The Journal of Toxicological Sciences
Online ISSN : 1880-3989
Print ISSN : 0388-1350
ISSN-L : 0388-1350
Volume 47, Issue 2
Displaying 1-4 of 4 articles from this issue
Original Article
  • Dedong Li, Jian Sun, Mingdong Yu, Ying Wang, Yuechun Lu, Bo Li
    2022 Volume 47 Issue 2 Pages 51-60
    Published: 2022
    Released on J-STAGE: February 01, 2022
    JOURNAL FREE ACCESS FULL-TEXT HTML

    MiR-128 is highly expressed in the central nervous system and may regulate the directional differentiation of bone marrow stromal stem cells into nerve cells. However, its role and mechanism in sevoflurane-induced progressive neurotoxicity in rats are rarely reported. Therefore, this study aims to explore the protection of miR-128-3p on sevoflurane-induced neurotoxicity. Hippocampal neurons were isolated and sevoflurane was used to treat the cells. Cell counting kit-8 (CCK-8) was used to detect cell viability. Immunofluorescence was used to detect enrichment of GFAP or βIII tubulin to identify nerve cells. Dual luciferase assay was used to identify the targeted binding relationship between miR-128-3p and NOVA1. The effect of miR-128-3p and sevoflurane on cells regarding apoptosis was detected by flow cytometry. The expression of apoptosis-related protein and oxidative stress-related proteins were detected by western blot. Enzyme-linked immuno-sorbent assay (ELISA) was used to measure inflammatory cytokine levels. Hippocampal neurons’ cell viability was significantly decreased by treatment with sevoflurane. MiR-128-3p was down-regulated after sevoflurane treatment in cells. Overexpressed miR-128-3p partially reversed the role of sevoflurane treatment in triggering cell apoptosis, enhancing the expression of Bax and cleaved caspase-3 and inhibiting Bcl-2 expression obviously. Overexpressed miR-128-3p partially reversed the role of sevoflurane treatment in promoting the expression of NOX1and NOX4, and inflammatory cytokine levels by targeting with NOVA1. MiR-128-3p might be a potential therapeutic target for the prevention or treatment of sevoflurane-induced neurotoxicity by targeting with NOVA1.

Original Article
  • Qinqin Zhang, Zhilan Wang, Jinfei Zhu, Zhili Peng, Cheng Tang
    2022 Volume 47 Issue 2 Pages 61-69
    Published: 2022
    Released on J-STAGE: February 01, 2022
    JOURNAL FREE ACCESS FULL-TEXT HTML

    Acute lung injury (ALI) is mainly mediated by the damage of pulmonary microvascular endothelial cells (PMVECs). LPS is one of the pathogenic factors leading to microcirculatory abnormalities of ALI. Ferulic acid (FA) exhibits therapeutic effects against various diseases. During lipopolysaccharide-induced acute respiratory distress syndrome, FA, when given beforehand, could depress inflammation and oxidative stress. However, the concrete role and underlying mechanism of FA in ALI have not been well characterized. Ten μg/mL Lipopolysaccharide (LPS) was used to treat rat PMVECs for 24 hr. qRT-PCR was used to detect the level of miR-17 and phosphatase and tensin homolog deleted on chromosome ten (PTEN). Western blot was used to analyze the associated proteins in the PI3K/Akt pathway, and the apoptosis-related proteins. Flow cytometric analysis was performed to detect the apoptosis of PMVECs. MTT assay was constructed to detect the cell viability. Luciferase assay was conducted to detect the target gene of miR-17 and PTEN. A cell model for in vitro studying the role of FA in ALI was established using PMVECs. Our data demonstrate that FA up-regulates miR-17 and declines apoptosis induced by LPS. FA inhibits apoptosis mediated by up-regulating miR-17. Furthermore, we found miR-17 targeted PTEN negatively. FA inhibits cleaved caspase-3 and Bax expression through the PI3K/Akt pathway mediated by up-regulating miR-17. Over-expression of PTEN could contribute to the similar expression trend of the PI3K/Akt signal pathway protein compared to miR-17 inhibitor transfected cells. FA inhibits PMVECs apoptosis induced by LPS via miR-17/PTEN to further regulate the activation of the PI3K/Akt pathway in ALI. We anticipate that our data will provoke additional studies for ALI clinical therapy.

