Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Eur J Drug Metab Pharmacokinet ; 49(3): 343-353, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38472634

RESUMO

BACKGROUND AND OBJECTIVE: In vitro glucuronidation of 17ß-estradiol (estradiol) is often performed to assess the role of uridine 5'-diphospho-glucuronosyltransferase 1A1 (UGT1A1) in xenobiotic/drug metabolism. The objective of this study was to determine the effects of four commonly used organic solvents [i.e., dimethyl sulfoxide (DMSO), methanol, ethanol, and acetonitrile] on the glucuronidation kinetics of estradiol, which can be glucuronidated at C3 and C17 positions. METHODS: The impacts of organic solvents on estradiol glucuronidation were determined by using expressed UGT enzymes and liver microsomes from both human and animals. RESULTS: In human liver microsomes (HLM), methanol, ethanol, and acetonitrile significantly altered estradiol glucuronidation kinetics with increased Vmax (up to 2.6-fold) and CLmax (up to 2.8-fold) values. Altered estradiol glucuronidation in HLM was deduced to be attributed to the enhanced metabolic activities of UGT1A1 and UGT2B7, whose activities differ at the two glucuronidation positions. The effects of organic solvents on estradiol glucuronidation were glucuronidation position-, isozyme-, and solvent-specific. Furthermore, both ethanol and acetonitrile have a greater tendency to modify the glucuronidation activity of estradiol in animal liver microsomes. CONCLUSION: Organic solvents such as methanol, ethanol, and acetonitrile showed great potential in adjusting the glucuronidation of estradiol. DMSO is the most suitable solvent due to its minimal influence on estradiol glucuronidation. Researchers should be cautious in selecting appropriate solvents to get accurate results when assessing the metabolism of a new chemical entity.


Assuntos
Dimetil Sulfóxido , Estradiol , Etanol , Glucuronídeos , Glucuronosiltransferase , Microssomos Hepáticos , Solventes , Microssomos Hepáticos/metabolismo , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Estradiol/metabolismo , Estradiol/farmacologia , Glucuronosiltransferase/metabolismo , Humanos , Solventes/farmacologia , Animais , Cinética , Etanol/metabolismo , Etanol/farmacologia , Glucuronídeos/metabolismo , Dimetil Sulfóxido/farmacologia , Metanol/farmacologia , Metanol/metabolismo , Acetonitrilas/farmacologia , Acetonitrilas/metabolismo
2.
J Cell Biochem ; 125(4): e30541, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38372186

RESUMO

Epithelial cells (ECs) have been proposed to contribute to myofibroblasts or fibroblasts through epithelial-mesenchymal transition (EMT) during renal fibrosis. However, since EMT may occur dynamically, transiently, and reversibly during kidney fibrosis, conventional lineage tracing based on Cre-loxP recombination in renal ECs could hardly capture the transient EMT activity, yielding inconsistent results. Moreover, previous EMT research has primarily focused on renal proximal tubule ECs, with few reports of distal tubules and collecting ducts. Here, we generated dual recombinases-mediated genetic lineage tracing systems for continuous monitoring of transient mesenchymal gene expression in E-cadherin+ and EpCAM+ ECs of distal tubules and collecting ducts during renal fibrosis. Activation of key EMT-inducing transcription factor (EMT-TF) Zeb1 and mesenchymal markers αSMA, vimentin, and N-cadherin, were investigated following unilateral ureteral obstruction (UUO). Our data revealed that E-cadherin+ and EpCAM+ ECs did not transdifferentiate into myofibroblasts, nor transiently expressed these mesenchymal genes during renal fibrosis. In contrast, in vitro a large amount of cultured renal ECs upregulated mesenchymal genes in response to TGF-ß, a major inducer of EMT.


