Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Total Environ ; 945: 174088, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38908587

RESUMEN

Vegetation degradation in arid and semi-arid regions reduces plant C inputs to the soil, which can impede soil nutrient cycling because of the limited C source for microbial metabolism. However, whether vegetation degradation aggravates microbial nutrient limitation in degraded ecosystems in arid and semi-arid regions is not fully understood. Here, we investigated changes in soil enzyme activity and microbial nutrient limitation along a well-documented gradient of degraded seabuckthorn (Hippophae rhamnoides L.) (slightly degraded, canopy dieback <25 %, moderately degraded, canopy dieback 25 %-75 %, and severely degraded, canopy dieback >75 %) in Liang (long ridge) and gully channel locations in the Pisha Sandstone region of the Loess Plateau, China. We found that as the magnitude of seabuckthorn degradation increased, activities of C-acquiring enzymes and ratios of C:N and C:P enzymes (0.54-0.80 and 0.52-0.77, respectively) increased whereas the N:P enzyme ratio (0.93-0.99) decreased. Stoichiometric modelling further indicated that microorganisms were limited by soil C and P (vector angle >45°) in the seabuckthorn plantation region, and the degradation of seabuckthorn plantation aggravated microbial C and P limitations. Partial least squares path modelling revealed that seabuckthorn degradation (canopy dieback) was the main factor explaining microbial C limitation variations, while soil physicochemical properties (pH and soil moisture content) and understory plant parameters (litter biomass) were the major factors underlying microbial P limitation of long ridge and gully channel formations, respectively. Our findings highlight synergistic changes between aboveground and belowground processes, suggesting an unexpected negative effect of vegetation degradation on soil microbial community and nutrient cycling. These insights offer a direction for the development of plantation nutrients management strategies in semi-arid and arid areas.


Asunto(s)
Hippophae , Fósforo , Microbiología del Suelo , Suelo , China , Fósforo/análisis , Fósforo/metabolismo , Suelo/química , Carbono/metabolismo , Ecosistema , Nitrógeno/metabolismo , Nitrógeno/análisis
3.
Int J Biochem Cell Biol ; 149: 106247, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35753650

RESUMEN

Lipid accumulation and progressive necroinflammation play pivotal roles in the development of diabetic nephropathy. C1q tumour necrosis factor-related protein-3 (CTRP3) is an adipokine with pleiotropic functions in cell proliferation, glucose and lipid metabolism, and inflammation. However, the mechanism and involvement of CTRP3 in lipid metabolism and the necroinflammation of renal tubular cells remain unclear. Here, we report that CTRP3 expression decreased in a time- and concentration-dependent manner in high glucose-stimulated HK-2 cells. We noted that the overexpression of CTRP3 or recombinant CTRP3 (rCTRP3) treatment prevented high glucose-induced lipid accumulation by inhibiting the expression of sterol regulatory element-binding protein-1 and increasing the expression of peroxisome proliferator-activated receptor-α and ATP-binding cassette A1. Moreover, the nucleotide-binding oligomerisation domain-like receptor protein 3-mediated inflammatory response and mixed lineage kinase domain-like protein-dependent necroinflammation were inhibited by CTRP3 overexpression or rCTRP3 treatment in HK-2 cells cultured in high glucose. Furthermore, lipotoxicity-induced by palmitic acid was found to be involved in necroinflammation in HK-2 cells, and CTRP3 displayed the same protective effect. CTRP3 also activated the adenosine monophosphate-activated protein kinase (AMPK) pathway, whereas adenine 9-ß-D-arabinofuranoside, an AMPK inhibitor, replicated the protective effects of CTRP3. Besides, using kidney biopsies from patients with diabetes, we found that decreased CTRP3 expression was accompanied by increased lipid deposition, as well as the structural and functional injury of renal tubular cells. Our findings demonstrate that CTRP3 affects lipid metabolism and necroinflammation in renal tubular cells via the AMPK signalling pathway. Thus, CTRP3 may be a potential therapeutic target in diabetic renal injury.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Monofosfato , Proteínas Portadoras , Glucosa/farmacología , Humanos , Riñón/patología , Lípidos , Factores de Necrosis Tumoral/metabolismo
4.
Front Med (Lausanne) ; 8: 657614, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34485320

RESUMEN

Years of research revealed that crosstalk extensively existed among kidney cells, cell factors and metabolites and played an important role in the development of diabetic kidney disease (DKD). In the last few years, single-cell RNA sequencing (scRNA-seq) technology provided new insight into cellular heterogeneity and genetic susceptibility regarding DKD at cell-specific level. The studies based on scRNA-seq enable a much deeper understanding of cell-specific processes such as interaction between cells. In this paper, we aim to review recent progress in single cell transcriptomic analyses of DKD, particularly highlighting on intra- or extra-glomerular cell crosstalk, cellular targets and potential therapeutic strategies for DKD.

5.
Med Sci Monit ; 25: 1220-1231, 2019 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-30765684

RESUMEN

BACKGROUND PGC-1α can be activated by deacetylation reactions catalyzed by SIRT1. Resveratrol is currently known as a potent activator of SIRT1. However, it is unknown whether the renal-protective effect of resveratrol is further related to activation of the podocyte SIRT1/PGC-1α pathway. MATERIAL AND METHODS High glucose was used to stimulate mouse podocytes. Resveratrol and PGC-1α siRNA transfection were used to perform co-intervention treatments. The protein and mRNA expression levels of SIRT1, PGC-1α, NRF1, and TFAM were detect by immunofluorescence, Western blot analysis, and qRT-PCR in the podocytes, respectively. DCHF-DA and MitoSOX™ staining were used to monitor the total ROS and mitochondrial ROS levels, respectively. The specific activities of complexes I and III were measured using Complex I and III Assay Kits. Mitochondrial membrane potential and cell apoptosis were measured using JC-1 staining and Annexin V-FITC/PI double-staining, respectively. RESULTS We found that high-glucose stimulation results in time-dependent decreases in the expression of SIRT1, PGC-1α, and its downstream genes NRF1 and mitochondrial transcription factor A (TFAM) for mouse podocytes, and increases ROS levels in cells and mitochondria. Moreover, the expression of nephrin was downregulated and the cell apoptotic rate was increased. Resveratrol treatment can improve abnormalities caused by high-glucose stimulation. In addition, it can also reduce the release of mitochondrial cytochrome C and DIABLO proteins to the cytoplasm and increase respiratory chain complex I and III activity and mitochondrial membrane potential. CONCLUSIONS Resveratrol can reduce the oxidative damage and apoptosis of podocytes induced by high-glucose stimulation via SIRT1/PGC-1α-mediated mitochondrial protection.


Asunto(s)
Estrés Oxidativo/efectos de los fármacos , Podocitos/efectos de los fármacos , Resveratrol/farmacología , Animales , Apoptosis/efectos de los fármacos , Técnicas de Cultivo de Célula , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Potencial de la Membrana Mitocondrial , Ratones , Mitocondrias , Proteínas Mitocondriales , Factor Nuclear 1 de Respiración/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/efectos de los fármacos , Procesamiento Proteico-Postraduccional , ARN Interferente Pequeño , Transducción de Señal/efectos de los fármacos , Sirtuina 1/efectos de los fármacos , Factores de Transcripción
6.
J Mol Endocrinol ; 61(3): 101-113, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30307163

RESUMEN

Tubular injury is one of the crucial determinants of progressive renal failure in diabetic nephropathy (DN), while epithelial-to-mesenchymal transition (EMT) of tubular cells contributes to the accumulation of matrix protein in the diabetic kidney. Activation of the nucleotide binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome leads to the maturation of interleukin (IL)-1B and is involved in the pathogenic mechanisms of diabetes. In this study, we explored the role of NLRP3 inflammasome on high glucose (HG) or transforming growth factor-B1 (TGFB1)-induced EMT in HK-2 cells. We evaluated EMT through the expression of α-smooth muscle actin (α-SMA) and E-cadherin as well as the induction of a myofibroblastic phenotype. Reactive oxygen species (ROS) was observed using the confocal microscopy. HG was shown to induce EMT at 48 h, which was blocked by NLRP3 silencing or antioxidant N-acetyl-L-cysteine (NAC). We found that NLRP3 interference could inhibit HG-induced ROS. Knockdown of NLRP3 could prevent HG-induced EMT by inhibiting the phosphorylation of SMAD3, P38 MAPK and ERK1/2. In addition, P38 MAPK and ERK1/2 might be involved in HG-induced NLRP3 inflammasome activation. Besides, TGFB1 induced the activation of NLRP3 inflammasome and the generation of ROS, which were blocked by NLRP3 interference or NAC. Tubular cells exposed to TGFB1 also underwent EMT, and this could be inhibited by NLRP3 shRNA or NAC. These results indicated that knockdown of NLRP3 antagonized HG-induced EMT by inhibiting ROS production, phosphorylation of SMAD3, P38MAPK and ERK1/2, highlighting NLRP3 as a potential therapy target for diabetic nephropathy.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Inflamasomas/metabolismo , Túbulos Renales/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Western Blotting , Línea Celular , Técnica del Anticuerpo Fluorescente , Humanos , Inflamasomas/efectos de los fármacos , Túbulos Renales/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
Lab Invest ; 98(9): 1211-1224, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29884908

RESUMEN

Thioredoxin-interacting protein (TXNIP) is associated with inflammation, tubulointerstitial fibrosis, and oxidative stress in diabetic kidney disease, yet the potential role of TXNIP in nondiabetic renal injury is not well known. This study aimed to investigate the effect of TXNIP on renal injury by creating a unilateral ureteral obstruction (UUO) model in TXNIP knockout (TKO) mice. We performed sham or UUO surgery in 8-week-old TXNIP KO male mice and age and sex-matched wild-type (WT) mice. Animals were killed at 3, 5, 7, or 14 days after surgery, and renal tissues were obtained for RNA, protein, and other analysis. Our results show that the expression of TXNIP was increased in a time-dependent manner in the ligated kidneys. TXNIP deletion reduced renal fibrosis, apoptosis, α-SMA, TGF-ß1 and CTGF expression, and activation of Smad3, p38 MAPK, and ERK1/2 in UUO kidneys. We also found UUO-induced renal F4/80+ macrophage infiltration, MCP-1 expression and activation of NF-κB and NLRP3 inflammasome were attenuated in TKO mice. Furthermore, our study revealed that TXNIP deficiency inhibited the expression of 8-OHdG, heme oxygenase-1 (HO-1) and NADPH oxidase 4 (Nox4) in UUO kidney. In summary, our study suggests that TXNIP plays a key role in the renal inflammation and fibrosis induced by UUO. Inhibition of TXNIP may be a strategy to slow the progression of chronic kidney diseases.


Asunto(s)
Proteínas Portadoras/metabolismo , Nefritis/etiología , Tiorredoxinas/metabolismo , Obstrucción Ureteral/complicaciones , Actinas/genética , Animales , Apoptosis , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Modelos Animales de Enfermedad , Fibrosis , Riñón/metabolismo , Riñón/patología , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nefritis/genética , Nefritis/metabolismo , Estrés Oxidativo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tiorredoxinas/antagonistas & inhibidores , Tiorredoxinas/genética , Factor de Crecimiento Transformador beta1/genética , Obstrucción Ureteral/genética , Obstrucción Ureteral/metabolismo
8.
Biochem J ; 474(16): 2733-2747, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28694352

RESUMEN

Protease-activated receptor-2 (PAR2), which belongs to a specific class of the G-protein-coupled receptors, is central to several inflammation processes. However, the precise molecular mechanism involved remains undefined. Autophagy has been previously shown to affect inflammation. In the present study, we examine the effect of PAR2 on kidney tubular epithelial autophagy and on autophagy-related inflammation and reveal the underlying mechanism involved. Autophagic activity and levels of autophagic marker LC3 were examined in human kidney tubular epithelial cells with PAR2 knockdown or overexpression. We administered the mammalian target of rapamycin (mTOR) inhibitor (rapamycin) or activator (MHY1485) to investigate the function of the phosphoinositide 3-kinase (PI3K)/Akt/mTOR pathway. We also used transforming growth factor-ß1 (TGF-ß1)-induced HK-2 cell inflammation models to investigate the role of PAR2-associated autophagy in kidney tubular epithelial inflammation. PAR2 antagonist and rapamycin were administered to mice after unilateral ureteral obstruction to detect the correlations between PAR2, autophagy, and inflammation. Our results show that PAR2 overexpression in HK-2 cells led to a greater reduction in autophagy via the PI3K/Akt/mTOR pathway activation and induces autophagy-related inflammation. Meanwhile, a knockdown of PAR2 via PAR2 RNAi transfection greatly increased autophagy and alleviated autophagy-associated inflammation. In unilateral ureteral obstruction (UUO) kidneys, PAR2 antagonist treatment greatly attenuated renal inflammation and interstitial injury by enhancing autophagy. Moreover, inhibition of mTOR, rapa, markedly increased autophagy and inhibited the UUO-induced inflammation. We conclude that PAR2 induces kidney tubular epithelial inflammation by inhibiting autophagy via the PI3K/Akt/mTOR signalling pathway. Our results are suggestive that PAR2 inhibition may play a role in the treatment of diseases with increased inflammatory responses in renal systems.


Asunto(s)
Autofagia/genética , Inflamación/genética , Receptor PAR-2/metabolismo , Serina-Treonina Quinasas TOR/genética , Animales , Apoptosis/genética , Humanos , Inflamación/patología , Riñón/metabolismo , Riñón/patología , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Receptor PAR-2/genética , Transducción de Señal , Sirolimus/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
9.
Tissue Cell ; 49(5): 537-544, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28732559

RESUMEN

Tumor necrosis factor-α (TNF-α) is suggested to induce mitochondrial dysfunction and apoptosis of renal tubular epithelial cells that possibly exacerbates renal function in chronic kidney disease (CKD). Here we investigated whether suppressor of cytokine signaling-1 (SOCS-1), an inhibitor of cytokine signaling, was involved in TNF-α-induced human renal tubular epithelial cells (HKCs) oxidative stress and apoptosis. TNF-α promoted the protein and mRNA expression of SOCS-1 in a time and dose dependent manner, along with increased cell apoptosis and activation of apoptosis signal regulating kinase-1(ASK1) in HKCs. Furthermore, overexpression of SOCS-1 in HKCs reduced TNF-α-mediated oxidative stress and apoptosis. Meanwhile, We also found that overexpression of SOCS-1 could regulate the activity of JAK/STAT signaling pathway. In addition, a specific JAK2 inhibitor, AG490, that both attenuated TNF-α-induced oxidative stress, also reduced apoptosis. Taken together, overexpression of SOCS-1 prevented TNF-α-mediated cell oxidative stress and apoptosis may be via suppression of JAK/STAT signaling pathway activation in HKCs.


Asunto(s)
Apoptosis/fisiología , Túbulos Renales/metabolismo , Mitocondrias/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Humanos , Estrés Oxidativo/fisiología
10.
Int J Biochem Cell Biol ; 90: 17-28, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28733250

RESUMEN

Autophagy is an important homoeostatic mechanism for the lysosomal degradation of protein aggregates and damaged cytoplasmic components. Recent studies suggest that autophagy which is induced by TGF-ß1 suppresses kidney fibrosis in renal tubular epithelial cells (RTECs) of obstructed kidneys. Sphingosine kinase 1(SK1), converting sphingosine into endogenous sphingosine-1-phosphate (S1P), was shown to modulate autophagy and involved in the processes of fibrotic diseases. Since SK1 activity is also up-regulated by TGF-ß1, we explored its effect on the induction of autophagy and development of renal fibrosis in this study. In vitro, SK1 expression and activity were markedly increased by TGF-ß1 stimulation in a time and concentration dependent manner, and concomitant changes in autophagic response were observed in HK-2 cells. Further, knockdown of SK-1 led to a decrease of autophagy whereas overexpression of SK1 caused a greater induction of autophagy. In addition, overexpression of SK1 resulted in decreased of mature TGF-ß levels through autophagic degradation. In vivo, SK1 enzymatic activity and autophagic response were both up-regulated in a mouse model of kidney fibrosis induced by unilateral ureteral obstruction (UUO); meanwhile, increased of mature TGF-ß1 and deposition of extracellular matrix (ECM) were observed in tubulointerstitial areas compared with sham-operated mice. However, aggravation of renal fibrosis was detected when SK1 inhibitor PF-543 was applied to suppress SK1 enzymatic activity in UUO mice. At the same time, autophagy was also inhibited by PF-543. Thus, our findings suggest that SK1 activation is renoprotective via induction of autophagy in the fibrotic process.


Asunto(s)
Autofagia , Células Epiteliales/patología , Túbulos Renales/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Autofagia/efectos de los fármacos , Línea Celular , Activación Enzimática/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Fibrosis , Humanos , Masculino , Metanol , Ratones , Pirrolidinas/farmacología , Sulfonas/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/patología
11.
Am J Nephrol ; 46(2): 131-138, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28723681

RESUMEN

BACKGROUND: Inflammation plays a crucial role in renal interstitial fibrosis, the pathway of chronic kidney diseases. Necroptosis is a novel form of regulated cell death, which plays a potential role in inflammation and renal diseases. The small molecule necrostatin-1 (Nec-1) is a specific inhibitor of necroptosis. This study was aimed at determining the role of necroptosis, RIP1/RIP3/mixed lineage kinase domain-like (MLKL) signaling pathway, in renal inflammation and interstitial fibrosis related to primitive tubulointerstitial injury. It was also aimed at evaluating the effect of Nec-1 in renal fibrosis induced by unilateral ureteral obstruction (UUO). METHODS: Renal histology, immunohistochemistry, western blot, and real-time polymerase chain reaction were performed using UUO C57BL/6J mice model. Moreover, we tested whether Nec-1 was renal-protective in the interstitial fibrosis kidney. Mice were exposed to UUO and injected intraperitoneal with Nec-1 or vehicle. RESULTS: The levels of RIP1/RIP3/MLKL protein and mRNA were increased in the obstructed kidneys 7 days after UUO; this was accompanied by changes in renal pathological lesions. Renal histological examination showed lesser renal damage in Nec-1-treated UUO mice. Renal inflammation, assessed by tumor necrosis factor-α, interleukin-1ß, and monocyte chemotactic protein-1 was markedly attenuated by Nec-1. Furthermore, Nec-1 treatment also significantly reduced TGF-ß and α-smooth muscle actin, indicating lesser renal interstitial fibrosis. CONCLUSION: These findings suggest that the participation of necroptosis in UUO is partly demonstrated. And necroptosis inhibition may have a potential role in the treatment of diseases with increased inflammatory response and interstitial fibrosis in renal.


Asunto(s)
Apoptosis/efectos de los fármacos , Imidazoles/farmacología , Indoles/farmacología , Riñón/patología , Nefritis/tratamiento farmacológico , Sustancias Protectoras/farmacología , Animales , Fibrosis , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Imidazoles/uso terapéutico , Indoles/uso terapéutico , Riñón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis/tratamiento farmacológico , Necrosis/etiología , Nefritis/etiología , Nefritis/patología , Sustancias Protectoras/uso terapéutico , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal/efectos de los fármacos , Obstrucción Ureteral/complicaciones
12.
Int J Mol Med ; 39(5): 1317-1324, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28339034

RESUMEN

The transforming growth factor-ß1 (TGF-ß1)/connective tissue growth factor (CTGF) pathway plays an important role in the pathogenesis and progression of chronic kidney disease. Oxidative stress is also involved in TGF-ß1 signalling. Sirtuin 1 (Sirt1) exerts a number of pleiotropic effects, protecting against renal disease, including inhibiting fibrosis and oxidative metabolism. In this study, we investigated the role of the Sirt1 activator, SRT1720, in unilateral ureteral obstruction (UUO)-induced tubulointerstitial fibrosis and aimed to determine whether this role depends on the inhibition of oxidative stress and the TGF-ß1/CTGF pathway. Renal fibrosis was induced by UUO in CD1 mice. SRT1720 (100 mg/kg) was administered by intraperitoneal injection for 3 days prior to UUO and this was continued for 7 days following UUO. Histological changes were examined by Masson's trichrome staining. The expression of fibrosis-related factors was evaluated by immunohistochemistry, western blot analysis and RT-qPCR. Apoptosis was also examined. We also examined the superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GPx) and reduced glutathione (GSH) levels. UUO induced renal fibrosis and apoptosis and decreased Sirt1 expression. The administration of SRT1720 increased the Sirt1 levels and partially attenuated UUO-induced renal fibrosis and apoptosis. Furthermore, SRT1720 attenuated the levels of oxidative stress (it decreased the MDA levels, and increased the SOD, GPx and GSH levels), which suggests that it protected the cells against ROS-induced damage. Moreover, SRT1720 effectively inhibited the levels of TGF-ß1/CTGF induced by UUO. On the whole, these findings indicate that the Sirt1 activator, SRT1720, exerts protective effects against UUO-induced tubulointerstitial fibrosis. The mechanisms of action of SRT1720 may include, at least in part, the suppression of renal oxidative stress and the TGF-ß1/CTGF signalling pathway. The Sirt1 activator may therefore be prove to be a potent therapeutic agent for the treatment of fibrotic kidney disease.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Estrés Oxidativo/efectos de los fármacos , Sirtuina 1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Expresión Génica , Enfermedades Renales/genética , Masculino , Ratones , Sirtuina 1/genética
13.
Int J Biochem Cell Biol ; 79: 1-13, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27497988

RESUMEN

Abnormal lipid metabolism contributes to the renal lipid accumulation, which is associated with diabetic kidney disease, but its precise mechanism remains unclear. The growing evidence demonstrates that thioredoxin-interacting protein is involved in regulating cellular glucose and lipid metabolism. Here, we investigated the effects of thioredoxin-interacting protein on lipid accumulation in diabetic kidney disease. In contrast to the diabetic wild-type mice, the physical and biochemical parameters were improved in the diabetic thioredoxin-interacting protein knockout mice. The increased renal lipid accumulation, expression of acetyl-CoA carboxylase, fatty acid synthase and sterol regulatory element binding protein-1, and phosphorylated Akt and mTOR associated with diabetes in wild-type mice was attenuated in diabetic thioredoxin-interacting protein knockout mice. Furthermore, thioredoxin-interacting protein knockout significantly increased the expression of peroxisome proliferator-activated receptor-α, acyl-coenzyme A oxidase 1 and carnitine palmitoyltransferaser 1 in diabetic kidneys. In vitro experiments, using HK-2 cells, revealed that knockdown of thioredoxin-interacting protein inhibited high glucose-mediated lipid accumulation, expression of acetyl-CoA carboxylase, fatty acid synthase and sterol regulatory element binding protein-1, as well as activation of Akt and mTOR. Moreover, knockdown of thioredoxin-interacting protein reversed high glucose-induced reduction of peroxisome proliferator-activated receptor-α, acyl-coenzyme A oxidase 1 and carnitine palmitoyltransferaser 1 expression in HK-2 cells. Importantly, blockade of Akt/mTOR signaling pathway with LY294002, a specific PI3K inhibitor, replicated these effects of thioredoxin-interacting protein silencing. Taken together, these data suggest that thioredoxin-interacting protein deficiency alleviates diabetic renal lipid accumulation through regulation of Akt/mTOR pathway, thioredoxin-interacting protein may be a potential therapeutic target for diabetic kidney disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Nefropatías Diabéticas/metabolismo , Metabolismo de los Lípidos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Tiorredoxinas/metabolismo , Acetil-CoA Carboxilasa/genética , Acetil-CoA Carboxilasa/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Relación Dosis-Respuesta a Droga , Acido Graso Sintasa Tipo I/genética , Acido Graso Sintasa Tipo I/metabolismo , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Glucosa/farmacología , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , PPAR alfa/metabolismo , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Tiorredoxinas/genética
14.
Am J Physiol Renal Physiol ; 310(6): F547-59, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26719366

RESUMEN

Oxidative stress is implicated in the pathogenesis of diabetic kidney injury. SS-31 is a mitochondria-targeted tetrapeptide that can scavenge reactive oxygen species (ROS). Here, we investigated the effect and molecular mechanism of mitochondria-targeted antioxidant peptide SS-31 on injuries in diabetic kidneys and mouse mesangial cells (MMCs) exposed to high-glucose (HG) ambience. CD-1 mice underwent uninephrectomy and streptozotocin treatment prior to receiving daily intraperitoneal injection of SS-31 for 8 wk. The diabetic mice treated with SS-31 had alleviated proteinuria, urinary 8-hydroxy-2-deoxyguanosine level, glomerular hypertrophy, and accumulation of renal fibronectin and collagen IV. SS-31 attenuated renal cell apoptosis and expression of Bax and reversed the expression of Bcl-2 in diabetic mice kidneys. Furthermore, SS-31 inhibited expression of transforming-growth factor (TGF)-ß1, Nox4, and thioredoxin-interacting protein (TXNIP), as well as activation of p38 MAPK and CREB and NADPH oxidase activity in diabetic kidneys. In vitro experiments using MMCs revealed that SS-31 inhibited HG-mediated ROS generation, apoptosis, expression of cleaved caspase-3, Bax/Bcl-2 ratio, and cytochrome c (cyt c) release from mitochondria. SS-31 normalized mitochondrial potential (ΔΨm) and ATP alterations, and inhibited the expression of TGF-ß1, Nox4, and TXNIP, as well as activation of p38 MAPK and CREB and NADPH oxidase activity in MMCs under HG conditions. SS-31 treatment also could reverse the reduction of thioredoxin (TRX) biologic activity and upregulate expression of thioredoxin 2 (TRX2) in MMCs under HG conditions. In conclusion, this study demonstrates a protective effect of SS-31 against HG-induced renal injury via an antioxidant mechanism in diabetic nephropathy.


Asunto(s)
Nefropatías Diabéticas/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Oligopéptidos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Proteínas Portadoras/metabolismo , Células Cultivadas , Colágeno Tipo IV/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Nefropatías Diabéticas/metabolismo , Evaluación Preclínica de Medicamentos , Fibronectinas/metabolismo , Glucosa , Masculino , Ratones , Mitocondrias/metabolismo , NADPH Oxidasa 4 , NADPH Oxidasas/metabolismo , Oligopéptidos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxinas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Biochem Biophys Res Commun ; 468(1-2): 281-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26505798

RESUMEN

The epithelial-to-mesenchymal transition (EMT) plays an important role in the progression of diabetic nephropathy. Our recent study showed that ROS mediated high glucose (HG)-induced EMT in renal tubular epithelial cells. CD36, a class-B scavenger receptor, has been reported to mediate the production of ROS in chronic kidney disease. In the present study, we examined the effect of inhibition of CD36 with CD36 siRNA or sulfosuccinimidyl-oleate (SSO), a CD36 antagonist, on HG-induced EMT in HK-2 cells. HG induced CD36 expression in a time-dependent manner in HK-2 cells. HG was shown to induce EMT at 72 h. This was blocked by knockdown of CD36 or treatment with SSO. Meanwhile, we also found that knockdown of CD36 or treatment with SSO inhibited HG-induced ROS generation, activation of ERK1/2 and Smad2, expression of TGF-ß1 and synthesis of fibronectin. These data suggest that inhibition of CD36 prevented HG-induced EMT in HK-2 cells, highlighting CD36 as a potential therapeutic target for diabetic nephropathy.


Asunto(s)
Antígenos CD36/metabolismo , Nefropatías Diabéticas/metabolismo , Células Epiteliales/citología , Transición Epitelial-Mesenquimal , Glucosa/metabolismo , Túbulos Renales/citología , Antígenos CD36/antagonistas & inhibidores , Línea Celular , Células Epiteliales/metabolismo , Fibronectinas/metabolismo , Humanos , Túbulos Renales/metabolismo , Sistema de Señalización de MAP Quinasas , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
16.
Drug Des Devel Ther ; 9: 5099-113, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26379423

RESUMEN

The dysregulation of cholesterol metabolism and inflammation plays a significant role in the progression of diabetic nephropathy (DN). Anthocyanins are polyphenols widely distributed in food and exert various biological effects including antioxidative, anti-inflammatory, and antihyperlipidemic effects. However, it remains unclear whether anthocyanins are associated with DN, and the mechanisms involved in the reciprocal regulation of inflammation and cholesterol efflux are yet to be elucidated. In this study, we evaluated the regulation of cholesterol metabolism and the anti-inflammatory effects exerted by anthocyanins (cyanidin-3-O-ß-glucoside chloride [C3G] or cyanidin chloride [Cy]) and investigated the underlying molecular mechanism of action using high-glucose (HG)-stimulated HK-2 cells. We found that anthocyanins enhanced cholesterol efflux and ABCA1 expression markedly in HK-2 cells. In addition, they increased peroxisome proliferator-activated receptor alpha (PPARα) and liver X receptor alpha (LXRα) expression and decreased the HG-induced expression of the proinflammatory cytokines intercellular adhesion molecule-1 (ICAM1), monocyte chemoattractant protein-1 (MCP1), and transforming growth factor-ß1 (TGFß1), as well as NFκB activation. Incubation with the PPARα-specific inhibitor GW6471 and LXRα shRNA attenuated the anthocyanin-mediated promotion of ABCA1 expression and cholesterol efflux, suggesting that anthocyanins activated PPARα-LXRα-ABCA1-dependent cholesterol efflux in HK-2 cells. Moreover, the knockout of LXRα abrogated the anti-inflammatory effect of anthocyanins, whereas the PPARα antagonist GW6471 does not have this effect. Further investigations revealed that LXRα might interfere with anthocyanin-induced decreased ICAM1, MCP1, and TGFß1 expression by reducing the nuclear translocation of NFκB. Collectively, these findings suggest that blocking cholesterol deposition and inhibiting the LXRα pathway-induced inflammatory response might be one of the main mechanisms by which anthocyanins exert their protective effects in DN.


Asunto(s)
Antocianinas/farmacología , Antiinflamatorios/farmacología , Colesterol/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Glucosa/metabolismo , Glucósidos/farmacología , Riñón/efectos de los fármacos , Nefritis/tratamiento farmacológico , Receptores Nucleares Huérfanos/agonistas , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Línea Celular , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Receptores X del Hígado , Nefritis/genética , Nefritis/metabolismo , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección
17.
Mol Cell Endocrinol ; 417: 27-35, 2015 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-26363223

RESUMEN

Renal tubular epithelial cells (RTEC) apoptosis, which plays a key role in the pathogenesis and progression of diabetic nephropathy (DN), is believed to be contributive to the hyperglycemia-induced kidney failure, though the exact mechanisms remain elusive. In this study, we investigated how inhibition of c-Src/p38 MAPK pathway would affect RTEC apoptosis. The c-Src inhibitor PP2 i.p. administered every other day for 8 weeks to diabetic db/db mice significantly reduced their kidney weights, daily urinary volumes, blood glucose, blood urea nitrogen, serum creatinine, triglyceride and urine albumin excretion, whereas deactivation of c-Src and p38 MAPK were also observed, along with decreases in both Bax/Bcl-2 ratio and cleaved caspase-3 level in the kidneys. In vitro, exposure of HK-2 cells (a human RTEC line), to high glucose (HG) promoted phosphorylation of c-Src and p38 MAPK, and subsequently, as revealed by western blotting, TUNEL assay and flow cytometry, increased cell death, which can be inhibited by PP2. Especially, a specific p38 MAPK inhibitor, SB203580, that both attenuated HG-induced c-Src activation and abrogated the expression of PPARγ and CHOP, also reduced apoptosis. Taken together, PP2 inhibits c-Src and therefore reduces apoptosis in RTEC, which at least in part, is due to suppressed p38 MAPK activation in diabetic kidney.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Túbulos Renales/citología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Pirimidinas/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Nefropatías Diabéticas/tratamiento farmacológico , Modelos Animales de Enfermedad , Esquema de Medicación , Células Epiteliales , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Masculino , Ratones , Pirimidinas/farmacología
18.
Drug Des Devel Ther ; 9: 4155-71, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26257513