Letter
  • Toshinori Hirai, Yoshihiko Morikawa, Noriko Sasaki, Hideo Kato, Daisuk ...
    2022 Volume 47 Issue 2 Pages 71-75
    Published: 2022
    Released on J-STAGE: February 01, 2022
    JOURNAL FREE ACCESS FULL-TEXT HTML
    Supplementary material

    This case involved a 27-year-old man with extreme obesity (body mass index 45.6 kg/m2) who had a history of fulminant hepatitis and living-donor liver transplantation at 11 years of age. He had been receiving oral sustained-release tacrolimus (TAC) 1.5 mg daily, and the trough concentration in the blood was below 2.0 ng/mL. He has an intrinsic cytochrome P450 3A5 (CYP3A5)*3/*3 (G/G) genotype and graft liver with CYP3A5*3 allele donated by his biological father. Additionally, there were no data on the phenotype of P-glycoprotein. He did not take medications, grapefruit, or St. John’s wort, which interact with CYP3A4 and P-glycoprotein. He intentionally took 30 mg of TAC and presented with symptoms of general malaise and poisoning. On the day of hospitalization (day 0), TAC was discontinued due to an elevated blood TAC concentration of > 60 ng/mL. Additionally, the blood TAC concentration exceeded 10 ng/mL for more than 3 days. He exhibited mild elevation of alanine aminotransferase, aspartate aminotransferase, and creatinine phosphokinase without apparent clinical symptoms. After discharge, blood TAC concentration decreased to 7.4 and 3.7 ng/mL on days 14 and 28, respectively, from the day of excessive TAC intake. Finally, the blood TAC concentration fell below 2.0 ng/mL on day 66. This case report showed that extreme obesity and the liver CYP3A5*3 allele delayed the elimination of TAC after excessive intake of the drug.

Original Article
  • Saori Watanabe-Matsumoto, Kikuo Yoshida, Yuriko Meiseki, Seiichi Ishid ...
    2022 Volume 47 Issue 2 Pages 77-87
    Published: 2022
    Released on J-STAGE: February 01, 2022
    JOURNAL FREE ACCESS FULL-TEXT HTML
    Supplementary material

    Although physiologically based kinetic (PBK) modeling is informative for the risk assessment of industrial chemicals, chemical-specific input values for partition coefficients and metabolic parameters, including Vmax and Km are mostly unavailable; however, in silico methods, such as quantitative structure-property relationship (QSPR) could fill the absence. To assess the PBK model validity using necessary toxicokinetic (TK) parameters predicted by QSPR, the PBK model of ethyl tert-butyl ether (ETBE) as a model substance was constructed, in which the values of the partition coefficients, Vmax, and Km of ETBE were predicted using those of the related chemicals previously reported in the literature, and toxicokinetics of inhaled ETBE were stochastically estimated using the Monte Carlo simulation. The calculated ETBE concentrations in venous blood were comparable to the measured values in humans, implying that the reproducibility of ETBE toxicokinetics in humans was established in this PBK model. The Monte Carlo simulation was used to conduct uncertainty and sensitivity analyses of the dose metrics in terms of maximum blood concentration (Cmax) and area under the blood concentration-time curve (AUC) and the estimated Cmax and AUC were highly and moderately reliable, respectively. Conclusively, the PBK model validity combined with in silico methods of QSPR was demonstrated in an ETBE model substance. QSPR-PBK modeling coupled with the Monte Carlo simulation is effective for estimating chemical toxicokinetics for which input values are unavailable and for evaluating the estimation validity.

feedback
Top