the magnitude of senescence markers but associated with enhanced kidney damage and fibrosis. In stress models, inflammatory cell infiltration was initially lower in knockout mice but later increased, leading to an accumulation of significantly more macrophages. SerpinB2 knockout tubular cells showed significantly reduced expression of the chemokine CCL2. Macrophages from knockout mice exhibited reduced phagocytosis and enhanced migration. Macrophage depletion and bone marrow transplantation experiments validated the functional relevance of these cell type-specific functions of SerpinB2. CONCLUSIONS: SerpinB2 influences tubule-macrophage crosstalk by supporting tubular CCL2 expression and regulating macrophage phagocytosis and migration. In mice, SerpinB2 expression seems to be needed for coordination and timely resolution of inflammation, successful repair, and kidney homeostasis during aging. Implications of SerpinB2 in human kidney disease deserve further exploration.


Asunto(s)
Lesión Renal Aguda/enzimología , Envejecimiento/inmunología , Senescencia Celular/inmunología , Túbulos Renales/enzimología , Riñón/enzimología , Macrófagos/fisiología , Inhibidor 2 de Activador Plasminogénico/fisiología , Daño por Reperfusión/enzimología , Obstrucción Ureteral/complicaciones , Lesión Renal Aguda/etiología , Lesión Renal Aguda/inmunología , Animales , Movimiento Celular , Quimiocina CCL2/biosíntesis , Quimiocina CCL2/genética , Técnicas de Cocultivo , Inducción Enzimática , Células Epiteliales/metabolismo , Fibrosis , Homeostasis , Riñón/irrigación sanguínea , Riñón/patología , Masculino , Ratones , Ratones Noqueados , Fagocitosis , Inhibidor 2 de Activador Plasminogénico/deficiencia , Daño por Reperfusión/inmunología , Transcriptoma , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/inmunología
4.
Clin Sci (Lond) ; 134(12): 1357-1376, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32490513

RESUMEN

Non-specific inhibition of Rho-associated kinases (ROCKs) alleviated renal fibrosis in the unilateral ureteral obstruction (UUO) model, while genetic deletion of ROCK1 did not affect renal pathology in mice. Thus, whether ROCK2 plays a role in renal tubulointerstitial fibrosis needs to be clarified. In the present study, a selective inhibitor against ROCK2 or genetic approach was used to investigate the role of ROCK2 in renal tubulointerstitial fibrosis. In the fibrotic kidneys of chronic kidney diseases (CKDs) patients, we observed an enhanced expression of ROCK2 with a positive correlation with interstitial fibrosis. In mice, the ROCK2 protein level was time-dependently increased in the UUO model. By treating CKD animals with KD025 at the dosage of 50 mg/kg/day via intraperitoneal injection, the renal fibrosis shown by Masson's trichrome staining was significantly alleviated along with the reduced expression of fibrotic genes. In vitro, inhibiting ROCK2 by KD025 or ROCK2 knockdown/knockout significantly blunted the pro-fibrotic response in transforming growth factor-ß1 (TGF-ß1)-stimulated mouse renal proximal tubular epithelial cells (mPTCs). Moreover, impaired cellular metabolism was reported as a crucial pathogenic factor in CKD. By metabolomics analysis, we found that KD025 restored the metabolic disturbance, including the impaired glutathione metabolism in TGF-ß1-stimulated tubular epithelial cells. Consistently, KD025 increased antioxidative stress enzymes and nuclear erythroid 2-related factor 2 (Nrf2) in fibrotic models. In addition, KD025 decreased the infiltration of macrophages and inflammatory response in fibrotic kidneys and blunted the activation of macrophages in vitro. In conclusion, inhibition of ROCK2 may serve as a potential novel therapy for renal tubulointerstitial fibrosis in CKD.


Asunto(s)
Células Epiteliales/enzimología , Túbulos Renales Proximales/patología , Enfermedades Metabólicas/enzimología , Quinasas Asociadas a rho/antagonistas & inhibidores , Adolescente , Animales , Antiinflamatorios/farmacología , Niño , Preescolar , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Femenino , Fibrosis , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Lactante , Inflamación/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Enfermedades Metabólicas/patología , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Células RAW 264.7 , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba/efectos de los fármacos , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/patología , Quinasas Asociadas a rho/metabolismo
5.
FASEB J ; 33(7): 8249-8262, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30951378