Assuntos
Transição Epitelial-Mesenquimal , Nefropatias , Humanos , Molécula de Adesão da Célula Epitelial/genética , Molécula de Adesão da Célula Epitelial/metabolismo , Fibrose , Nefropatias/metabolismo , Células Epiteliais/metabolismo , Caderinas/genética , Caderinas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
3.
Mol Med Rep ; 13(2): 1427-33, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26648565

RESUMO

NogoA is a myelin­associated protein, which is important in the inhibition of axonal fiber growth and in regeneration following injury of the mammalian central nervous system. A previous study suggested that NogoA may be key in the process of Parkinson's disease (PD), which is the second most common chronic neurodegenerative disorder worldwide. The regulatory mechanism underlying the effect of NogoA on the process of PD remains to be fully elucidated. The present study aimed to investigate the effect and underlying mechanism of NogoA on cellular viability, apoptosis and autophagy induced by 1-methyl-4-phenylpyridinium (MPP+) in PC12 cells, a commonly used in vitro PD model. PC12 cells were treated with 1 mM MPP+ for 24 h and the cells were harvested for western blotting. The results demonstrated that the protien expression levels of NogoA were increased in the PC12 cells treated with MPP+. Subsequently, NogoA small interfering RNA was synthesized and transfected into PC12 cells to silence the expression of NogoA. NogoA knockdown significantly reduced the MPP+­induced decrease in cell viability and apoptosis, detected using a cell counting kit­8 and flow cytometric analysis, respectively. Interference in the expression of NogoA increased the MPP+­induced decrease in mitochondrial membrane potential, determined quantitatively by flow cytometry using JC-1 dye, and the protein levels of Beclin­1. In addition, MPP+ treatment activated the mammalian target of rapamycin (mTOR)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. Knockdown of NogoA significantly inhibited the expression levels of mTOR and STAT3. Furthermore, overexpression of NogoA had similar neurotoxic effects on the PC12 cells as MPP+ treatment. Treatment with rapamycin, an inhibitor of the mTOR/STAT3 signaling pathway had a similar effect to that of NogoA knockdown in the MPP+­treated PC12 cells. Taken together, the results from the present study demonstrated that NogoA may regulate MPP+­induced neurotoxicity in PC12 cells via the mTOR/STAT3 signaling pathway and provided an explanation regarding the regulatory mechanism of NogoA on the process of PD.


Assuntos
1-Metil-4-fenilpiridínio/toxicidade , Técnicas de Silenciamento de Genes , Proteínas da Mielina/metabolismo , Neurotoxinas/toxicidade , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/efeitos dos fármacos , Proteína Beclina-1 , Sobrevivência Celular , Proteínas Ligadas por GPI/metabolismo , Inativação Gênica/efeitos dos fármacos , Proteínas Nogo , Receptor Nogo 1 , Células PC12 , Ratos , Receptores de Superfície Celular/metabolismo , Sirolimo/farmacologia
4.
Biomed Res Int ; 2015: 817914, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26583134

RESUMO

Parkinson's disease (PD) is a common degenerative disease that lacks efficient treatment. Myelin-associated neurite outgrowth inhibitor A (Nogo-A) is relevant with inhibition of nerve regeneration and may play vital role in pathogenesis of PD. The study aimed to establish the shRNA expression plasmids of Nogo-A gene and explore the regulatory effects of Nogo-A silencing on the expression of inflammation factor tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) as well as tyrosine hydroxylase (TH) in lipopolysaccharide- (LPS-) stimulated rat PC12 cells. The results showed that both mRNA and protein levels of Nogo-A in pGenesil-nogoA-shRNA group were downregulated. The viabilities of PC12 cells decreased with increase of LPS concentrations. LPS significantly increased the supernatant TNF-alpha and IL-6 concentrations and reduced TH protein expression in PC12 cells, while silencing Nogo-A could block these effects. These results suggested that LPS can activate PC12 cells to secrete inflammatory cytokines and lower the TH expression, which can be regulated by Nogo-A gene silencing. Nogo-A silencing might provide new ideas for PD treatment in the future.


Assuntos
Inflamação/genética , Interleucina-6/metabolismo , Proteínas da Mielina/genética , Doença de Parkinson/genética , Fator de Necrose Tumoral alfa/metabolismo , Tirosina 3-Mono-Oxigenase/biossíntese , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Humanos , Inflamação/induzido quimicamente , Inflamação/patologia , Interleucina-6/genética , Lipopolissacarídeos/toxicidade , Proteínas da Mielina/antagonistas & inibidores , Regeneração Nervosa/genética , Proteínas Nogo , Células PC12 , Doença de Parkinson/patologia , Doença de Parkinson/terapia , RNA Mensageiro/biossíntese , Ratos , Fator de Necrose Tumoral alfa/genética , Tirosina 3-Mono-Oxigenase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA