Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Heliyon ; 10(3): e24983, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38318047

RESUMO

Renal fibrosis is a pathological feature of chronic kidney disease (CKD), progressing toward end-stage kidney disease (ESKD). The aim of this study is to investigate the therapeutic potential of altenusin, a farnesoid X receptor (FXR) agonist derived from fungi, on renal fibrosis. The effect of altenusin was determined (i) in vitro using the transforming growth factor ß1 (TGF-ß1)-induced epithelial to mesenchymal transition (EMT) of human renal proximal tubular cells and (ii) in vivo using mouse unilateral ureteral obstruction (UUO). The findings revealed that incubation of 10 ng/ml TGF-ß1 promotes morphological change in RPTEC/TERT1 cells, a human renal proximal tubular cell line, from epithelial to fibroblast-like cells. TGF-ß1 markedly increased EMT markers namely α-smooth muscle actin (α-SMA), fibronectin, and matrix metalloproteinase 9 (MMP-9), while decreased the epithelial marker E-cadherin. Co-incubation TGF-ß1 with altenusin preserved the epithelial characteristics of the renal epithelial cells by antagonizing TGF-ß/Smad signaling pathway, specifically a decreased phosphorylation of Smad2/3 with an increased level of Smad7. Interestingly, the antagonizing effect of altenusin does not require FXR activation. Moreover, altenusin could reverse TGF-ß1-induced fibroblast-like cells to epithelial-like cells. Treatment on UUO mice with 30 mg/kg altenusin significantly reduced the expression of α-SMA, fibronectin, and collagen type 1A1 (COL1A1). The reduction in the renal fibrosis markers is correlated with the decreased phosphorylation of Smad2/3 levels but does not improve E-cadherin protein expression. Collectively, altenusin reduces EMT in human renal proximal tubular cells and renal fibrosis by antagonizing the TGF-ß/Smad signaling pathway.

2.
EJIFCC ; 34(2): 142-152, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37455841

RESUMO

Background: Breath analyser tests are used worldwide to obtain proof of alcohol intoxication and often used in the conviction of traffic violators. These tests are conducted to quickly and painlessly determine the existing concentration of alcohol in arterial blood by measuring the amount of ethanol in exhaled breath, which can be identified with an electrochemical sensor.At present, the calibration and maintenance of analysers used for these tests are typically performed regularly but lack quality control. Consequently, test results may not be accurate because of calibration deterioration.The aim of this study was to develop and evaluate the uncertainty of control materials used in breath-alcohol testing at the Bangkok Metropolitan Police Station. Material and methods: Ethyl alcohol (99.99%; Certified Reference Material grade) diluted at three different concentrations was kept under design conditions. The concentrations were 28, 67, and 134 mg/dL, determined by performing headspace gas chromatography, and the uncertainty was set as ±1.3925, ±2.8736, and ±1.8231 mg/dL (±4.97%, ±4.29%, and ±2.72% for the concentrations, respectively), as per ISO Guide 35:2017. Results: The total error percentages of the developed control materials were 4.97%, 4.29%, and 2.72% for concentrations of 28, 67, and 134 mg/dL, respectively. Each concentration of the materials was tested by using measurements from 70 breath-alcohol analysers belonging to the Bangkok Metropolitan Police Station. Conclusion: These control materials are applicable to quality assurance and standards tests and may help to ensure the accuracy of breath-alcohol testing in the future.

3.
Int J Mol Sci ; 23(8)2022 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-35457146

RESUMO

Renal cyst expansion in polycystic kidney disease (PKD) involves abnormalities in both cyst-lining-cell proliferation and fluid accumulation. Suppression of these processes may retard the progression of PKD. Evidence suggests that the activation of 5' AMP-activated protein kinase (AMPK) inhibits cystic fibrosis transmembrane conductance regulator (CFTR)-mediated chloride secretion, leading to reduced progression of PKD. Here we investigated the pharmacological effects of panduratin A, a bioactive compound known as an AMPK activator, on CFTR-mediated chloride secretion and renal cyst development using in vitro and animal models of PKD. We demonstrated that AMPK was activated in immortalized normal renal cells and autosomal dominant polycystic kidney disease (ADPKD) cells following treatment with panduratin A. Treatment with panduratin A reduced the number of renal cyst colonies corresponding with a decrease in cell proliferation and phosphorylated p70/S6K, a downstream target of mTOR signaling. Additionally, panduratin A slowed cyst expansion via inhibition of the protein expression and transport function of CFTR. In heterozygous Han:Sprague-Dawley (Cy/+) rats, an animal model of PKD, intraperitoneal administration of panduratin A (25 mg/kg BW) for 5 weeks significantly decreased the kidney weight per body weight ratios and the cystic index. Panduratin A also reduced collagen deposition in renal tissue. Intraperitoneal administration of panduratin A caused abdominal bleeding and reduced body weight. However, 25 mg/kg BW of panduratin A via oral administration in the PCK rats, another non-orthologous PKD model, showed a significant decrease in the cystic index without severe adverse effects, indicating that the route of administration is critical in preventing adverse effects while still slowing disease progression. These findings reveal that panduratin A might hold therapeutic properties for the treatment of PKD.


Assuntos
Cistos , Doenças Renais Policísticas , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Peso Corporal , Proliferação de Células , Chalconas , Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Rim/metabolismo , Masculino , Doenças Renais Policísticas/tratamento farmacológico , Doenças Renais Policísticas/metabolismo , Ratos , Ratos Sprague-Dawley
4.
J Pharmacol Sci ; 148(4): 369-376, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35300812

RESUMO

Cystic fibrosis transmembrane conductance regulator (CFTR) plays crucial role in renal cyst expansion via increase in fluid accumulation. Inhibition of CFTR has been proposed to retard cyst development and enlargement in polycystic kidney disease (PKD). Pinostrobin, a bioactive natural flavonoid, possesses several pharmacological effects. The present study investigated pharmacological effects of pinostrobin on CFTR-mediated Cl- secretion and renal cyst expansion in in vitro and in vivo models. Pinostrobin (10 and 50 µM) reduced number of MDCK cell-derived cyst colonies and inhibited cyst expansion via inhibition of cell proliferation and CFTR-mediated Cl- secretion. The inhibitory effect of pinostrobin was not due to the decrease in cell viability and activity of Na+-K+-ATPase. We also investigated the natural analogue pinocembrin as well as the synthetic analogue pinostrobin oxime. Both pinocembrin and pinostrobin oxime did not reduce CFTR-mediated Cl- secretion. In PKD rats, oral administration of pinostrobin (40 mg/kg/day) exhibited a decreasing in cystic area compared to vehicle-treated rats. Pinostrobin treatment inhibited renal expression of CFTR protein in PKD rats. Our findings highlighted the potential therapeutic application of pinostrobin in PKD.


Assuntos
Cistos , Flavanonas , Rim , Doenças Renais Policísticas , Animais , Proliferação de Células , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Cães , Flavanonas/farmacologia , Flavanonas/uso terapêutico , Rim/efeitos dos fármacos , Rim/metabolismo , Células Madin Darby de Rim Canino , Doenças Renais Policísticas/tratamento farmacológico , Doenças Renais Policísticas/metabolismo , Ratos
5.
Molecules ; 26(21)2021 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-34771049

RESUMO

BACKGROUND: Panduratin A is a bioactive cyclohexanyl chalcone exhibiting several pharmacological activities, such as anti-inflammatory, anti-oxidative, and anti-cancer activities. Recently, the nephroprotective effect of panduratin A in cisplatin (CDDP) treatment was revealed. The present study examined the potential of certain compounds derived from panduratin A to protect against CDDP-induced nephrotoxicity. METHODS: Three derivatives of panduratin A (DD-217, DD-218, and DD-219) were semi-synthesized from panduratin A. We investigated the effects and corresponding mechanisms of the derivatives of panduratin A for preventing nephrotoxicity of CDDP in both immortalized human renal proximal tubular cells (RPTEC/TERT1 cells) and mice. RESULTS: Treating the cell with 10 µM panduratin A significantly reduced the viability of RPTEC/TERT1 cells compared to control (panduratin A: 72% ± 4.85%). Interestingly, DD-217, DD-218, and DD-219 at the same concentration did not significantly affect cell viability (92% ± 8.44%, 90% ± 7.50%, and 87 ± 5.2%, respectively). Among those derivatives, DD-218 exhibited the most protective effect against CDDP-induced renal proximal tubular cell apoptosis (control: 57% ± 1.23%; DD-218: 19% ± 10.14%; DD-219: 33% ± 14.06%). The cytoprotective effect of DD-218 was mediated via decreases in CDDP-induced mitochondria dysfunction, intracellular reactive oxygen species (ROS) generation, activation of ERK1/2, and cleaved-caspase 3 and 7. In addition, DD-218 attenuated CDDP-induced nephrotoxicity by a decrease in renal injury and improved in renal dysfunction in C57BL/6 mice. Importantly, DD-218 did not attenuate the anti-cancer efficacy of CDDP in non-small-cell lung cancer cells or colon cancer cells. CONCLUSIONS: This finding suggests that DD-218, a derivative of panduratin A, holds promise as an adjuvant therapy in patients receiving CDDP.


Assuntos
Chalconas/farmacologia , Cisplatino/efeitos adversos , Células Epiteliais/efeitos dos fármacos , Túbulos Renais Proximais/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Chalconas/síntese química , Chalconas/química , Técnicas de Química Sintética , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estrutura Molecular , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/síntese química , Substâncias Protetoras/química , Transporte Proteico/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
6.
Biol Pharm Bull ; 44(6): 830-837, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34078815

RESUMO

BACKGROUND: Cisplatin is an effective chemotherapy but its main side effect, acute kidney injury, limits its use. Panduratin A, a bioactive compound extracted from Boesenbergia rotunda, shows several biological activities such as anti-oxidative effects. The present study investigated the nephroprotective effect of panduratin A on cisplatin-induced renal injury. METHODS: We investigated the effect of panduratin A on the toxicity of cisplatin in both mice and human renal cell cultures using RPTEC/TERT1 cells. RESULTS: The results demonstrated that panduratin A ameliorates cisplatin-induced renal toxicity in both mice and RPTEC/TERT1 cells by reducing apoptosis. Mice treated with a single intraperitoneal (i.p.) injection of cisplatin (20 mg/kg body weight (BW)) exhibited renal tubule injury and impaired kidney function as shown by histological examination and increased serum creatinine. Co-administration of panduratin A (50 mg/kg BW) orally improved kidney function and ameliorated renal tubule injury of cisplatin by inhibiting activation of extracellular signal-regulated kinase (ERK)1/2 and caspase 3. In human renal proximal tubular cells, cisplatin induced cell apoptosis by activating pro-apoptotic proteins (ERK1/2 and caspase 3), and reducing the anti-apoptotic protein (Bcl-2). These effects were significantly ameliorated by co-treatment with panduratin A. Interestingly, panduratin A did not alter intracellular accumulation of cisplatin. It did not alter the anti-cancer efficacy of cisplatin in either human colon or non-small cell lung cancer cell lines. CONCLUSIONS: The present study highlights panduratin A has a potential protective effect on cisplatin's nephrotoxicity.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Antineoplásicos/efeitos adversos , Chalconas/uso terapêutico , Cisplatino/efeitos adversos , Substâncias Protetoras/uso terapêutico , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Apoptose , Linhagem Celular , Chalconas/farmacologia , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Túbulos Renais Proximais/citologia , Masculino , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia
7.
Toxicol Lett ; 236(3): 145-53, 2015 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-25980575

RESUMO

Liver X receptors (LXRs) including LXRα and LXRß are members of the nuclear receptor superfamily of ligand-activated transcription factors, which are expressed in high metabolic organs such as the liver, kidney, and adipose tissue. LXRs have been shown to act as antioxidants and anti-inflammatory in several organs. The present study investigated the effects of LXR activation on cadmium-induced cell death in renal proximal tubular cells. Treating human renal proximal tubular cells, HK-2 cells, with 20µM CdCl2 for 24h led to cell death via apoptosis but not necrosis. Interestingly, pretreating HK-2 cells with T0901317, a LXR agonist, significantly inhibited the apoptotic cell death induced by CdCl2. The protective effect of T0901317 was eliminated by incubation with fenofibrate, a LXR antagonist, indicating that the effect of T0901317 on cadmium-induced apoptotic cell death was mediated by LXR activation. In addition, the effect of CdCl2 was attenuated by a reactive oxygen species (ROS) scavenger, N-acetyl-l-cysteine (NAC). An increase in ROS induced by CdCl2 was mediated by inhibition of catalase but not superoxide dismutase (SOD) which was attenuated by T0901317. Western blot analysis revealed that CdCl2 stimulated expression of c-jun N-terminal kinase (JNK) phosphorylation and the stimulation were inhibited by NAC, indicating the induction of JNK phosphorylation was stimulated following ROS production. Moreover, the increases of ROS and JNK phosphorylation induced by CdCl2 were attenuated by LXR activation. This study provides the first evidence to show LXR activation reduces cadmium-induced apoptotic cell death of human renal proximal tubular cells by inhibition of ROS production and JNK activation.


Assuntos
Apoptose/efeitos dos fármacos , Cloreto de Cádmio/toxicidade , Túbulos Renais Proximais/citologia , Receptores Nucleares Órfãos/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Receptores X do Fígado , MAP Quinase Quinase 4/metabolismo , Espécies Reativas de Oxigênio/metabolismo
8.
Am J Physiol Renal Physiol ; 303(12): F1610-6, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23077096

RESUMO

Liver X receptors (LXRs) are nuclear receptors that regulate cholesterol, fatty acid, and glucose metabolism in various tissues. However, the renal action of LXRs is not well understood. Here we investigated the effects of LXR-activating ligands on modulation of epithelial sodium channel (ENaC)-mediated sodium transport in collecting duct cells. Exposure of the M1 cells to the synthetic LXR agonists T0901317 and GW3965 or the natural ligand 22R-hydroxycholesterol for 24 h decreased amiloride-sensitive sodium transport, corresponding with an increase of transepithelial resistance. The inhibition of amiloride-sensitive sodium transport after incubation with T0901317 or GW3965 was not mediated by a reduction of Na(+)/K(+)-ATPase-mediated basolateral sodium transport. On the other hand, T0901317 and GW3965 decreased mRNA abundance and membrane expression of ENaC. Preincubation the monolayer with GW3965 attenuated aldosterone-induced stimulation sodium transport. In primary cultures of collecting duct cells, T0901317 and GW3965 similarly inhibited ENaC transport function as in M1 cells. This is the first evidence showing LXR-activating ligands modulate ENaC-mediated sodium transport in collecting duct cells. These results suggest that LXRs may represent a novel therapeutic target for treatment of conditions with dysregulation of ENaC such as hypertension.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Coletores/metabolismo , Receptores Nucleares Órfãos/agonistas , Receptores Nucleares Órfãos/metabolismo , Sódio/metabolismo , Animais , Anticolesterolemiantes/farmacologia , Benzoatos/farmacologia , Benzilaminas/farmacologia , Transporte Biológico/efeitos dos fármacos , Células Cultivadas , Hidrocarbonetos Fluorados/farmacologia , Hidroxicolesteróis/farmacologia , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/efeitos dos fármacos , Ligantes , Receptores X do Fígado , Camundongos , ATPase Trocadora de Sódio-Potássio/metabolismo , Sulfonamidas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...