RESUMEN

In this study, we examined the effect of MC1568, a selective class IIa histone deacetylase (HDAC) inhibitor, on the development and progression of renal fibrosis in a murine model of renal fibrosis induced by unilateral ureteral obstruction (UUO). All 4 class IIa HDAC isoforms, in particular HDAC4, were up-regulated in renal epithelial cells of the injured kidney. Administration of MC1568 immediately after UUO injury reduced expression of α-smooth muscle actin (α-SMA), fibronectin, and collagen 1. MC1568 treatment or small interfering RNA-mediated silencing of HDAC4 also suppressed expression of those proteins in cultured renal epithelial cells. Mechanistically, MC1568 abrogated UUO-induced phosphorylation of Smad3, NF-κB, and up-regulation of integrin ɑVß6 in the kidney and inhibited TGF-ß1-induced responses in cultured renal epithelial cells. MC1568 also increased renal expression of klotho, bone morphogenetic protein 7, and Smad7. Moreover, delayed administration of MC1568 at 3 d after ureteral obstruction reversed the expression of α-SMA, fibronectin, and collagen 1 and increased expression of matrix metalloproteinase (MMP)-2 and -9. Collectively, these results suggest that selectively targeting class IIa HDAC isoforms (in particular HDAC4) may inhibit development and progression of renal fibrosis by suppressing activation and expression of multiple profibrotic molecules and increasing expression of antifibrotic proteins and MMPs.-Xiong, C., Guan, Y., Zhou, X., Liu, L., Zhuang, M. A., Zhang, W., Zhang, Y., Masucci, M. V., Bayliss, G., Zhao, T. C., Zhuang, S. Selective inhibition of class IIa histone deacetylases alleviates renal fibrosis.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Enfermedades Renales/enzimología , Pirroles/farmacología , Obstrucción Ureteral/enzimología , Animales , Proteína Morfogenética Ósea 7/metabolismo , Línea Celular Transformada , Fibrosis , Enfermedades Renales/patología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/patología
6.
Am J Physiol Renal Physiol ; 316(2): F360-F371, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30565999

RESUMEN

Renal fibrosis is the pathological hallmark of chronic kidney disease (CKD) and manifests as glomerulosclerosis and tubulointerstitial fibrosis. Reactive oxygen species contribute significantly to renal inflammation and fibrosis, but most research has focused on superoxide and hydrogen peroxide (H2O2). The animal heme peroxidases myeloperoxidase (MPO), eosinophil peroxidase (EPX), and peroxidasin (PXDN) uniquely metabolize H2O2 into highly reactive and destructive hypohalous acids, such as hypobromous and hypochlorous acid. However, the role of these peroxidases and their downstream hypohalous acids in the pathogenesis of renal fibrosis is unclear. Our study defines the contribution of MPO, EPX, and PXDN to renal inflammation and tubulointerstitial fibrosis in the murine unilateral ureteral obstruction (UUO) model. Using a nonspecific inhibitor of animal heme peroxidases and peroxidase-specific knockout mice, we find that loss of EPX or PXDN, but not MPO, reduces renal fibrosis. Furthermore, we demonstrate that eosinophils, the source of EPX, accumulate in the renal interstitium after UUO. These findings point to EPX and PXDN as potential therapeutic targets for renal fibrosis and CKD and suggest that eosinophils modulate the response to renal injury.


Asunto(s)
Peroxidasa del Eosinófilo/metabolismo , Eosinófilos/enzimología , Proteínas de la Matriz Extracelular/metabolismo , Riñón/enzimología , Nefritis Intersticial/enzimología , Peroxidasa/metabolismo , Peroxidasas/metabolismo , Obstrucción Ureteral/enzimología , Animales , Movimiento Celular , Modelos Animales de Enfermedad , Peroxidasa del Eosinófilo/deficiencia , Peroxidasa del Eosinófilo/genética , Eosinófilos/patología , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/genética , Femenino , Fibrosis , Riñón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Nefritis Intersticial/etiología , Nefritis Intersticial/patología , Nefritis Intersticial/prevención & control , Peroxidasa/deficiencia , Peroxidasa/genética , Peroxidasas/deficiencia , Peroxidasas/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/patología , Peroxidasina
7.
Clin Sci (Lond) ; 133(2): 239-252, 2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30617188

RESUMEN

Kidney fibrosis is the common pathophysiological mechanism in end-stage renal disease characterized by excessive accumulation of myofibroblast-derived extracellular matrix. Natriuretic peptides have been demonstrated to have cyclic guanosine monophosphate (cGMP)-dependent anti-fibrotic properties likely due to interference with pro-fibrotic tissue growth factor ß (TGF-ß) signaling. However, in vivo, natriuretic peptides are rapidly degraded by neutral endopeptidases (NEP). In a unilateral ureteral obstruction (UUO) mouse model for kidney fibrosis we assessed the anti-fibrotic effects of SOL1, an orally active compound that inhibits NEP and endothelin-converting enzyme (ECE). Mice (n=10 per group) subjected to UUO were treated for 1 week with either solvent, NEP-/ECE-inhibitor SOL1 (two doses), reference NEP-inhibitor candoxatril or the angiotensin II receptor type 1 (AT1)-antagonist losartan. While NEP-inhibitors had no significant effect on blood pressure, they did increase urinary cGMP levels as well as endothelin-1 (ET-1) levels. Immunohistochemical staining revealed a marked decrease in renal collagen (∼55% reduction, P<0.05) and α-smooth muscle actin (α-SMA; ∼40% reduction, P<0.05). Moreover, the number of α-SMA positive cells in the kidneys of SOL1-treated groups inversely correlated with cGMP levels consistent with a NEP-dependent anti-fibrotic effect. To dissect the molecular mechanisms associated with the anti-fibrotic effects of NEP inhibition, we performed a 'deep serial analysis of gene expression (Deep SAGE)' transcriptome and targeted metabolomics analysis of total kidneys of all treatment groups. Pathway analyses linked increased cGMP and ET-1 levels with decreased nuclear receptor signaling (peroxisome proliferator-activated receptor [PPAR] and liver X receptor/retinoid X receptor [LXR/RXR] signaling) and actin cytoskeleton organization. Taken together, although our transcriptome and metabolome data indicate metabolic dysregulation, our data support the therapeutic potential of NEP inhibition in the treatment of kidney fibrosis via cGMP elevation and reduced myofibroblast formation.


Asunto(s)
Benzazepinas/farmacología , Enfermedades Renales/prevención & control , Riñón/efectos de los fármacos , Miofibroblastos/efectos de los fármacos , Neprilisina/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Obstrucción Ureteral/tratamiento farmacológico , Animales , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Riñón/enzimología , Riñón/patología , Enfermedades Renales/enzimología , Enfermedades Renales/genética , Enfermedades Renales/patología , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/enzimología , Miofibroblastos/patología , Células 3T3 NIH , Neprilisina/metabolismo , Transducción de Señal/efectos de los fármacos , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/genética , Obstrucción Ureteral/patología
8.
Am J Physiol Renal Physiol ; 315(6): F1822-F1832, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30280598

RESUMEN

Renal fibrosis is a common pathological feature in chronic kidney disease (CKD), including diabetic kidney disease (DKD) and obstructive nephropathy. Multiple microRNAs have been implicated in the pathogenesis of both DKD and obstructive nephropathy, although the overall role of microRNAs in tubular injury and renal fibrosis in CKD is unclear. Dicer (a key RNase III enzyme for microRNA biogenesis) was specifically ablated from kidney proximal tubules in mice via the Cre-lox system to deplete micoRNAs. Proximal tubular Dicer knockout (PT- Dicer KO) mice and wild-type (WT) littermates were subjected to streptozotocin (STZ) treatment to induce DKD or unilateral ureteral obstruction (UUO) to induce obstructive nephropathy. Renal hypertrophy, renal tubular apoptosis, kidney inflammation, and tubulointerstitial fibrosis were examined. Compared with WT mice, PT- Dicer KO mice showed more severe tubular injury and renal inflammation following STZ treatment. These mice also developed higher levels of tubolointerstitial fibrosis. Meanwhile, PT- Dicer KO mice had a significantly higher Smad2/3 expression in kidneys than WT mice (at 6 mo of age) in both control and STZ-treated mice. Similarly, UUO induced more severe renal injury, inflammation, and interstitial fibrosis in PT- Dicer KO mice than WT. Although we did not detect obvious Smad2/3 expression in sham-operated mice (2-3 mo old), significantly more Smad2/3 was induced in obstructed PT- Dicer KO kidneys. These results supported a protective role of Dicer-dependent microRNA synthesis in renal injury and fibrosis development in CKD, specifically in DKD and obstructive nephropathy. Depletion of Dicer and microRNAs may upregulate Smad2/3-related signaling pathway to enhance the progression of CKD.


Asunto(s)
ARN Helicasas DEAD-box/deficiencia , Nefropatías Diabéticas/enzimología , Túbulos Renales Proximales/enzimología , Nefritis/enzimología , Insuficiencia Renal Crónica/enzimología , Ribonucleasa III/deficiencia , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Obstrucción Ureteral/enzimología , Animales , ARN Helicasas DEAD-box/genética , Nefropatías Diabéticas/complicaciones , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis , Túbulos Renales Proximales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Nefritis/etiología , Nefritis/genética , Nefritis/patología , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Ribonucleasa III/genética , Transducción de Señal , Regulación hacia Arriba , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/genética , Obstrucción Ureteral/patología
9.
Am J Physiol Renal Physiol ; 315(3): F445-F453, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29465305

RESUMEN

It is well known that proteinuria following urinary tract obstruction is mainly of a tubular nature. However, it is unknown whether there are also changes in glomerular permeability. In this study, we compared glomerular sieving coefficients (θ) of polydisperse fluorescein isothiocyanate (FITC)-Ficoll 70/400 following a 120- or 180-min unilateral ureteral obstruction (UUO) in anesthetized Sprague-Dawley rats. Samples were collected from the obstructed kidney at 5, 15, and 30 min postrelease and analyzed by means of high-pressure size-exclusion chromatography. After 120-min UUO, mean θ for Ficoll70Å was increased ( P < 0.01) from 2.2 ± 0.5 × 10-5 (baseline) to 10.6 ± 10 × 10-5 15 min postrelease (highest value). After 180-min UUO, mean θ for Ficoll70Å was further increased ( P < 0.001) from 1.4 ± 0.5 × 10-5 (baseline) to 40 ± 10 × 10-5 at 5 min postrelease (highest value). Administration of a reactive oxygen species (ROS) scavenger (Tempol; 1 mg·kg-1·min-1) partly abrogated the permeability effects following 120-min UUO but not after 180 min. Moreover, administration of the RhoA kinase inhibitor Y-27632, the nitric oxide synthase inhibitor NG-nitro-l-arginine methyl ester, or Rac-1 inhibition did not ameliorate glomerular hyperpermeability following 180-min UUO. We show, for the first time, that acute UUO results in marked elevations in glomerular permeability. In addition, our data suggest a time-dependent pathophysiology of UUO-induced hyperpermeability, where reactive oxygen species generation may play an important role in the early stages.


Asunto(s)
Antioxidantes/farmacología , Óxidos N-Cíclicos/farmacología , Inhibidores Enzimáticos/farmacología , Tasa de Filtración Glomerular/efectos de los fármacos , Glomérulos Renales/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Obstrucción Ureteral/tratamiento farmacológico , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Amidas/farmacología , Aminoquinolinas/farmacología , Animales , Presión Arterial/efectos de los fármacos , Modelos Animales de Enfermedad , Frecuencia Cardíaca/efectos de los fármacos , Glomérulos Renales/enzimología , Glomérulos Renales/fisiopatología , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteinuria/tratamiento farmacológico , Proteinuria/enzimología , Proteinuria/fisiopatología , Piridinas/farmacología , Pirimidinas/farmacología , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Marcadores de Spin , Factores de Tiempo , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/fisiopatología , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
10.
Kidney Int ; 93(1): 173-187, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29042082

RESUMEN

Src activation has been associated with fibrogenesis after kidney injury. Macrophage-myofibroblast transition is a newly identified process to generate collagen-producing myofibroblasts locally in the kidney undergoing fibrosis in a TGF-ß/Smad3-dependent manner. The potential role of the macrophage-myofibroblast transition in Src-mediated renal fibrosis is unknown. In studying this by RNA sequencing at single-cell resolution, we uncovered a unique Src-centric regulatory gene network as a key underlying mechanism of macrophage-myofibroblast transition. A total of 501 differentially expressed genes associated with macrophage-myofibroblast transition were identified. However, Smad3-knockout largely reduced the transcriptome diversity. More importantly, inhibition of Src largely suppresses ureteral obstruction-induced macrophage-myofibroblast transition in the injured kidney in vivo along with transforming growth factor-ß1-induced elongated fibroblast-like morphology, α-smooth muscle actin expression and collagen production in bone marrow derived macrophages in vitro. Unexpectedly, we further uncovered that Src serves as a direct Smad3 target gene and also specifically up-regulated in macrophages during macrophage-myofibroblast transition. Thus, macrophage-myofibroblast transition contributes to Src-mediated tissue fibrosis. Hence, targeting Src may represent as a precision therapeutic strategy for macrophage-myofibroblast transition-driven fibrotic diseases.


Asunto(s)
Transdiferenciación Celular , Cicatriz/enzimología , Enfermedades Renales/enzimología , Riñón/enzimología , Macrófagos/enzimología , Miofibroblastos/enzimología , Familia-src Quinasas/metabolismo , Animales , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Células Cultivadas , Cicatriz/genética , Cicatriz/patología , Cicatriz/prevención & control , Modelos Animales de Enfermedad , Fibrosis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Riñón/efectos de los fármacos , Riñón/patología , Enfermedades Renales/genética , Enfermedades Renales/patología , Enfermedades Renales/prevención & control , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Inhibidores de Proteínas Quinasas/farmacología , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de la Célula Individual , Proteína smad3/genética , Proteína smad3/metabolismo , Obstrucción Ureteral/tratamiento farmacológico , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/genética , Familia-src Quinasas/genética
11.
Int J Mol Sci ; 19(10)2018 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-30332759

RESUMEN

Urinary tract obstruction and the subsequent development of hydronephrosis can cause kidney injuries, which results in chronic kidney disease. Although it is important to detect kidney injuries at an early stage, new biomarkers of hydronephrosis have not been identified. In this study, we examined whether vanin-1 could be a potential biomarker for hydronephrosis. Male Sprague-Dawley rats were subjected to unilateral ureteral obstruction (UUO). On day 7 after UUO, when the histopathological renal tubular injuries became obvious, the vanin-1 level in the renal pelvic urine was significantly higher than that in voided urine from sham-operated rats. Furthermore, vanin-1 remained at the same level until day 14. There was no significant difference in the serum vanin-1 level between sham-operated rats and rats with UUO. In the kidney tissue, the mRNA and protein expressions of vanin-1 significantly decreased, whereas there was increased expression of transforming growth factor (TGF)-ß1 and Snail-1, which plays a pivotal role in the pathogenesis of renal fibrosis via epithelial-to-mesenchymal transition (EMT). These results suggest that vanin-1 in the renal pelvic urine is released from the renal tubular cells of UUO rats and reflects renal tubular injuries at an early stage. Urinary vanin-1 may serve as a candidate biomarker of renal tubular injury due to hydronephrosis.


Asunto(s)
Amidohidrolasas/orina , Hidronefrosis/enzimología , Hidronefrosis/orina , Pelvis Renal/enzimología , Pelvis Renal/lesiones , Aldehídos/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Fibrosis , Proteínas Ligadas a GPI/orina , Hidronefrosis/patología , Pelvis Renal/diagnóstico por imagen , Pelvis Renal/patología , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/patología , Obstrucción Ureteral/orina
12.
Nephrology (Carlton) ; 22(8): 589-597, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27245114

RESUMEN

AIM: Recent studies indicate that pirfenidone (PFD) may have anti-fibrotic effects in many tissues, but the potential molecular mechanism remains unknown. The purpose of this study is to investigate the potential effects of PFD on epithelial-to-mesenchymal transition (EMT) and renal fibrosis in a unilateral ureteral obstruction (UUO) rat model and the involved molecular mechanism related to cultured human renal proximal tubular epithelial cells (HK-2). METHODS: Sixty rats were randomly divided into three groups: sham-operated, vehicle-treated UUO, and PFD-treated UUO. Kidney specimens were collected at day 7 or 14 after UUO. PFD treatment was also performed for human HK-2. The tubulointerstitial injury, interstitial collagen deposition, and expression of type I and III collagen, α-SMA, S100A4, fibronection and E-cadherin were assessed. In addition, extracellular signal regulated kinase (ERK1/2), p38 MAPK (p38), and c-Jun N-terminal kinase/stress-activated protein kinase (JNK) were also detected. RESULTS: In vitro, PFD significantly attenuated TGF-ß1-induced EMT and extracellular matrix (ECM) synthesis, as determined by reducing expression of α-SMA, type I and III collagen, S100A4, fibronection, and increased expression of E-cadherin. PFD treatment attenuated TGF-ß1-induced up-regulation of phosphorylation of ERK1/2, p38 and JNK. In vivo, PFD reduced the degree of tubulointerstitial injury and renal fibrosis, which was associated with reduced expression of TGF-ß1, type III collagen, α-SMA, S100A4, fibronection, and increased expression of E-cadherin. CONCLUSION: These results suggest that pirfenidone is able to attenuate EMT and fibrosis in vivo and in vitro through antagonizing the MAPK pathway, providing a potential treatment to alleviate renal tubulointerstitial fibrosis.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Enfermedades Renales/prevención & control , Túbulos Renales Proximales/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Piridonas/farmacología , Fármacos Renales/farmacología , Obstrucción Ureteral/tratamiento farmacológico , Actinas/metabolismo , Animales , Antígenos CD , Cadherinas/metabolismo , Línea Celular , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibronectinas/metabolismo , Fibrosis , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Enfermedades Renales/enzimología , Enfermedades Renales/etiología , Enfermedades Renales/patología , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/patología , Masculino , Fosforilación , Ratas Sprague-Dawley , Proteína de Unión al Calcio S100A4/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
BMC Nephrol ; 18(1): 232, 2017 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-28697727

RESUMEN

BACKGROUND: Intermedin [IMD, adrenomedullin-2 (ADM-2)] attenuates renal fibrosis by inhibition of oxidative stress. However, the precise mechanisms remain unknown. Heme oxygenase-1 (HO-1), an antioxidant agent, is associated with antifibrogenic effects. ADM is known to induce HO-1. Whether IMD has any effect on HO-1 is unclear. Herein, we determined whether the antifibrotic properties of IMD are mediated by induction of HO-1. METHODS: Renal fibrosis was induced by unilateral ureteral obstruction (UUO) performed on male Wistar rats. Rat proximal tubular epithelial cell line (NRK-52E) was exposed to rhTGF-ß1 (10 ng/ml) to establish an in vitro model of epithelial-mesenchymal transition (EMT). IMD was over-expressed in vivo and in vitro using the vector pcDNA3.1-IMD. Zinc protoporphyrin (ZnPP) was used to block HO-1 enzymatic activity. IMD effects on HO-1 expression in the obstructed kidney of UUO rat and in TGF-ß1-stimulated NRK-52E were analyzed by real-time RT-PCR, Western blotting or immunohistochemistry. HO activity in the obstructed kidney, contralateral kidney of UUO rat and NRK-52E was examined by measuring bilirubin production. Renal fibrosis was determined by Masson trichrome staining and collagen I expression. Macrophage infiltration and IL-6 expression were evaluated using immunohistochemical analysis. In vivo and in vitro EMT was assessed by measuring α-smooth muscle actin (α-SMA) and E-cadherin expression using Western blotting or immunofluorescence, respectively. RESULTS: HO-1 expression and HO activity were increased in IMD-treated UUO kidneys or NRK-52E. The obstructed kidneys of UUO rats demonstrated significant interstitial fibrosis on day 7 after operation. In contrast, kidneys that were treated with IMD gene transfer exhibited minimal interstitial fibrosis. The obstructed kidneys of UUO rats also had greater macrophage infiltration and IL-6 expression. IMD restrained infiltration of macrophages and expression of IL-6 in UUO kidneys. The degree of EMT was extensive in obstructed kidneys of UUO rats as indicated by decreased expression of E-cadherin and increased expression of α-SMA. In vitro studies using NRK-52E confirmed these observations. EMT was suppressed by IMD gene delivery. However, all of the above beneficial effects of IMD were eliminated by ZnPP, an inhibitor of HO enzyme activity. CONCLUSION: This study demonstrates that IMD attenuates renal fibrosis by induction of HO-1.


Asunto(s)
Adrenomedulina/administración & dosificación , Hemo Oxigenasa (Desciclizante)/biosíntesis , Enfermedades Renales/enzimología , Enfermedades Renales/prevención & control , Neuropéptidos/administración & dosificación , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/terapia , Adrenomedulina/genética , Animales , Células Cultivadas , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Fibrosis/enzimología , Fibrosis/genética , Fibrosis/terapia , Técnicas de Transferencia de Gen , Hemo Oxigenasa (Desciclizante)/genética , Enfermedades Renales/genética , Masculino , Neuropéptidos/genética , Distribución Aleatoria , Ratas , Ratas Wistar , Obstrucción Ureteral/genética
14.
Am J Physiol Renal Physiol ; 310(1): F27-40, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26469975

RESUMEN

Renal fibrosis is an inevitable outcome of chronic kidney disease (CKD). Erythropoietin (EPO) has been recently reported to be able to mitigate renal fibrosis. The mechanism underlying the protective effect of EPO, however, remains elusive. In the present study, employing a mouse model of renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction (UUO), we demonstrated that EPO markedly reduced the disruption of the tubular basement membrane (TBM) through attenuating the activation of tissue plasminogen activator (tPA) and matrix metalloproteinase 9 (MMP9), the major matrix proteolytic network in the obstructed kidney. Instead of acting directly on tPA in the kidney, EPO strongly increased the level of circulating microRNA (miR)-144, which was delivered to the injured renal fibroblasts via extracellular vesicles (EVs) to target the tPA 3'-untranslated region and suppress tPA expression. The protective effect of EPO on mouse TBM was inhibited by miR-144 antagomir. Furthermore, in vitro results confirmed that EPO could stimulate bone marrow-derived Sca-1(+)CD44(+)CD11b(-)CD19(-) cells to secrete miR-144-containing EVs, which markedly suppressed tPA expression, as well as metalloproteinase 9 (MMP9) level and activity, in cultured renal fibroblasts. In conclusion, our study provides the first evidence that EPO protects mouse renal TBM through promoting bone marrow cells to generate and secrete miR-144, which, in turn, is efficiently delivered into the mouse kidney via EVs to inhibit the activation of the tPA/MMP9-mediated proteolytic network. This finding thus suggests that EPO, a hormone widely used to treat anemia in CKD, is a potential therapeutic strategy for renal fibrosis.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Eritropoyetina/farmacología , Vesículas Extracelulares/efectos de los fármacos , Membrana Basal Glomerular/efectos de los fármacos , Enfermedades Renales/prevención & control , Túbulos Renales/efectos de los fármacos , MicroARNs/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Obstrucción Ureteral/tratamiento farmacológico , Regiones no Traducidas 3' , Animales , Sitios de Unión , Células de la Médula Ósea/enzimología , Línea Celular , Citoprotección , Modelos Animales de Enfermedad , Activación Enzimática , Represión Enzimática , Vesículas Extracelulares/enzimología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibrosis , Membrana Basal Glomerular/enzimología , Membrana Basal Glomerular/patología , Enfermedades Renales/enzimología , Enfermedades Renales/genética , Enfermedades Renales/patología , Túbulos Renales/enzimología , Túbulos Renales/patología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , MicroARNs/sangre , MicroARNs/genética , Ratas , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Activador de Tejido Plasminógeno/genética , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/genética , Obstrucción Ureteral/patología
15.
Am J Physiol Renal Physiol ; 308(11): F1297-305, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25810437

RESUMEN

Unilateral ureteral obstruction (UUO) is associated with increased hydrostatic pressure, inflammation, and oxidative stress in the renal parenchyma. Previous studies have demonstrated marked cyclooxygenase (COX)-2 induction in renal medullary interstitial cells (RMICs) in response to UUO. The aim of the present study was to evaluate the effect of quercetin, a naturally occurring antioxidant, on COX-2 induction in vivo and in vitro. Rats subjected to 24 h of UUO were treated intraperitoneally with quercetin (50 mg·kg(-1)·day(-1)). Quercetin partly prevented COX-2 induction in the renal inner medulla in response to UUO. Moreover, RMICs exposed to conditions associated with obstruction, inflammation (produced by IL-1ß), oxidative stress (produced by H2O2), and mechanical stress (produced by stretch) showed increased COX-2 expression. Interestingly, quercetin reduced COX-2 induction in RMICs subjected to stretched. Similarly, PGE2 production was markedly increased in RMICs exposed to stretch and was reversed to control levels by quercetin treatment. Furthermore, stretch-induced phosphorylation of ERK1/2 was blocked by quercetin, and inhibition of ERK1/2 attenuated stretch-induced COX-2 induction in RMICs. These results indicate that quercetin attenuated the induction of COX-2 expression and activity in RMICs exposed to mechanical stress as a consequence of acute UUO and that the MAPK ERK1/2 pathway might be involved in this quercetin-mediated reduction in COX-2.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Médula Renal/efectos de los fármacos , Quercetina/farmacología , Obstrucción Ureteral/enzimología , Enfermedad Aguda , Animales , Peróxido de Hidrógeno/metabolismo , Médula Renal/metabolismo , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Estrés Mecánico , Obstrucción Ureteral/tratamiento farmacológico
16.
Am J Physiol Renal Physiol ; 309(12): F1035-48, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26671967

RESUMEN

Renal oxidative stress is increased in response to ureteral obstruction. In vitro, cyclooxygenase (COX)-2 activity contributes to protection against oxidants. In the present study, we tested the hypothesis that COX-2 activity counters oxidative stress and apoptosis in an in vivo model of obstructive nephropathy. Renal oxidative stress markers, antioxidant enzymes, and markers of tubular injury, tubular dilation, and apoptosis were investigated in COX-2 knockout (COX-2(-/-)) and wild-type (WT) mice subjected to 3 or 7 days of unilateral ureteral obstruction (UUO). In a separate series, WT sham-operated and UUO mice were treated with a selective COX-2 inhibitor, parecoxib. COX-2 increased in response to UUO; the oxidative stress markers 4-hydroxynonenal and nitrotyrosine protein residues increased in kidney tissue with no genotype difference after UUO, whereas the antioxidant enzymes heme oxygenase-1 and SOD2 displayed higher levels in COX-2(-/-) mice. Tubular injury was aggravated by COX-2 deletion, as measured by tubular dilatation, an increase in kidney injury molecule-1, cortical caspase-3 content, and apoptosis index. In conclusion, COX-2 is necessary to protect against tubular injury and apoptosis after UUO but not necessary to protect against oxidative stress. COX-2 is not likely to directly regulate antioxidant enzymes heme oxygenase-1 and SOD in the kidney.


Asunto(s)
Apoptosis , Ciclooxigenasa 2/metabolismo , Obstrucción Ureteral/enzimología , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/farmacología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Ratones , Ratones Noqueados , Estrés Oxidativo/efectos de los fármacos , Obstrucción Ureteral/patología
17.
Mediators Inflamm ; 2015: 693260, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25688176

RESUMEN

Soluble epoxide hydrolase (sEH) is abundantly expressed in kidney and plays a potent role in regulating inflammatory response in inflammatory diseases. However, the role of sEH in progression of chronic kidney diseases such as obstructive nephropathy is still elusive. In current study, wild-type (WT) and sEH deficient (sEH (-/-)) mice were subjected to the unilateral ureteral obstruction (UUO) surgery and the kidney injury was evaluated by histological examination, western blotting, and ELISA. The protein level of sEH in kidney was increased in UUO-treated mice group compared to nonobstructed group. Additionally, UUO-induced hydronephrosis, renal tubular injury, inflammation, and fibrosis were ameliorated in sEH (-/-) mice with the exception of glomerulosclerosis. Moreover, sEH (-/-) mice with UUO showed lower levels of inflammation-related and fibrosis-related protein such as monocyte chemoattractant protein-1, macrophage inflammatory protein-2, interleukin-1ß (IL-1ß), IL-6, inducible nitric oxide synthase, collagen 1A1, and α-actin. The levels of superoxide anion radical and hydrogen peroxide as well as NADPH oxidase activity were also decreased in UUO kidneys of sEH (-/-) mice compared to that observed in WT mice. Collectively, our findings suggest that sEH plays an important role in the pathogenesis of experimental obstructive nephropathy and may be a therapeutic target for the treatment of obstructive nephropathy-related diseases.


Asunto(s)
Epóxido Hidrolasas/genética , Epóxido Hidrolasas/metabolismo , Fibrosis/enzimología , Fibrosis/terapia , Enfermedades Renales/enzimología , Enfermedades Renales/terapia , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/terapia , Animales , Western Blotting , Inmunohistoquímica , Inflamación/enzimología , Inflamación/terapia , Masculino , Ratones , Ratones Endogámicos C57BL
18.
Am J Physiol Renal Physiol ; 307(7): F881-9, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25080524

RESUMEN

Mechanical deformation after congenital ureteral obstruction is traduced into biochemical signals leading to tubular atrophy due to epithelial cell apoptosis. We investigated whether Na(+)/H(+) exchanger 1 (NHE1) could be responsible for HK-2 cell apoptosis induction in response to mechanical stretch through its ability to function as a control point of RhoA and MAPK signaling pathways. When mechanical stretch was applied to HK-2 cells, cell apoptosis was associated with diminished NHE1 expression and RhoA activation. The RhoA signaling pathway was confirmed to be upstream from the MAPK cascade when HK-2 cells were transfected with the active RhoA-V14 mutant, showing higher ERK1/2 expression and decreased p38 activation associated with NHE1 downregulation. NHE1 participation in apoptosis induction was confirmed by specific small interfering RNA NHE1 showing caspase-3 activation and decreased Bcl-2 expression. The decreased NHE1 expression was correlated with abnormal NHE1 activity addressed by intracellular pH measurements. These results demonstrate that mitochondrial proximal tubule cell apoptosis in response to mechanical stretch is orchestrated by signaling pathways initiated by the small GTPase RhoA and followed by the opposing effects of ERK1/2 and p38 MAPK phosphorylation, regulating NHE1 decreased expression and activity.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Intercambiadores de Sodio-Hidrógeno/metabolismo , Estrés Mecánico , Obstrucción Ureteral/enzimología , Proteína de Unión al GTP rhoA/metabolismo , Apoptosis , Línea Celular , Humanos , Túbulos Renales Proximales/fisiopatología , Receptor Cross-Talk , Obstrucción Ureteral/fisiopatología
19.
Am J Physiol Renal Physiol ; 306(2): F259-70, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24226522

RESUMEN

Oxidative stress resulting from unilateral ureteral obstruction (UUO) may be aggravated by increased production of ROS. Previous studies have demonstrated increased cyclooxygenase (COX)-2 expression in renal medullary interstitial cells (RMICs) in response to UUO. We investigated, both in vivo and in vitro, the role of ROS in the induction of COX-2 in rats subjected to UUO and in RMICs exposed to oxidative and mechanical stress. Rats subjected to 3-day UUO were treated with two mechanistically distinct antioxidants, the NADPH oxidase inhibitor diphenyleneiodonium (DPI) and the complex I inhibitor rotenone (ROT), to interfere with ROS production. We found that UUO-mediated induction of COX-2 in the inner medulla was attenuated by both antioxidants. In addition, DPI and ROT reduced tubular damage and oxidative stress after UUO. Moreover, mechanical stretch induced COX-2 and oxidative stress in RMICs. Likewise, RMICs exposed to H2O2 as an inducer of oxidative stress showed increased COX-2 expression and activity, both of which were reduced by DPI and ROT. Similarly, ROS production, which was increased after exposure of RMICs to H2O2, was also reduced by DPI and ROT. Furthermore, oxidative stress-induced phosphorylation of ERK1/2 and p38 was blocked by both antioxidants, and inhibition of ERK1/2 and p38 attenuated the induction of COX-2 in RMICs. Notably, COX-2 inhibitors further exacerbated the oxidative stress level in H2O2-exposed RMICs. We conclude that oxidative stress as a consequence of UUO stimulates COX-2 expression through the activation of multiple MAPKs and that the induction of COX-2 may exert a cytoprotective function in RMICs.


Asunto(s)
Ciclooxigenasa 2/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Obstrucción Ureteral/enzimología , Animales , Antioxidantes/farmacología , Apoptosis/fisiología , Western Blotting , Células Cultivadas , Dinoprostona/metabolismo , Electroforesis en Gel de Poliacrilamida , Inducción Enzimática/fisiología , Técnicas para Inmunoenzimas , Etiquetado Corte-Fin in Situ , Médula Renal/citología , Médula Renal/metabolismo , Masculino , Mitocondrias/metabolismo , NADPH Oxidasas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Adhesión en Parafina , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Estrés Mecánico
20.
Am J Physiol Renal Physiol ; 306(9): F996-1007, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24573388

RESUMEN

Our previous studies support the protective effect of cGMP and cGMP-dependent protein kinase I (PKG-I) pathway on the development of renal fibrosis. Therefore, in the present studies, we determined whether pharmacologically or genetically increased PKG activity attenuates renal fibrosis in a unilateral ureteral obstruction (UUO) model and also examined the mechanisms involved. To increase PKG activity, we used the phosphodiesterase 5 inhibitor sildenafil and PKG transgenic mice. UUO model was induced in wild-type or PKG-I transgenic mice by ligating the left lateral ureteral and the renal fibrosis was observed after 14 days of ligation. Sildenafil was administered into wild-type UUO mice for 14 days. In vitro, macrophage and proximal tubular cell function was also analyzed. We found that sildenafil treatment or PKG transgenic mice had significantly reduced UUO-induced renal fibrosis, which was associated with reduced TGF-ß signaling and reduced macrophage infiltration into kidney interstitial. In vitro data further demonstrated that both macrophages and proximal tubular cells were important sources of UUO-induced renal TGF-ß levels. The interaction between macrophages and tubular cells contributes to TGF-ß-induced renal fibrosis. Taken together, these data suggest that increasing PKG activity ameliorates renal fibrosis in part through regulation of macrophage and tubular cell function, leading to reduced TGF-ß-induced fibrosis.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Enfermedades Renales/prevención & control , Túbulos Renales Proximales/efectos de los fármacos , Inhibidores de Fosfodiesterasa 5/farmacología , Piperazinas/farmacología , Sulfonas/farmacología , Obstrucción Ureteral/tratamiento farmacológico , Actinas/metabolismo , Angiotensina II/farmacología , Animales , Cadherinas/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Mediadores de Inflamación/metabolismo , Enfermedades Renales/enzimología , Enfermedades Renales/genética , Enfermedades Renales/patología , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/patología , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Macrófagos/patología , Masculino , Ratones , Ratones Transgénicos , Fosforilación , Purinas/farmacología , Transducción de Señal/efectos de los fármacos , Citrato de Sildenafil , Proteína Smad2/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/genética , Obstrucción Ureteral/patología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda