首页 > 最新文献

Human & experimental toxicology最新文献

英文 中文
Exosomes derived from M2 macrophages promote fibroblast autophagy to contribute to hypertrophic scar formation via CXCL2/CXCR7/mTOR pathway. 源自M2巨噬细胞的外泌体通过CXCL2/CXCR7/mTOR途径促进成纤维细胞自噬,从而促成肥厚性疤痕的形成。
Pub Date : 2024-01-01 DOI: 10.1177/09603271241303320
Min Shi, Lu Zhang, Fangfang Bi, Xiaohong Ma
<p><strong>Introduction: </strong>Abnormal activation of hypertrophic scar fibroblasts (HSF) plays an important role in the excessive fibrosis of hypertrophic scars (HS). However, the regulatory mechanism of HSF abnormal activation is not fully unclear. Early studies had shown that M2 macrophages were increased during scar formation. The aim of this study was to investigate the mechanism of M2 macrophage-derived exosomes (M2-EXOs) mediating HSF abnormal activation.</p><p><strong>Methods: </strong>The blood samples of 20 normal people and 20 HS patients were collected from Xi'an Hospital of Traditional Chinese Medicine, and the level of M2 macrophages in the blood was measured by flow cytometry. Subsequently, HSFs were co-cultured with M2-THP-1 for 48 h to analyze the effect of M2 macrophages on the function of HSFs <i>in vitro</i>. HSFs were treated with exogenous chemokine (C-X-C motif) ligand 2 (CXCL2) or anti-CXCL2 to analyze the effect of CXCL2 on HSFs function and autophagy. HSFs were treated with exogenous CXCL2 and/or anti-CXCR7, and CXCL2 and/or 3MA to explore the molecular mechanism of CXCL2-mediated HS. Finally, a mouse HS model was constructed, and the effect of M2-Exos on the growth of HS was explored by subcutaneous injection of CXCL2 or M2-Exos in the scar site <i>in vivo</i>.</p><p><strong>Results: </strong>We found that the proportion of M2 macrophages in the blood of HS patients increased. CXCL2-rich M2-EXOs promoted the abnormal proliferation, migration, and collagen deposition of HSFs <i>in vitro</i>. CXCL2 increased the level of p-mTOR in HSF and promoted the expression of autophagy proteins LC3II/I and Atg5 <i>in vitro</i>. Further results showed that CXCL2 activated autophagy through CXCR7/PI3K/mTOR signal transduction, thereby promoting collagen deposition and fibrosis <i>in vitro</i>. Autophagy inhibitor 3-MA reversed the effect of CXCL2 on HSFs <i>in vitro</i>. In addition, in the HS mouse model, after treatment with M2-EXOs or CXCL2 <i>in vivo</i>, the scar recovery time was significantly prolonged and the scar damage was aggravated.</p><p><strong>Discussion: </strong>These results suggest that the CXCL2/CXCR7/mTOR pathway may be a promising target for the treatment of HS. Abnormal activation of hypertrophic scar fibroblasts (HSFs) plays an important role in the excessive fibrosis of hypertrophic scars (HS). However, the regulatory mechanism of HSFs abnormal activation is not fully unclear. Early studies had shown that M2 macrophages were increased during scar formation. The aim of this study was to investigate the mechanism of M2 macrophage-derived exosomes (M2-EXOs) mediating HSFs abnormal activation. Here, we analyzed the proportion of M2 macrophages in total macrophages in the HS patient's blood, and we found that the proportion of M2 macrophages were elevated in the blood of HS patients. We found that C-X-C motif chemokine 2 (CXCL2)-rich M2-EXOs promoted abnormal proliferation, migration, and collagen deposition in
导言:肥厚性疤痕成纤维细胞(HSF)的异常激活在肥厚性疤痕(HS)的过度纤维化中起着重要作用。然而,HSF异常活化的调控机制尚不完全清楚。早期研究表明,M2 巨噬细胞在疤痕形成过程中会增加。本研究旨在探讨M2巨噬细胞衍生的外泌体(M2-EXOs)介导HSF异常激活的机制:方法:从西安市中医医院采集20名正常人和20名HS患者的血液样本,用流式细胞术检测血液中M2巨噬细胞的水平。随后,将 HSFs 与 M2-THP-1 共同培养 48 小时,分析 M2 巨噬细胞对体外 HSFs 功能的影响。用外源性趋化因子(C-X-C motif)配体 2(CXCL2)或抗 CXCL2 处理 HSFs,分析 CXCL2 对 HSFs 功能和自噬的影响。用外源性 CXCL2 和/或抗 CXCR7 以及 CXCL2 和/或 3MA 处理 HSFs,以探讨 CXCL2 介导 HS 的分子机制。最后,构建了小鼠 HS 模型,并通过在体内瘢痕部位皮下注射 CXCL2 或 M2-Exos 探讨了 M2-Exos 对 HS 生长的影响:结果:我们发现HS患者血液中的M2巨噬细胞比例增加。富含 CXCL2 的 M2-EXO 在体外促进 HSFs 的异常增殖、迁移和胶原沉积。CXCL2 提高了 HSF 中 p-mTOR 的水平,促进了体外自噬蛋白 LC3II/I 和 Atg5 的表达。进一步的结果表明,CXCL2通过CXCR7/PI3K/mTOR信号转导激活自噬,从而促进体外胶原沉积和纤维化。自噬抑制剂 3-MA 逆转了 CXCL2 对体外 HSFs 的影响。此外,在 HS 小鼠模型中,体内使用 M2-EXOs 或 CXCL2 治疗后,瘢痕恢复时间明显延长,瘢痕损伤加重:这些结果表明,CXCL2/CXCR7/mTOR通路可能是治疗HS的一个有前景的靶点。肥厚性疤痕成纤维细胞(HSFs)的异常活化在肥厚性疤痕(HS)的过度纤维化中起着重要作用。然而,HSFs异常活化的调控机制尚不完全清楚。早期研究表明,M2 巨噬细胞在疤痕形成过程中会增加。本研究旨在探究 M2 巨噬细胞衍生的外泌体(M2-EXOs)介导 HSFs 异常激活的机制。我们分析了 HS 患者血液中 M2 巨噬细胞占巨噬细胞总数的比例,发现 HS 患者血液中 M2 巨噬细胞比例升高。我们发现,富含 C-X-C motif 趋化因子 2(CXCL2)的 M2-EXO 可促进 HSFs 在体外异常增殖、迁移和胶原沉积。CXCL2 可提高 mTOR 蛋白的磷酸化水平,促进 HSF 体外自噬相关蛋白 LC3II/I 和 Atg5 的表达水平。CXCL2通过趋化因子(C-X-C基序)受体7(CXCR7)/PI3K/mTOR信号转导激活自噬,促进体外胶原沉积和纤维化。自噬抑制剂3-甲基腺嘌呤(3-MA)可逆转体外CXCL2对HSFs的影响。同时,在 HS 小鼠模型中,体内使用 M2-EXOs 或 CXCL2 处理后,瘢痕恢复时间明显延长,瘢痕损伤加重。这些结果表明,CXCL2/CXCR7/mTOR通路可能是治疗HS的一个有前景的靶点。
{"title":"Exosomes derived from M2 macrophages promote fibroblast autophagy to contribute to hypertrophic scar formation via CXCL2/CXCR7/mTOR pathway.","authors":"Min Shi, Lu Zhang, Fangfang Bi, Xiaohong Ma","doi":"10.1177/09603271241303320","DOIUrl":"10.1177/09603271241303320","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Introduction: &lt;/strong&gt;Abnormal activation of hypertrophic scar fibroblasts (HSF) plays an important role in the excessive fibrosis of hypertrophic scars (HS). However, the regulatory mechanism of HSF abnormal activation is not fully unclear. Early studies had shown that M2 macrophages were increased during scar formation. The aim of this study was to investigate the mechanism of M2 macrophage-derived exosomes (M2-EXOs) mediating HSF abnormal activation.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;The blood samples of 20 normal people and 20 HS patients were collected from Xi'an Hospital of Traditional Chinese Medicine, and the level of M2 macrophages in the blood was measured by flow cytometry. Subsequently, HSFs were co-cultured with M2-THP-1 for 48 h to analyze the effect of M2 macrophages on the function of HSFs &lt;i&gt;in vitro&lt;/i&gt;. HSFs were treated with exogenous chemokine (C-X-C motif) ligand 2 (CXCL2) or anti-CXCL2 to analyze the effect of CXCL2 on HSFs function and autophagy. HSFs were treated with exogenous CXCL2 and/or anti-CXCR7, and CXCL2 and/or 3MA to explore the molecular mechanism of CXCL2-mediated HS. Finally, a mouse HS model was constructed, and the effect of M2-Exos on the growth of HS was explored by subcutaneous injection of CXCL2 or M2-Exos in the scar site &lt;i&gt;in vivo&lt;/i&gt;.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;We found that the proportion of M2 macrophages in the blood of HS patients increased. CXCL2-rich M2-EXOs promoted the abnormal proliferation, migration, and collagen deposition of HSFs &lt;i&gt;in vitro&lt;/i&gt;. CXCL2 increased the level of p-mTOR in HSF and promoted the expression of autophagy proteins LC3II/I and Atg5 &lt;i&gt;in vitro&lt;/i&gt;. Further results showed that CXCL2 activated autophagy through CXCR7/PI3K/mTOR signal transduction, thereby promoting collagen deposition and fibrosis &lt;i&gt;in vitro&lt;/i&gt;. Autophagy inhibitor 3-MA reversed the effect of CXCL2 on HSFs &lt;i&gt;in vitro&lt;/i&gt;. In addition, in the HS mouse model, after treatment with M2-EXOs or CXCL2 &lt;i&gt;in vivo&lt;/i&gt;, the scar recovery time was significantly prolonged and the scar damage was aggravated.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Discussion: &lt;/strong&gt;These results suggest that the CXCL2/CXCR7/mTOR pathway may be a promising target for the treatment of HS. Abnormal activation of hypertrophic scar fibroblasts (HSFs) plays an important role in the excessive fibrosis of hypertrophic scars (HS). However, the regulatory mechanism of HSFs abnormal activation is not fully unclear. Early studies had shown that M2 macrophages were increased during scar formation. The aim of this study was to investigate the mechanism of M2 macrophage-derived exosomes (M2-EXOs) mediating HSFs abnormal activation. Here, we analyzed the proportion of M2 macrophages in total macrophages in the HS patient's blood, and we found that the proportion of M2 macrophages were elevated in the blood of HS patients. We found that C-X-C motif chemokine 2 (CXCL2)-rich M2-EXOs promoted abnormal proliferation, migration, and collagen deposition in ","PeriodicalId":94029,"journal":{"name":"Human & experimental toxicology","volume":"43 ","pages":"9603271241303320"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142669925","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Trimethylamine-N oxide enhances post-stroke depression progression via ROS-p38/MAPK signaling.
Pub Date : 2024-01-01 DOI: 10.1177/09603271241306396
Yikui Hu, Yujie Liu, Hua Wang, Xun Wang

Introduction: Post-stroke depression (PSD) is one of the common complications after stroke. Trimethylamine-N oxide (TMAO) has been reported to exacerbate brain injury after ischemic stroke, and its expression is positively related to the severity of depressive symptoms. This study was performed to investigate the role and mechanism of TMAO in PSD.

Methods: The classical stroke model was combined with spatial constraint stress to establish a PSD mouse model, and the effect of TMAO on the depression-like behavior of the PSD mouse model was evaluated using the tail suspension test and the forced swimming test. ELISA was employed to measure the concentration of TMAO and cortisol. RT-qPCR, Western blot, and Immunohistochemistry assay were implemented to determine mRNA and protein expression. Endothelial permeability was assessed using the fluorescein isothiocyanate-dextran permeability assay in vitro. Reactive oxygen species (ROS) level was examined with a ROS detection kit.

Results: TMAO dose-dependently exacerbated depression-like behavior, induced the activation of the p38/mitogen-activated protein kinase (MAPK) signaling pathway, downregulated the abundance of tight junction proteins, and resulted in the dysregulation of neurotrophic mediators in the PSD mouse model in vivo. Further, TMAO increased endothelial permeability and activated reactive oxygen species-p38/MAPK signaling. By introducing the p38/MAPK pathway inhibitor SB203580, TMAO was found to downregulate the expressions of tight junction proteins and promoted endothelial permeability via p38/MAPK signaling activation.

Discussion: By activating the ROS-P38/MAPK pathway, TMAO enhanced the permeability of the blood-brain barrier after stroke. These mechanisms resulted in the dysregulation of neurotrophic mediators, ultimately leading to PSD progression.

{"title":"Trimethylamine-N oxide enhances post-stroke depression progression via ROS-p38/MAPK signaling.","authors":"Yikui Hu, Yujie Liu, Hua Wang, Xun Wang","doi":"10.1177/09603271241306396","DOIUrl":"https://doi.org/10.1177/09603271241306396","url":null,"abstract":"<p><strong>Introduction: </strong>Post-stroke depression (PSD) is one of the common complications after stroke. Trimethylamine-N oxide (TMAO) has been reported to exacerbate brain injury after ischemic stroke, and its expression is positively related to the severity of depressive symptoms. This study was performed to investigate the role and mechanism of TMAO in PSD.</p><p><strong>Methods: </strong>The classical stroke model was combined with spatial constraint stress to establish a PSD mouse model, and the effect of TMAO on the depression-like behavior of the PSD mouse model was evaluated using the tail suspension test and the forced swimming test. ELISA was employed to measure the concentration of TMAO and cortisol. RT-qPCR, Western blot, and Immunohistochemistry assay were implemented to determine mRNA and protein expression. Endothelial permeability was assessed using the fluorescein isothiocyanate-dextran permeability assay in vitro. Reactive oxygen species (ROS) level was examined with a ROS detection kit.</p><p><strong>Results: </strong>TMAO dose-dependently exacerbated depression-like behavior, induced the activation of the p38/mitogen-activated protein kinase (MAPK) signaling pathway, downregulated the abundance of tight junction proteins, and resulted in the dysregulation of neurotrophic mediators in the PSD mouse model in vivo. Further, TMAO increased endothelial permeability and activated reactive oxygen species-p38/MAPK signaling. By introducing the p38/MAPK pathway inhibitor SB203580, TMAO was found to downregulate the expressions of tight junction proteins and promoted endothelial permeability via p38/MAPK signaling activation.</p><p><strong>Discussion: </strong>By activating the ROS-P38/MAPK pathway, TMAO enhanced the permeability of the blood-brain barrier after stroke. These mechanisms resulted in the dysregulation of neurotrophic mediators, ultimately leading to PSD progression.</p>","PeriodicalId":94029,"journal":{"name":"Human & experimental toxicology","volume":"43 ","pages":"9603271241306396"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142804205","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Oxidative stress reactions in rats with pulmonary injuries induced by sulfur mustard (1 LD50).
Pub Date : 2024-01-01 DOI: 10.1177/09603271241308772
Lu Liu, Xiaoxuan Hu, Na Zhang, Yuxu Zhong, Xiao-Ji Zhu, Tao Liu

Objective: Sulfur mustard (SM) is an important chemical warfare agent. The mechanisms underlying SM toxicity have not been completely elucidated. However, oxidative stress and the subsequent damage to macromolecules have been considered ascrucial steps in SM toxicity. In this study, a rat model of SM-induced acute pulmonary injury was established using an equal toxicity dose (1LD50). This study employed two methods to directly compare oxidative stress indices in serum enzymes and the epithelial cells of the alveolar septa.

Methods: Male Sprague-Dawley rats were randomly divided into intraperitoneal SM, intraperitoneal propylene glycol control, tracheal SM, tracheal propylene glycol control, and control groups. SM-induced serum enzyme levels and protein expression in the epithelial cells of the alveolar septa were measured using enzyme-linked immunosorbent assay and immunohistochemistry.

Results: Serum levels of superoxide dismutase, catalase, and glutathione peroxidase were upregulated in the intraperitoneal SM group compared with those in the tracheal SM group. Positive expression ratios of CuZn-superoxide dismutase, Mn-superoxide dismutase, paraoxonase-1, and apolipoprotein-1 proteins in the epithelial cells of the alveolar septa in the intraperitoneal SM group were elevated compared with those in the tracheal SM group.

Conclusion: Under SM (1LD50) exposure, there were significantly higher serum enzyme levels and protein expressions in the epithelial cells of the alveolar septa of rats injected with SM intraperitoneally compared with SM administered by intratracheal instillation. The results demonstrated that the differences in oxidative stress indices at the molecular level in SM-induced pulmonary injury were dependent on the route of exposure.

{"title":"Oxidative stress reactions in rats with pulmonary injuries induced by sulfur mustard (1 LD<sub>50</sub>).","authors":"Lu Liu, Xiaoxuan Hu, Na Zhang, Yuxu Zhong, Xiao-Ji Zhu, Tao Liu","doi":"10.1177/09603271241308772","DOIUrl":"https://doi.org/10.1177/09603271241308772","url":null,"abstract":"<p><strong>Objective: </strong>Sulfur mustard (SM) is an important chemical warfare agent. The mechanisms underlying SM toxicity have not been completely elucidated. However, oxidative stress and the subsequent damage to macromolecules have been considered ascrucial steps in SM toxicity. In this study, a rat model of SM-induced acute pulmonary injury was established using an equal toxicity dose (1LD<sub>50</sub>). This study employed two methods to directly compare oxidative stress indices in serum enzymes and the epithelial cells of the alveolar septa.</p><p><strong>Methods: </strong>Male Sprague-Dawley rats were randomly divided into intraperitoneal SM, intraperitoneal propylene glycol control, tracheal SM, tracheal propylene glycol control, and control groups. SM-induced serum enzyme levels and protein expression in the epithelial cells of the alveolar septa were measured using enzyme-linked immunosorbent assay and immunohistochemistry.</p><p><strong>Results: </strong>Serum levels of superoxide dismutase, catalase, and glutathione peroxidase were upregulated in the intraperitoneal SM group compared with those in the tracheal SM group. Positive expression ratios of CuZn-superoxide dismutase, Mn-superoxide dismutase, paraoxonase-1, and apolipoprotein-1 proteins in the epithelial cells of the alveolar septa in the intraperitoneal SM group were elevated compared with those in the tracheal SM group.</p><p><strong>Conclusion: </strong>Under SM (1LD<sub>50</sub>) exposure, there were significantly higher serum enzyme levels and protein expressions in the epithelial cells of the alveolar septa of rats injected with SM intraperitoneally compared with SM administered by intratracheal instillation. The results demonstrated that the differences in oxidative stress indices at the molecular level in SM-induced pulmonary injury were dependent on the route of exposure.</p>","PeriodicalId":94029,"journal":{"name":"Human & experimental toxicology","volume":"43 ","pages":"9603271241308772"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142879113","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
LncRNA-NEAT1 facilitates autophagy to boost pemetrexed resistance in lung adenocarcinoma via the mir-379-3p/HIF1A pathway. LncRNA-NEAT1通过mir-379-3p/HIF1A途径促进自噬,从而增强肺腺癌对培美曲塞的耐药性。
Pub Date : 2024-01-01 DOI: 10.1177/09603271241292169
Wei Hu, Wenjun Cao, Jiheng Liu

Background: As a primary chemotherapeutic agent for lung adenocarcinoma (LUAD), pemetrexed (PEM) faces the challenge of resistance development in cancer cells due to its chronic use, which compromises its therapeutic benefits. LncRNA-NEAT1, implicated in the promotion of cancer, is a key player in LUAD. The objective of this study is to explore the contribution of lncRNA-NEAT1 to PEM resistance in LUAD and to dissect the molecular mechanisms involved.

Method: The expression levels of lncRNA-NEAT1 in LUAD tissues and cells were deciphered using the TCGA database and qRT-PCR. To delve into the functional implications of lncRNA-NEAT1, we engineered plasmids to modulate its expression levels in PEM-resistant A549 cells. PEM resistance in the modified cells was then quantitatively assessed via a panel of assays including cell counting kit-8 (CCK-8), and colony formation, and flow cytometry. To predict the interaction sites between lncRNA-NEAT1 and miR-379-3p, along with the miR-379-3p and hypoxia-inducible factor (HIF1A), we referred to the StarBase and TargetScan databases. The interplay between these RNA molecules was further characterized by RNA immunoprecipitation (RIP) and dual-luciferase reporter assays, while the expression of autophagy-related proteins LC3I, LC3II, and Beclin1 was profiled using western blot (WB).

Results: Abundant lncRNA-NEAT1 expression was observed in LUAD tissues and cell lines. Its depletion resulted in impeded growth of A549/PEM cells, enhanced apoptotic rates, and a lowered threshold for PEM to exert a half-maximal inhibitory effect. The interplay between lncRNA-NEAT1 and miR-379-3p, as evidenced by dual-luciferase reporter assays, RIP, and qRT-PCR, led to the upregulation of HIF1A. WB and CCK-8 outcomes illustrated that the autophagy and PEM resistance were compromised when HIF1A expression was curtailed by miR-379-3p mimics in A549/PEM cells. The restoration of these effects was observed upon lncRNA-NEAT1-mediated downregulation of miR-379-3p.

Conclusion: Our study illuminates the role of lncRNA-NEAT1 in LUAD, where it mediates resistance to PEM through the activation of autophagy via the miR-379-3p/HIF1A axis. This work paves the way for new therapeutic strategies for managing PEM resistance in LUAD patients.

背景:作为肺腺癌(LUAD)的主要化疗药物,培美曲塞(PEM)因其长期使用而面临着癌细胞产生耐药性的挑战,这损害了其治疗效果。LncRNA-NEAT1 与癌症的发生有关,是 LUAD 中的一个关键角色。本研究旨在探讨lncRNA-NEAT1对LUAD的PEM耐药性的贡献,并剖析其中的分子机制:方法:利用TCGA数据库和qRT-PCR技术破译lncRNA-NEAT1在LUAD组织和细胞中的表达水平。为了深入研究lncRNA-NEAT1的功能意义,我们设计了质粒来调节其在抗PEM的A549细胞中的表达水平。然后通过细胞计数试剂盒-8(CCK-8)、菌落形成和流式细胞术等一系列检测方法对改造细胞的PEM耐药性进行定量评估。为了预测lncRNA-NEAT1与miR-379-3p以及miR-379-3p与缺氧诱导因子(HIF1A)之间的相互作用位点,我们参考了StarBase和TargetScan数据库。通过RNA免疫沉淀(RIP)和双荧光素酶报告实验进一步确定了这些RNA分子之间的相互作用,同时使用Western印迹(WB)分析了自噬相关蛋白LC3I、LC3II和Beclin1的表达:结果:在LUAD组织和细胞系中观察到大量lncRNA-NEAT1表达。结果:在LUAD组织和细胞系中观察到大量lncRNA-NEAT1表达,其耗竭导致A549/PEM细胞生长受阻、凋亡率升高以及PEM发挥半最大抑制作用的阈值降低。双荧光素酶报告实验、RIP和qRT-PCR证明,lncRNA-NEAT1和miR-379-3p之间的相互作用导致了HIF1A的上调。WB和CCK-8结果表明,当miR-379-3p模拟物抑制A549/PEM细胞中HIF1A的表达时,自噬和PEM抗性会受到影响。在lncRNA-NEAT1介导的miR-379-3p下调作用下,这些效应得以恢复:我们的研究阐明了lncRNA-NEAT1在LUAD中的作用,它通过miR-379-3p/HIF1A轴激活自噬介导对PEM的抵抗。这项研究为治疗 LUAD 患者对 PEM 的耐药性提供了新的治疗策略。
{"title":"LncRNA-NEAT1 facilitates autophagy to boost pemetrexed resistance in lung adenocarcinoma via the mir-379-3p/HIF1A pathway.","authors":"Wei Hu, Wenjun Cao, Jiheng Liu","doi":"10.1177/09603271241292169","DOIUrl":"https://doi.org/10.1177/09603271241292169","url":null,"abstract":"<p><strong>Background: </strong>As a primary chemotherapeutic agent for lung adenocarcinoma (LUAD), pemetrexed (PEM) faces the challenge of resistance development in cancer cells due to its chronic use, which compromises its therapeutic benefits. LncRNA-NEAT1, implicated in the promotion of cancer, is a key player in LUAD. The objective of this study is to explore the contribution of lncRNA-NEAT1 to PEM resistance in LUAD and to dissect the molecular mechanisms involved.</p><p><strong>Method: </strong>The expression levels of lncRNA-NEAT1 in LUAD tissues and cells were deciphered using the TCGA database and qRT-PCR. To delve into the functional implications of lncRNA-NEAT1, we engineered plasmids to modulate its expression levels in PEM-resistant A549 cells. PEM resistance in the modified cells was then quantitatively assessed via a panel of assays including cell counting kit-8 (CCK-8), and colony formation, and flow cytometry. To predict the interaction sites between lncRNA-NEAT1 and miR-379-3p, along with the miR-379-3p and hypoxia-inducible factor (HIF1A), we referred to the StarBase and TargetScan databases. The interplay between these RNA molecules was further characterized by RNA immunoprecipitation (RIP) and dual-luciferase reporter assays, while the expression of autophagy-related proteins LC3I, LC3II, and Beclin1 was profiled using western blot (WB).</p><p><strong>Results: </strong>Abundant lncRNA-NEAT1 expression was observed in LUAD tissues and cell lines. Its depletion resulted in impeded growth of A549/PEM cells, enhanced apoptotic rates, and a lowered threshold for PEM to exert a half-maximal inhibitory effect. The interplay between lncRNA-NEAT1 and miR-379-3p, as evidenced by dual-luciferase reporter assays, RIP, and qRT-PCR, led to the upregulation of HIF1A. WB and CCK-8 outcomes illustrated that the autophagy and PEM resistance were compromised when HIF1A expression was curtailed by miR-379-3p mimics in A549/PEM cells. The restoration of these effects was observed upon lncRNA-NEAT1-mediated downregulation of miR-379-3p.</p><p><strong>Conclusion: </strong>Our study illuminates the role of lncRNA-NEAT1 in LUAD, where it mediates resistance to PEM through the activation of autophagy via the miR-379-3p/HIF1A axis. This work paves the way for new therapeutic strategies for managing PEM resistance in LUAD patients.</p>","PeriodicalId":94029,"journal":{"name":"Human & experimental toxicology","volume":"43 ","pages":"9603271241292169"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142484138","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Assessment of safety through mutagenicity and subchronic toxicity studies with black pepper extract preparation. 通过黑胡椒提取物制剂的诱变性和亚慢性毒性研究评估其安全性。
Pub Date : 2024-01-01 DOI: 10.1177/09603271241298531
Lee Chae, Sungwon Lee, Swetha Mahadevan, Mark R Bauter, Colleen Wojenski, Kristin Robertson, Brian Premkumar, Brinda Mahadevan

Introduction: Black pepper (Piper nigrum) is a rich source of natural and bioactive components such as N-trans-feruloyltyramine (NFT). In this paper, we discuss the results of the subchronic toxicity and mutagenicity studies conducted to understand the potential for adverse effects if any, of Black Pepper Extract Preparation (BPE).

Methods: To evaluate mutagenicity, an Ames test was conducted with BPE in the presence and absence of S9 metabolic activation. Long-term safety was inferred through a 90-day subchronic toxicology study using adult rats. Dose levels were selected with expected human intake levels of NFT (120 mg/kg/day), with an acceptable safety factor, for preclinical safety and tolerability. Sprague Dawley rats were fed diets targeting dietary intakes (doses) of 0, 125, 350, or 700 mg/kg/day of BPE for 90 days, an NFT dose level equivalent to 68, 190, and 380 mg/kg/day.

Results: In vitro Ames test up to 5000 µg/plate with and without S9 metabolic activation showed no BPE-related increases in revertant colony numbers and was non-mutagenic. There were no BPE-related changes in viability, clinical signs, body weight, food consumption, and organ weights. BPE dietary administration did not induce any treatment-related changes in hematology, clinical chemistry, other macroscopic or microscopic endpoints.

Discussion and conclusion: The highest dose tested with BPE (700 mg/kg/day) was the no-observed-adverse-effect level (NOAEL) that revealed no adverse effects. Based on toxicological endpoints evaluated, this NOAEL for BPE corresponded to a human equivalent NFT dose level of 380 mg/kg/day, dependent upon a (∼50%) concentration of NFT in BPE.

简介黑胡椒(Piper nigrum)含有丰富的天然生物活性成分,如 N-反式阿魏酰酪胺(N-trans-feruloyltyramine,NFT)。本文讨论了亚慢性毒性和诱变性研究的结果,以了解黑胡椒提取物制剂(BPE)的潜在不良影响(如果有的话):为了评估诱变性,在有 S9 代谢活化和没有 S9 代谢活化的情况下对 BPE 进行了 Ames 试验。通过对成年大鼠进行为期 90 天的亚慢性毒理学研究,推断其长期安全性。剂量水平是根据人类对 NFT 的预期摄入水平(120 毫克/千克/天)和可接受的安全系数选定的,以确保临床前安全性和耐受性。给 Sprague Dawley 大鼠喂食目标膳食摄入量(剂量)为 0、125、350 或 700 毫克/千克/天的 BPE,持续 90 天,NFT 剂量水平相当于 68、190 和 380 毫克/千克/天:结果:体外阿姆斯试验(5000 微克/板)(含或不含 S9 代谢活化)显示,逆转菌落数的增加与 BPE 无关,且无突变性。存活率、临床症状、体重、食量和器官重量都没有发生与 BPE 相关的变化。BPE 膳食给药不会引起血液学、临床化学、其他宏观或微观终点发生任何与治疗相关的变化:BPE 测试的最高剂量(700 毫克/千克/天)为无观测不良效应水平(NOAEL),未发现任何不良影响。根据评估的毒理学终点,BPE 的无观测不良效应水平相当于 380 毫克/千克/天的人体等效 NFT 剂量水平,这取决于 BPE 中 NFT 的浓度(∼50%)。
{"title":"Assessment of safety through mutagenicity and subchronic toxicity studies with black pepper extract preparation.","authors":"Lee Chae, Sungwon Lee, Swetha Mahadevan, Mark R Bauter, Colleen Wojenski, Kristin Robertson, Brian Premkumar, Brinda Mahadevan","doi":"10.1177/09603271241298531","DOIUrl":"https://doi.org/10.1177/09603271241298531","url":null,"abstract":"<p><strong>Introduction: </strong>Black pepper (<i>Piper nigrum</i>) is a rich source of natural and bioactive components such as N-trans-feruloyltyramine (NFT). In this paper, we discuss the results of the subchronic toxicity and mutagenicity studies conducted to understand the potential for adverse effects if any, of Black Pepper Extract Preparation (BPE).</p><p><strong>Methods: </strong>To evaluate mutagenicity, an Ames test was conducted with BPE in the presence and absence of S9 metabolic activation. Long-term safety was inferred through a 90-day subchronic toxicology study using adult rats. Dose levels were selected with expected human intake levels of NFT (120 mg/kg/day), with an acceptable safety factor, for preclinical safety and tolerability. Sprague Dawley rats were fed diets targeting dietary intakes (doses) of 0, 125, 350, or 700 mg/kg/day of BPE for 90 days, an NFT dose level equivalent to 68, 190, and 380 mg/kg/day.</p><p><strong>Results: </strong>In vitro Ames test up to 5000 µg/plate with and without S9 metabolic activation showed no BPE-related increases in revertant colony numbers and was non-mutagenic. There were no BPE-related changes in viability, clinical signs, body weight, food consumption, and organ weights. BPE dietary administration did not induce any treatment-related changes in hematology, clinical chemistry, other macroscopic or microscopic endpoints.</p><p><strong>Discussion and conclusion: </strong>The highest dose tested with BPE (700 mg/kg/day) was the no-observed-adverse-effect level (NOAEL) that revealed no adverse effects. Based on toxicological endpoints evaluated, this NOAEL for BPE corresponded to a human equivalent NFT dose level of 380 mg/kg/day, dependent upon a (∼50%) concentration of NFT in BPE.</p>","PeriodicalId":94029,"journal":{"name":"Human & experimental toxicology","volume":"43 ","pages":"9603271241298531"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142607697","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Liver and ovarian toxicities boosted by bisphenol and gamma radiation in female albino rats. 双酚和伽马射线对雌性白化大鼠肝脏和卵巢的毒性增强
Pub Date : 2024-01-01 DOI: 10.1177/09603271231219264
Asmaa A Hassan, Sherein S Abdelgayed, Somaya Z Mansour

Bisphenol A (BPA), a carbon-based synthetic polymer compound, was newly classified as an environmental toxicant and an endocrine-disrupting chemical leading to abnormalities in cell proliferation, apoptosis, or migration that contributes to cancer development and progression. This study aims to evaluate the effect of the elevation of γ- radiation dose and BPA on the liver and ovaries of female rats. In this study, eighty female albino rats (130-150 g) were used in this work. Rats in this experiment received BPA in ethanol (50 mg/kg b. wt.) for 30 days, day after day, and in the irradiated groups, animals were administered BPA and then exposed to γ- radiation in doses (2, 4, and 6 Gy) one shot dose. Several members of the cytochrome family were examined. Exposure to γ-radiation and BPA showed an increase in cytochrome P450 and b5 fold change. Further, BPA and γ-radiation activate α and β estrogen receptors and also downregulate aromatase (CYT19) fold change. The current results also revealed that BPA and/or γ-radiation regulate the protein expression of the PI3K/Akt signaling pathway. The steroidogenic acute regulatory protein (StAR) appeared to be targeted by BPA and γ-radiation and its relative expression was elevated significantly by raising the γ-radiation dose. In conclusion, exposure to BPA, an endocrine-disrupting chemical, leads to marked toxicity. Additionally, toxicity is heightened by increasing the γ-radiation dose, either alone or in combination with BPA.

双酚 A(BPA)是一种碳基合成高分子化合物,新近被列为环境有毒物质和干扰内分泌的化学物质,会导致细胞增殖、凋亡或迁移异常,从而导致癌症的发生和发展。本研究旨在评估γ 辐射剂量和双酚 A 升高对雌性大鼠肝脏和卵巢的影响。本研究使用了 80 只白化雌性大鼠(体重 130-150 克)。在辐照组中,给动物注射双酚 A 后,再以 2、4 和 6 Gy 剂量一次性照射γ-射线。对细胞色素家族的几个成员进行了检测。暴露于γ射线和双酚 A 显示细胞色素 P450 和 b5 的折叠变化增加。此外,双酚 A 和 γ 辐射激活了 α 和 β 雌激素受体,并下调了芳香化酶(CYT19)的折叠变化。目前的研究结果还显示,双酚 A 和/或γ-辐射调节 PI3K/Akt 信号通路的蛋白表达。类固醇生成急性调节蛋白(StAR)似乎是双酚A和γ-辐射的靶标,其相对表达量随着γ-辐射剂量的增加而显著升高。总之,暴露于双酚 A 这种干扰内分泌的化学物质会导致明显的毒性。此外,增加γ-辐射剂量(无论是单独使用还是与双酚A一起使用)会增强毒性。
{"title":"Liver and ovarian toxicities boosted by bisphenol and gamma radiation in female albino rats.","authors":"Asmaa A Hassan, Sherein S Abdelgayed, Somaya Z Mansour","doi":"10.1177/09603271231219264","DOIUrl":"10.1177/09603271231219264","url":null,"abstract":"<p><p>Bisphenol A (BPA), a carbon-based synthetic polymer compound, was newly classified as an environmental toxicant and an endocrine-disrupting chemical leading to abnormalities in cell proliferation, apoptosis, or migration that contributes to cancer development and progression. This study aims to evaluate the effect of the elevation of γ- radiation dose and BPA on the liver and ovaries of female rats. In this study, eighty female albino rats (130-150 g) were used in this work. Rats in this experiment received BPA in ethanol (50 mg/kg b. wt.) for 30 days, day after day, and in the irradiated groups, animals were administered BPA and then exposed to γ- radiation in doses (2, 4, and 6 Gy) one shot dose. Several members of the cytochrome family were examined. Exposure to γ-radiation and BPA showed an increase in cytochrome P450 and b5 fold change. Further, BPA and γ-radiation activate α and β estrogen receptors and also downregulate aromatase (CYT19) fold change. The current results also revealed that BPA and/or γ-radiation regulate the protein expression of the PI3K/Akt signaling pathway. The steroidogenic acute regulatory protein (StAR) appeared to be targeted by BPA and γ-radiation and its relative expression was elevated significantly by raising the γ-radiation dose. In conclusion, exposure to BPA, an endocrine-disrupting chemical, leads to marked toxicity. Additionally, toxicity is heightened by increasing the γ-radiation dose, either alone or in combination with BPA.</p>","PeriodicalId":94029,"journal":{"name":"Human & experimental toxicology","volume":"43 ","pages":"9603271231219264"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139543861","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ginsenoside Rg5 induces NSCLC cell apoptosis and autophagy through PI3K/Akt/mTOR signaling pathway. 人参皂苷Rg5通过PI3K/Akt/mTOR信号通路诱导NSCLC细胞凋亡和自噬。
Pub Date : 2024-01-01 DOI: 10.1177/09603271241229140
Caidie Zhang, Yan Jin

Objective: Ginsenoside Rg5 (Rg5) is a minor ginsenoside of ginseng and has a strong anti-tumor potential. This study focused on deciphering the function of Rg5 in non-small cell lung cancer (NSCLC) and investigating its related mechanism.

Methods: After treating human NSCLC cell lines (H1650 and A549) and bronchial epithelial cells (BEAS-2B) with increasing concentration of Rg5, cell viability was examined using methyl thiazolyl tetrazolium (MTT) assay. NSCLC cell proliferation and apoptosis were evaluated by colony formation assay and flow cytometry, respectively. The levels of proteins associated with cell cycle progression, cell apoptosis, and autophagy as well as the key markers in the PI3K/Akt/mTOR pathway were measured using western blot. A xenograft nude mouse model was established to explore the function of Rg5 in vivo.

Results: NSCLC cell viability was dose- and time-dependently suppressed after Rg5 treatment. Rg5 restrained NSCLC cell proliferation by inducing G2/M phase arrest via regulation of cell cycle-related genes including p21, cyclin B1, and Cdc2. Additionally, Rg5 promoted caspase-dependent apoptosis in NSCLC cells by regulating the intrinsic mitochondrial signaling pathway. Rg5 induced autophagy via the regulation of autophagy-related proteins. The in vivo experiments revealed the inhibitory impact of Rg5 on xenograft growth. Rg5 also inactivated the PI3K/Akt/mTOR signaling pathway in NSCLC cells and mouse tumors.

Conclusion: Rg5 induced autophagy and caspase-dependent apoptosis in NSCLC cells by inhibiting the PI3K/Akt/mTOR signaling pathway, suggesting that Rg5 might become a promising and novel anti-tumor agent for the clinical treatment of NSCLC patients.

研究目的人参皂苷Rg5(Rg5)是人参中的一种次要人参皂苷,具有很强的抗肿瘤潜力。本研究的重点是破译 Rg5 在非小细胞肺癌(NSCLC)中的功能及其相关机制:方法:用不同浓度的 Rg5 处理人 NSCLC 细胞系(H1650 和 A549)和支气管上皮细胞(BEAS-2B)后,用甲基噻唑基四氮唑(MTT)测定法检测细胞活力。NSCLC 细胞增殖和凋亡分别通过集落形成试验和流式细胞术进行评估。采用 Western 印迹法测定了与细胞周期进展、细胞凋亡和自噬相关的蛋白质水平,以及 PI3K/Akt/mTOR 通路中的关键标记物。建立了一个异种移植裸鼠模型,以探讨Rg5在体内的功能:结果:Rg5处理后,NSCLC细胞活力受到剂量和时间依赖性抑制。Rg5通过调控细胞周期相关基因(包括p21、细胞周期蛋白B1和Cdc2)诱导G2/M期停滞,从而抑制NSCLC细胞增殖。此外,Rg5 还能通过调节线粒体固有信号通路,促进 NSCLC 细胞发生依赖于 Caspase 的凋亡。Rg5通过调节自噬相关蛋白诱导自噬。体内实验显示了Rg5对异种移植生长的抑制作用。Rg5还能使NSCLC细胞和小鼠肿瘤中的PI3K/Akt/mTOR信号通路失活:结论:Rg5通过抑制PI3K/Akt/mTOR信号通路诱导NSCLC细胞自噬和依赖于Caspase的细胞凋亡,表明Rg5可能成为临床治疗NSCLC患者的一种前景广阔的新型抗肿瘤药物。
{"title":"Ginsenoside Rg5 induces NSCLC cell apoptosis and autophagy through PI3K/Akt/mTOR signaling pathway.","authors":"Caidie Zhang, Yan Jin","doi":"10.1177/09603271241229140","DOIUrl":"10.1177/09603271241229140","url":null,"abstract":"<p><strong>Objective: </strong>Ginsenoside Rg5 (Rg5) is a minor ginsenoside of ginseng and has a strong anti-tumor potential. This study focused on deciphering the function of Rg5 in non-small cell lung cancer (NSCLC) and investigating its related mechanism.</p><p><strong>Methods: </strong>After treating human NSCLC cell lines (H1650 and A549) and bronchial epithelial cells (BEAS-2B) with increasing concentration of Rg5, cell viability was examined using methyl thiazolyl tetrazolium (MTT) assay. NSCLC cell proliferation and apoptosis were evaluated by colony formation assay and flow cytometry, respectively. The levels of proteins associated with cell cycle progression, cell apoptosis, and autophagy as well as the key markers in the PI3K/Akt/mTOR pathway were measured using western blot. A xenograft nude mouse model was established to explore the function of Rg5 <i>in vivo</i>.</p><p><strong>Results: </strong>NSCLC cell viability was dose- and time-dependently suppressed after Rg5 treatment. Rg5 restrained NSCLC cell proliferation by inducing G2/M phase arrest via regulation of cell cycle-related genes including p21, cyclin B1, and Cdc2. Additionally, Rg5 promoted caspase-dependent apoptosis in NSCLC cells by regulating the intrinsic mitochondrial signaling pathway. Rg5 induced autophagy via the regulation of autophagy-related proteins. The <i>in vivo</i> experiments revealed the inhibitory impact of Rg5 on xenograft growth. Rg5 also inactivated the PI3K/Akt/mTOR signaling pathway in NSCLC cells and mouse tumors.</p><p><strong>Conclusion: </strong>Rg5 induced autophagy and caspase-dependent apoptosis in NSCLC cells by inhibiting the PI3K/Akt/mTOR signaling pathway, suggesting that Rg5 might become a promising and novel anti-tumor agent for the clinical treatment of NSCLC patients.</p>","PeriodicalId":94029,"journal":{"name":"Human & experimental toxicology","volume":"43 ","pages":"9603271241229140"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139577295","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Subchronic oral toxicity assessment of Bacillus velezensis strain BV379 in sprague-dawley rats. BV379 株 BV379 芽孢杆菌对斯普拉格-道利大鼠的亚慢性口服毒性评估。
Pub Date : 2024-01-01 DOI: 10.1177/09603271241278977
Mark R Bauter, Laura M Brutscher, Laurie C Dolan, Jessica L Spears

Introduction: The spore-forming bacterial species Bacillus velezensis is commonly utilized in feed for livestock and aquaculture. In recent years, there has been increased interest in introducing B. velezensis into human supplements and food. Before it can be safely administered in humans, the safety of each B. velezensis strain needs to be established. The objective of this study was to evaluate the in vivo safety of Bacillus velezensis strain BV379 by high-dose oral administration to rats in a 28-day subchronic toxicity study.

Methods: In this study, 80 animals were assigned to four groups: vehicle control, 1 × 1010, 4 × 1010, or 10 × 1010 CFU/kg bw/day by gavage. The following toxicological assessments were performed: ophthalmological examinations; observations for viability, signs of gross toxicity, and behavioral changes; in-life parameters, including body weight and food consumption; urinalysis, hematology, clinical chemistry, and coagulation assessments; macroscopic and microscopic tissue assessments; and bacterial enumeration in selected tissues.

Results: Under the conditions of this study, no adverse clinical endpoints were attributed to the administration of Bacillus velezensis strain BV379, which was well-tolerated up to the highest dose of 10 × 1010 CFU/kg bw/day.

Conclusion: These results support the in vivo pre-clinical safety of Bacillus velezensis strain BV379 for use in food and supplements.

导言:芽孢杆菌(Bacillus velezensis)通常用于家畜和水产养殖的饲料中。近年来,人们对将 B. velezensis 引入人类补充剂和食品的兴趣日益浓厚。在对人类安全施用之前,需要确定每种 B. velezensis 菌株的安全性。本研究的目的是通过对大鼠进行为期 28 天的亚慢性毒性研究,评估高剂量口服 Velezensis 杆菌菌株 BV379 在体内的安全性:在这项研究中,80只大鼠被分为四组:给药对照组、1×1010、4×1010或10×1010 CFU/kg体重/天组。进行了以下毒理学评估:眼科检查;观察存活率、大体毒性迹象和行为变化;生活参数,包括体重和食物消耗量;尿液分析、血液学、临床化学和凝血评估;宏观和微观组织评估;以及选定组织中的细菌计数:结果:在本研究的条件下,施用韦氏芽孢杆菌菌株 BV379 未导致任何不良临床终点:这些结果证明了在食品和补充剂中使用韦氏芽孢杆菌菌株 BV379 的体内临床前安全性。
{"title":"Subchronic oral toxicity assessment of <i>Bacillus velezensis</i> strain BV379 in sprague-dawley rats.","authors":"Mark R Bauter, Laura M Brutscher, Laurie C Dolan, Jessica L Spears","doi":"10.1177/09603271241278977","DOIUrl":"https://doi.org/10.1177/09603271241278977","url":null,"abstract":"<p><strong>Introduction: </strong>The spore-forming bacterial species <i>Bacillus velezensis</i> is commonly utilized in feed for livestock and aquaculture. In recent years, there has been increased interest in introducing <i>B</i>. <i>velezensis</i> into human supplements and food. Before it can be safely administered in humans, the safety of each <i>B</i>. <i>velezensis</i> strain needs to be established. The objective of this study was to evaluate the in vivo safety of <i>Bacillus velezensis</i> strain BV379 by high-dose oral administration to rats in a 28-day subchronic toxicity study.</p><p><strong>Methods: </strong>In this study, 80 animals were assigned to four groups: vehicle control, 1 × 10<sup>10</sup>, 4 × 10<sup>10</sup>, or 10 × 10<sup>10</sup> CFU/kg bw/day by gavage. The following toxicological assessments were performed: ophthalmological examinations; observations for viability, signs of gross toxicity, and behavioral changes; in-life parameters, including body weight and food consumption; urinalysis, hematology, clinical chemistry, and coagulation assessments; macroscopic and microscopic tissue assessments; and bacterial enumeration in selected tissues.</p><p><strong>Results: </strong>Under the conditions of this study, no adverse clinical endpoints were attributed to the administration of <i>Bacillus velezensis</i> strain BV379, which was well-tolerated up to the highest dose of 10 × 10<sup>10</sup> CFU/kg bw/day.</p><p><strong>Conclusion: </strong>These results support the in vivo pre-clinical safety of <i>Bacillus velezensis</i> strain BV379 for use in food and supplements.</p>","PeriodicalId":94029,"journal":{"name":"Human & experimental toxicology","volume":"43 ","pages":"9603271241278977"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142335177","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MG132 inhibits proliferation and induces apoptosis of acute lymphoblastic leukemia via Akt/FOXO3a/Bim pathway. MG132 通过 Akt/FOXO3a/Bim 通路抑制急性淋巴细胞白血病的增殖并诱导其凋亡。
Pub Date : 2024-01-01 DOI: 10.1177/09603271241303030
Zhong Fan, Wen-Hao Lin, Cong Liang, Yu Li, Chun-Jin Peng, Jie-Si Luo, Wen-Yan Tang, Li-Min Zheng, Dan-Ping Huang, Zhi-Yong Ke, Li-Na Wang, Xiao-Li Zhang, Li-Bin Huang

Background: Acute lymphoblastic leukemia (ALL) is one of the most common pediatric cancers, characterized by the malignant proliferation of leukemic cells. Despite advancements in treatment, the prognosis for refractory and relapsed ALL remains poor, underscoring the need for novel therapeutic targets and approaches.

Methods: To investigate the anti-leukemic properties of MG132, MTS assays were employed to assess cell viability, and flow cytometry was used to evaluate apoptosis. Mechanistic studies, including qRT-PCR, Western blotting, and lentivirus-mediated FOXO3a knockdown, were conducted to explore MG132's effects on the Akt/FOXO3a/Bim signaling pathway. A xenograft mouse model was utilized to validate the in vivo efficacy of MG132 in suppressing tumor growth.

Results: MG132 inhibited cell proliferation and induced apoptosis in both ALL cell lines and primary cells in a concentration-dependent manner. Mechanistic studies revealed that MG132 promoted FOXO3a nuclear localization by suppressing Akt phosphorylation and preventing FOXO3a degradation, leading to increased Bim expression. Furthermore, FOXO3a knockdown significantly reduced MG132's anti-proliferative effects. In vivo, MG132 markedly inhibited tumor growth in the xenograft model.

Conclusion: These findings suggest that MG132 exerts potent anti-leukemic effects through modulation of the Akt/FOXO3a/Bim axis, offering a promising therapeutic avenue for treating ALL.

背景:急性淋巴细胞白血病(ALL)是最常见的儿童癌症之一,其特点是白血病细胞恶性增殖。尽管在治疗方面取得了进展,但难治性和复发性ALL的预后仍然很差,这凸显了对新型治疗靶点和方法的需求:为了研究MG132的抗白血病特性,我们采用MTS测定法评估细胞活力,并用流式细胞术评估细胞凋亡。为了探讨MG132对Akt/FOXO3a/Bim信号通路的影响,进行了包括qRT-PCR、Western印迹和慢病毒介导的FOXO3a基因敲除在内的机理研究。利用异种移植小鼠模型验证了MG132抑制肿瘤生长的体内疗效:结果:MG132以浓度依赖性方式抑制ALL细胞系和原代细胞的细胞增殖并诱导其凋亡。机理研究发现,MG132通过抑制Akt磷酸化和阻止FOXO3a降解来促进FOXO3a核定位,从而导致Bim表达增加。此外,敲除 FOXO3a 能显著降低 MG132 的抗增殖作用。在体内,MG132能明显抑制异种移植模型中肿瘤的生长:这些研究结果表明,MG132通过调节Akt/FOXO3a/Bim轴发挥了强有力的抗白血病作用,为治疗ALL提供了一种前景广阔的治疗途径。
{"title":"MG132 inhibits proliferation and induces apoptosis of acute lymphoblastic leukemia via Akt/FOXO3a/Bim pathway.","authors":"Zhong Fan, Wen-Hao Lin, Cong Liang, Yu Li, Chun-Jin Peng, Jie-Si Luo, Wen-Yan Tang, Li-Min Zheng, Dan-Ping Huang, Zhi-Yong Ke, Li-Na Wang, Xiao-Li Zhang, Li-Bin Huang","doi":"10.1177/09603271241303030","DOIUrl":"https://doi.org/10.1177/09603271241303030","url":null,"abstract":"<p><strong>Background: </strong>Acute lymphoblastic leukemia (ALL) is one of the most common pediatric cancers, characterized by the malignant proliferation of leukemic cells. Despite advancements in treatment, the prognosis for refractory and relapsed ALL remains poor, underscoring the need for novel therapeutic targets and approaches.</p><p><strong>Methods: </strong>To investigate the anti-leukemic properties of MG132, MTS assays were employed to assess cell viability, and flow cytometry was used to evaluate apoptosis. Mechanistic studies, including qRT-PCR, Western blotting, and lentivirus-mediated FOXO3a knockdown, were conducted to explore MG132's effects on the Akt/FOXO3a/Bim signaling pathway. A xenograft mouse model was utilized to validate the in vivo efficacy of MG132 in suppressing tumor growth.</p><p><strong>Results: </strong>MG132 inhibited cell proliferation and induced apoptosis in both ALL cell lines and primary cells in a concentration-dependent manner. Mechanistic studies revealed that MG132 promoted FOXO3a nuclear localization by suppressing Akt phosphorylation and preventing FOXO3a degradation, leading to increased Bim expression. Furthermore, FOXO3a knockdown significantly reduced MG132's anti-proliferative effects. In vivo, MG132 markedly inhibited tumor growth in the xenograft model.</p><p><strong>Conclusion: </strong>These findings suggest that MG132 exerts potent anti-leukemic effects through modulation of the Akt/FOXO3a/Bim axis, offering a promising therapeutic avenue for treating ALL.</p>","PeriodicalId":94029,"journal":{"name":"Human & experimental toxicology","volume":"43 ","pages":"9603271241303030"},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142718154","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Problems with the effectiveness of L-carnitine and paraffin oil in acute aluminum phosphide poisoning. L-肉碱和石蜡油治疗急性磷化铝中毒的疗效问题。
Pub Date : 2023-01-01 DOI: 10.1177/09603271231210974
Maryam Zaare Nahandi, Ali Banagozar Mohammadi
{"title":"Problems with the effectiveness of L-carnitine and paraffin oil in acute aluminum phosphide poisoning.","authors":"Maryam Zaare Nahandi,&nbsp;Ali Banagozar Mohammadi","doi":"10.1177/09603271231210974","DOIUrl":"10.1177/09603271231210974","url":null,"abstract":"","PeriodicalId":94029,"journal":{"name":"Human & experimental toxicology","volume":"42 ","pages":"9603271231210974"},"PeriodicalIF":0.0,"publicationDate":"2023-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50164248","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Human & experimental toxicology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1