RESUMEN

Transforming growth factor-ß1 (TGF-ß1) plays an important role in the pathogenesis and progression of chronic kidney disease. Connective tissue growth factor (CTGF) is a critical fibrogenic mediator of TGF-ß1. Mammalian sirtuin 1 (Sirt1) is reported to attenuate renal fibrosis by inhibiting the TGF-ß1 pathway. This study was designed to detect whether the delivery of CTGF siRNA in vivo directly ameliorates renal fibrosis. Furthermore, the relationship with Sirt1 underlying the protective effect of CTGF siRNA on interstitial fibrosis and apoptosis was explored. Here, we report that the expressions of CTGF and TGF-ß1 were increased while Sirt1 expression and activity were both dramatically decreased in mouse kidneys with unilateral ureteral obstruction. Recombinant human TGF-ß1 treatment in HK-2 cells increased CTGF levels and remarkably decreased Sirt1 levels and was accompanied by apoptosis and release of fibrosis-related factors. Recombinant human CTGF stimulation also directly induced apoptosis and fibrosis. The CTGF siRNA plasmid ameliorated tubular cell apoptosis and tubulointerstitial fibrosis, but did not affect Sirt1 expression and activity both in vivo and in vitro. Furthermore, overexpression of Sirt1 abolished TGF-ß1-induced cell apoptosis and fibrosis, while Sirt1 overexpression suppressed CTGF expression via stimulation by TGF-ß1. This study provides evidence that treatment strategies involving the delivery of siRNA targeting potentially therapeutic transgenes may be efficacious. Our results suggest that the decrease in Sirt1 is associated with the upregulated expression of CTGF in renal fibrosis, and may aid in the design of new therapies for the prevention of renal fibrosis.


Asunto(s)
Apoptosis , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Enfermedades Renales/prevención & control , Túbulos Renales/enzimología , ARN Interferente Pequeño/metabolismo , Tratamiento con ARN de Interferencia , Sirtuina 1/metabolismo , Obstrucción Ureteral/terapia , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Fibrosis , Regulación de la Expresión Génica , Humanos , Enfermedades Renales/enzimología , Enfermedades Renales/genética , Enfermedades Renales/patología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Masculino , Ratones , ARN Interferente Pequeño/genética , Transducción de Señal , Sirtuina 1/genética , Transfección , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/genética , Obstrucción Ureteral/patología
19.
Cell Signal ; 25(12): 2788-96, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24041652

RESUMEN

Epithelial to mesenchymal transition (EMT) of tubular cells contributes to the renal accumulation of matrix protein that is associated with diabetic nephropathy. Both high glucose and transforming growth factor-ß (TGF-ß) are able to induce EMT in cell culture. In this study, we examined the role of the thioredoxin-interacting protein (TXNIP) on EMT induced by high glucose or TGF-ß1 in HK-2 cells. EMT was assessed by the expression of α-smooth muscle actin (α-SMA) and E-cadherin and the induction of a myofibroblastic phenotype. High glucose (30mM) was shown to induce EMT at 72h. This was blocked by knockdown of TXNIP or antioxidant NAC. Meanwhile, we also found that knockdown of TXNIP or antioxidant NAC inhibited high glucose-induced generation of reactive oxygen species (ROS), phosphorylation of p38 MAPK and ERK1/2 and expression of TGF-ß1. HK-2 cells that were exposed to TGF-ß1 (4ng/ml) also underwent EMT. The expression of TXNIP gene and protein was increased in HK-2 cells treated with TGF-ß1. Transfection with TXNIP shRNA was able to attenuate TGF-ß1 induced-EMT. These results suggested that knockdown of TXNIP antagonized high glucose-induced EMT by inhibiting ROS production, activation of p38 MAPK and ERK1/2, and expression of TGF-ß1, highlighting TXNIP as a potential therapy target for diabetic nephropathy.


Asunto(s)
Proteínas Portadoras/genética , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Túbulos Renales/citología , Proteínas Portadoras/metabolismo , Células Epiteliales/citología , Humanos , Sistema de Señalización de MAP Quinasas , Factor de Crecimiento Transformador beta/metabolismo
20.
J Cell Biochem ; 114(5): 1029-38, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23129176

RESUMEN

Recent studies have shown that Notch pathway plays a key role in the pathogenesis of diabetic nephropathy (DN), however, the exact mechanisms remain elusive. Here we demonstrated that high glucose (HG) upregulated Notch pathway in podocytes accompanied with the alteration of Bcl-2 and p53 pathways, subsequently leading to podocytes apoptosis. Inhibition of Notch pathway by chemical inhibitor or specific short hairpin RNA (shRNA) vector in podocytes prevented Bcl-2- and p53-dependent cell apoptosis. These findings suggest that Notch pathway mediates HG-induced podocytes apoptosis via Bcl-2 and p53 pathways.


Asunto(s)
Apoptosis/efectos de los fármacos , Glucosa/farmacología , Podocitos/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Técnicas de Silenciamiento del Gen , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Inhibidores de Proteasas/farmacología , ARN Interferente Pequeño/metabolismo , Proteínas Serrate-Jagged , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA