首页 > 最新文献

Neurotoxicity Research最新文献

英文 中文
Melatonin Alleviates Erastin-Induced Cell Death by Inhibiting Ferroptosis and Amyloid Precursor Protein Processing in Neuronal Cell Lines. 褪黑素通过抑制神经细胞系的铁下垂和淀粉样前体蛋白加工来减轻erastin诱导的细胞死亡。
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2025-05-30 DOI: 10.1007/s12640-025-00747-1
Suwakon Wongjaikam, Puntita Siengdee, Alliya Somnus, Piyarat Govitrapong

Ferroptosis is an iron-dependent and membrane lipid peroxidation-mediated form of programmed or regulated cell death. A number of recent studies have demonstrated that ferroptosis contributes to Alzheimer's disease (AD)-mediated nerve cell death. Melatonin demonstrates strong antioxidant properties and offers protective benefits for the brain in the context of AD. However, it is not fully known whether melatonin protects against ferroptosis and whether ferroptosis affects amyloid precursor protein (APP) processing. In this study, we studied the effects of melatonin on SH-SY5Y cells-induced ferroptosis using erastin, and ferrostatin-1 was used as a ferroptosis inhibitor. To confirm the occurrence of ferroptosis, we conducted measurements of cell cytotoxicity, intracellular iron, reactive oxygen species (ROS), and 4-hydroxynonenal (4-HNE). The protein expressions that were regulated by either ferroptosis or APP processing were measured. Our results revealed that erastin increased intracellular iron levels, ROS, and 4-HNE lipid peroxidation in SH-SY5Y cells, resulting in an increased percentage of cell death. Erastin disrupted the regulation of proteins involved in ferroptosis and increased the production of amyloid beta (Aβ) through APP proteolysis. Following melatonin treatment, intracellular iron, ROS, and 4-HNE levels were significantly reduced. Additionally, the cystine/glutamate antiporter (system xc-) and glutathione peroxidase 4 (GPX4) were increased, and acyl-CoA synthetase long chain family member 4 (ACSL4) was diminished. APP, β-site-APP cleaving enzyme 1 (BACE1), presenilin 1 (PS1) and Aβ production were alleviated in erastin-treated SH-SY5Y cells. In conclusion, melatonin effectively inhibits ferroptosis-related cell death and AD-like conditions induced by erastin in SH-SY5Y human neuroblastoma cell lines.

铁死亡是一种铁依赖性和膜脂过氧化介导的程序性或受调节的细胞死亡形式。最近的一些研究表明,铁下垂有助于阿尔茨海默病(AD)介导的神经细胞死亡。褪黑素显示出强大的抗氧化特性,并在AD的情况下为大脑提供保护作用。然而,褪黑素是否对铁下垂有保护作用以及铁下垂是否影响淀粉样前体蛋白(APP)的加工尚不完全清楚。在本研究中,我们使用erastin研究褪黑素对SH-SY5Y细胞诱导的铁下垂的影响,并使用铁抑素-1作为铁下垂抑制剂。为了证实铁下垂的发生,我们测量了细胞毒性、细胞内铁、活性氧(ROS)和4-羟基壬烯醛(4-HNE)。测定受铁下垂或APP加工调节的蛋白表达。我们的研究结果显示,在SH-SY5Y细胞中,erastin增加细胞内铁水平、ROS和4-HNE脂质过氧化,导致细胞死亡百分比增加。Erastin破坏了与铁下垂有关的蛋白质的调节,并通过APP蛋白水解增加了β淀粉样蛋白(Aβ)的产生。褪黑素治疗后,细胞内铁、ROS和4-HNE水平显著降低。此外,胱氨酸/谷氨酸反转运蛋白(system xc-)和谷胱甘肽过氧化物酶4 (GPX4)增加,酰基辅酶a合成酶长链家族成员4 (ACSL4)减少。在erastin处理的SH-SY5Y细胞中,APP、β-位点-APP切割酶1 (BACE1)、早老素1 (PS1)和Aβ的产生均有所减少。综上所述,褪黑素可有效抑制SH-SY5Y人神经母细胞瘤细胞系中由erastin诱导的铁中毒相关细胞死亡和ad样疾病。
{"title":"Melatonin Alleviates Erastin-Induced Cell Death by Inhibiting Ferroptosis and Amyloid Precursor Protein Processing in Neuronal Cell Lines.","authors":"Suwakon Wongjaikam, Puntita Siengdee, Alliya Somnus, Piyarat Govitrapong","doi":"10.1007/s12640-025-00747-1","DOIUrl":"10.1007/s12640-025-00747-1","url":null,"abstract":"<p><p>Ferroptosis is an iron-dependent and membrane lipid peroxidation-mediated form of programmed or regulated cell death. A number of recent studies have demonstrated that ferroptosis contributes to Alzheimer's disease (AD)-mediated nerve cell death. Melatonin demonstrates strong antioxidant properties and offers protective benefits for the brain in the context of AD. However, it is not fully known whether melatonin protects against ferroptosis and whether ferroptosis affects amyloid precursor protein (APP) processing. In this study, we studied the effects of melatonin on SH-SY5Y cells-induced ferroptosis using erastin, and ferrostatin-1 was used as a ferroptosis inhibitor. To confirm the occurrence of ferroptosis, we conducted measurements of cell cytotoxicity, intracellular iron, reactive oxygen species (ROS), and 4-hydroxynonenal (4-HNE). The protein expressions that were regulated by either ferroptosis or APP processing were measured. Our results revealed that erastin increased intracellular iron levels, ROS, and 4-HNE lipid peroxidation in SH-SY5Y cells, resulting in an increased percentage of cell death. Erastin disrupted the regulation of proteins involved in ferroptosis and increased the production of amyloid beta (Aβ) through APP proteolysis. Following melatonin treatment, intracellular iron, ROS, and 4-HNE levels were significantly reduced. Additionally, the cystine/glutamate antiporter (system xc<sup>-</sup>) and glutathione peroxidase 4 (GPX4) were increased, and acyl-CoA synthetase long chain family member 4 (ACSL4) was diminished. APP, β-site-APP cleaving enzyme 1 (BACE1), presenilin 1 (PS1) and Aβ production were alleviated in erastin-treated SH-SY5Y cells. In conclusion, melatonin effectively inhibits ferroptosis-related cell death and AD-like conditions induced by erastin in SH-SY5Y human neuroblastoma cell lines.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"43 3","pages":"25"},"PeriodicalIF":2.9,"publicationDate":"2025-05-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144182917","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Streptozotocin and L-Buthionine-Sulfoximine Decrease Neuron Membrane Lipid Packing and Alter Insulin Signaling. 链脲佐菌素和l -丁硫氨酸-亚砜亚胺减少神经元膜脂堆积和改变胰岛素信号。
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2025-05-29 DOI: 10.1007/s12640-025-00749-z
Yandara A Martins, Camila A E F Cardinali, Andressa P Costa, Andréa S Torrão

Membrane composition, permeability and fluidity are essential for proper cellular function. According to the membrane aging hypothesis, aging-related diseases, including neurodegenerative disorders, arise from the aging of cell membranes. Membrane proteins, such as the insulin receptor, rely on an optimal membrane environment for proper partitioning and functionality. Our goal was to investigate the effects of streptozotocin (STZ) and L-buthionine-sulfoximine (BSO), two commonly used agents to model aging and neurodegeneration, on membrane composition and permeability, as well as their impact on insulin signaling. Mouse neuroblastoma 2a cell line (neuro-2a) were treated with STZ (6 h) and BSO (24 h). Cell viability was assessed by the MTT assay. Cholesterol and sphingomyelin content were quantified by commercial kits, while membrane polarity was evaluated with the Laurdan probe. Gene expression of Srebf2 and Cyp46a1 was analyzed by qPCR. Proteins from the insulin signaling pathway were examined by immunoblotting. STZ treatment reduced neuronal cholesterol content, downregulated Srebf2 and Cyp46a1 gene expression, and decreased membrane packing. In contrast, BSO-treated cells exhibited increased sphingomyelin content, upregulated Srebf2 and Cyp46a1 gene expression, and decreased membrane packing. Both treatments induced an insulin-resistant state, which we attribute to alterations in the membrane environment.

膜的组成、渗透性和流动性是细胞正常功能所必需的。根据膜老化假说,与衰老有关的疾病,包括神经退行性疾病,都是由细胞膜老化引起的。膜蛋白,如胰岛素受体,依赖于最佳的膜环境来进行适当的分配和功能。我们的目的是研究链脲佐菌素(STZ)和l -丁硫氨酸-亚砜胺(BSO)这两种常用的模拟衰老和神经变性的药物对膜组成和通透性的影响,以及它们对胰岛素信号传导的影响。小鼠神经母细胞瘤2a细胞系(neuro2a)分别用STZ (6 h)和BSO (24 h)处理。MTT法测定细胞活力。胆固醇和鞘磷脂含量用商用试剂盒定量,膜极性用Laurdan探针评估。采用qPCR分析Srebf2和Cyp46a1基因的表达情况。免疫印迹法检测胰岛素信号通路蛋白。STZ处理降低了神经元胆固醇含量,下调了Srebf2和Cyp46a1基因的表达,减少了膜的包装。相比之下,bso处理的细胞鞘磷脂含量增加,Srebf2和Cyp46a1基因表达上调,膜堆积减少。两种治疗方法都诱导胰岛素抵抗状态,我们将其归因于膜环境的改变。
{"title":"Streptozotocin and L-Buthionine-Sulfoximine Decrease Neuron Membrane Lipid Packing and Alter Insulin Signaling.","authors":"Yandara A Martins, Camila A E F Cardinali, Andressa P Costa, Andréa S Torrão","doi":"10.1007/s12640-025-00749-z","DOIUrl":"10.1007/s12640-025-00749-z","url":null,"abstract":"<p><p>Membrane composition, permeability and fluidity are essential for proper cellular function. According to the membrane aging hypothesis, aging-related diseases, including neurodegenerative disorders, arise from the aging of cell membranes. Membrane proteins, such as the insulin receptor, rely on an optimal membrane environment for proper partitioning and functionality. Our goal was to investigate the effects of streptozotocin (STZ) and L-buthionine-sulfoximine (BSO), two commonly used agents to model aging and neurodegeneration, on membrane composition and permeability, as well as their impact on insulin signaling. Mouse neuroblastoma 2a cell line (neuro-2a) were treated with STZ (6 h) and BSO (24 h). Cell viability was assessed by the MTT assay. Cholesterol and sphingomyelin content were quantified by commercial kits, while membrane polarity was evaluated with the Laurdan probe. Gene expression of Srebf2 and Cyp46a1 was analyzed by qPCR. Proteins from the insulin signaling pathway were examined by immunoblotting. STZ treatment reduced neuronal cholesterol content, downregulated Srebf2 and Cyp46a1 gene expression, and decreased membrane packing. In contrast, BSO-treated cells exhibited increased sphingomyelin content, upregulated Srebf2 and Cyp46a1 gene expression, and decreased membrane packing. Both treatments induced an insulin-resistant state, which we attribute to alterations in the membrane environment.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"43 3","pages":"24"},"PeriodicalIF":2.9,"publicationDate":"2025-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144174291","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Protective Effect of Annexin A1 on Autophagy Via the CAMK2/BECN1 Signaling Pathway in PC12 Cells Stimulated with H2O2. 膜联蛋白A1通过CAMK2/BECN1信号通路对H2O2刺激PC12细胞自噬的保护作用
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2025-04-24 DOI: 10.1007/s12640-025-00745-3
Shan Hui, Yi Long, Lemei Zhu, Junpei Hu, Qing Zheng, Lihui Liang

This study investigates the potential protective role of annexin A1 (ANXA1) in cell models of H2O2-induced Alzheimer's disease. PC12 cells exposed to varying concentrations of H2O2 exhibited a dose-dependent decrease in cell viability. H2O2 exposure led to elevated reactive oxygen species (ROS) levels, reduced superoxide dismutase (SOD) and catalase (CAT) activities, and a decline in ANXA1 protein expression. Under oxidative stress, ANXA1 overexpression increased cell viability, reduced apoptosis rate, enhanced the expression of microtubule-associated protein 3 (LC3) II/I while reducing phosphorylated calcium/calmodulin-dependent protein kinase II (p-CAMK2)/CAMK2 and phosphorylated beclin 1 (p-BECN1)/BECN1. Conversely, ANXA1 knockdown produced contrasting effects. Overexpression of ANXA1, accompanied by administration of KN-93 (a competitive inhibitor of CAMK2), can synergistically diminished p-CAMK2/CAMK2 and p-BECN1/BECN1 levels while significantly increasing LC3 II/I levels, autophagosomes, and autolysosomes. In conclusion, ANXA1 demonstrated a protective role in H2O2-induced oxidative stress damage model in PC12 cells by inhibiting the CAMK2/BECN1 signaling pathway and enhancing autophagy.

本研究探讨了膜联蛋白A1 (ANXA1)在h2o2诱导的阿尔茨海默病细胞模型中的潜在保护作用。暴露于不同浓度H2O2的PC12细胞表现出剂量依赖性的细胞活力下降。H2O2暴露导致活性氧(ROS)水平升高,超氧化物歧化酶(SOD)和过氧化氢酶(CAT)活性降低,ANXA1蛋白表达下降。氧化应激下,ANXA1过表达可提高细胞活力,降低细胞凋亡率,增强微管相关蛋白3 (LC3) II/I的表达,降低磷酸化钙/钙调素依赖性蛋白激酶II (p-CAMK2)/CAMK2和磷酸化beclin 1 (p-BECN1)/BECN1。相反,ANXA1敲低会产生相反的效果。过表达ANXA1,同时给予km -93 (CAMK2的竞争性抑制剂),可以协同降低p-CAMK2/CAMK2和p-BECN1/BECN1水平,同时显著增加LC3 II/I水平、自噬体和自溶体。综上所述,ANXA1通过抑制CAMK2/BECN1信号通路和增强自噬,在h2o2诱导的PC12细胞氧化应激损伤模型中发挥保护作用。
{"title":"The Protective Effect of Annexin A1 on Autophagy Via the CAMK2/BECN1 Signaling Pathway in PC12 Cells Stimulated with H<sub>2</sub>O<sub>2</sub>.","authors":"Shan Hui, Yi Long, Lemei Zhu, Junpei Hu, Qing Zheng, Lihui Liang","doi":"10.1007/s12640-025-00745-3","DOIUrl":"10.1007/s12640-025-00745-3","url":null,"abstract":"<p><p>This study investigates the potential protective role of annexin A1 (ANXA1) in cell models of H<sub>2</sub>O<sub>2</sub>-induced Alzheimer's disease. PC12 cells exposed to varying concentrations of H<sub>2</sub>O<sub>2</sub> exhibited a dose-dependent decrease in cell viability. H<sub>2</sub>O<sub>2</sub> exposure led to elevated reactive oxygen species (ROS) levels, reduced superoxide dismutase (SOD) and catalase (CAT) activities, and a decline in ANXA1 protein expression. Under oxidative stress, ANXA1 overexpression increased cell viability, reduced apoptosis rate, enhanced the expression of microtubule-associated protein 3 (LC3) II/I while reducing phosphorylated calcium/calmodulin-dependent protein kinase II (p-CAMK2)/CAMK2 and phosphorylated beclin 1 (p-BECN1)/BECN1. Conversely, ANXA1 knockdown produced contrasting effects. Overexpression of ANXA1, accompanied by administration of KN-93 (a competitive inhibitor of CAMK2), can synergistically diminished p-CAMK2/CAMK2 and p-BECN1/BECN1 levels while significantly increasing LC3 II/I levels, autophagosomes, and autolysosomes. In conclusion, ANXA1 demonstrated a protective role in H<sub>2</sub>O<sub>2</sub>-induced oxidative stress damage model in PC12 cells by inhibiting the CAMK2/BECN1 signaling pathway and enhancing autophagy.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"43 3","pages":"23"},"PeriodicalIF":2.9,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144004156","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exploring Retinal Neurodegeneration in Alzheimer's Disease: A Molecular and Cellular Perspective. 从分子和细胞的角度探讨阿尔茨海默病的视网膜神经变性。
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2025-04-12 DOI: 10.1007/s12640-025-00744-4
Rishika Dhapola, Prajjwal Sharma, Sneha Kumari, Balachandar Vellingiri, Bikash Medhi, Dibbanti HariKrishnaReddy

Increasing evidence of ocular impairments in Alzheimer's disease (AD) has drawn the attention of researchers worldwide towards retinal neurodegeneration in AD. The AD-associated changes observed in the retina include visual discrepancies, pupil size modulations, retinal nerve layer changes, retinal blood flow alterations and histopathological changes. The brain cells that act as pathological triggers for the progression of retinal neurodegeneration associated with AD are microglia, astrocytes and neurons. Various molecular pathways lead to structural and functional abnormalities in the retina, significantly affecting the brain including Aβ accumulation, apoptosis, inflammation and oxidative stress. Therapeutic agents under development that ameliorate disease conditions by targeting retinal anomalies include mesenchymal stem cell-conditioned media, BDNF, glatiramer acetate, salvianolic acid B, Lycium barbarum extract and exosomes. Investigating real-time alterations in the retina in AD may not only affect diagnostic approaches but also help to clarify neuropathological pathways and offer helpful measurements for assessing novel therapeutic approaches for AD.

越来越多的证据表明阿尔茨海默病(AD)的眼部损害引起了全世界研究者对阿尔茨海默病视网膜神经变性的关注。在视网膜上观察到的ad相关改变包括视觉差异、瞳孔大小调节、视网膜神经层改变、视网膜血流改变和组织病理学改变。作为与AD相关的视网膜神经变性进展的病理触发的脑细胞是小胶质细胞、星形胶质细胞和神经元。多种分子通路导致视网膜结构和功能异常,显著影响大脑,包括Aβ积累、细胞凋亡、炎症和氧化应激。通过靶向视网膜异常改善疾病状况的正在开发的治疗剂包括间充质干细胞条件培养基、BDNF、醋酸格拉替默、丹酚酸B、枸杞提取物和外泌体。研究阿尔茨海默病视网膜的实时变化不仅可以影响诊断方法,还有助于阐明神经病理通路,并为评估阿尔茨海默病的新治疗方法提供有用的测量方法。
{"title":"Exploring Retinal Neurodegeneration in Alzheimer's Disease: A Molecular and Cellular Perspective.","authors":"Rishika Dhapola, Prajjwal Sharma, Sneha Kumari, Balachandar Vellingiri, Bikash Medhi, Dibbanti HariKrishnaReddy","doi":"10.1007/s12640-025-00744-4","DOIUrl":"https://doi.org/10.1007/s12640-025-00744-4","url":null,"abstract":"<p><p>Increasing evidence of ocular impairments in Alzheimer's disease (AD) has drawn the attention of researchers worldwide towards retinal neurodegeneration in AD. The AD-associated changes observed in the retina include visual discrepancies, pupil size modulations, retinal nerve layer changes, retinal blood flow alterations and histopathological changes. The brain cells that act as pathological triggers for the progression of retinal neurodegeneration associated with AD are microglia, astrocytes and neurons. Various molecular pathways lead to structural and functional abnormalities in the retina, significantly affecting the brain including Aβ accumulation, apoptosis, inflammation and oxidative stress. Therapeutic agents under development that ameliorate disease conditions by targeting retinal anomalies include mesenchymal stem cell-conditioned media, BDNF, glatiramer acetate, salvianolic acid B, Lycium barbarum extract and exosomes. Investigating real-time alterations in the retina in AD may not only affect diagnostic approaches but also help to clarify neuropathological pathways and offer helpful measurements for assessing novel therapeutic approaches for AD.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"43 2","pages":"22"},"PeriodicalIF":2.9,"publicationDate":"2025-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143974823","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Role and Mechanism of TRIM13 Regulation of TRAF6 Ubiquitination in the Synergy of Inflammatory Responses and Neurotoxicity Induced by METH and HIV- 1 Tat Protein in Astrocytes. TRIM13调控TRAF6泛素化在冰毒和HIV- 1 Tat蛋白诱导的星形胶质细胞炎症反应和神经毒性协同作用中的作用及机制
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2025-04-07 DOI: 10.1007/s12640-025-00743-5
Yi Tan, Lin Miao, Chan Wang, Haowei Wang, Yi Li, Yizhen Huang, Hanxin Teng, Yunqing Tian, Genmeng Yang, Xiaofeng Zeng, Juan Li

Methamphetamine (METH) abuse and HIV infection are major public health concerns worldwide. While both METH and HIV- 1 Tat proteins can induce neurotoxicity and synergistic effects on the nervous system, the mechanisms by which they act synergistically remain unclear. Our recent research shows that neuroinflammation plays an important role in neurotoxicity induced by METH and HIV- 1 Tat proteins, but the regulatory mechanism has not been clarified. Tripartite Motif Containing 13 (TRIM13) is a protein known to regulate the inflammatory response through ubiquitination of Tumor Necrosis Factor Receptor Associated Factor 6 (TRAF6). This study investigated the role of TRIM13 and TRAF6 in the inflammatory response of U- 87 MG cells induced by METH and HIV- 1 Tat proteins. U- 87 MG cells were treated with 2 mM METH and/or 100 nM HIV- 1 Tat protein. Western blot (WB), immunofluorescence (IF), and co-immunoprecipitation (Co-IP) experiments were employed to elucidate the role of TRIM13 and TRAF6. The results demonstrated that METH and HIV- 1 Tat protein could synergistically induce an inflammatory response in U- 87 MG cells. Furthermore, the knockdown of TRIM13 significantly enhanced this inflammatory response, while the inhibition of TRAF6 significantly weakened it. Additionally, the study revealed that TRIM13 could degrade TRAF6 via ubiquitination. In conclusion, this study suggests that TRIM13 regulates TRAF6 ubiquitination to dampen the inflammatory response of U- 87 MG cells induced by METH and HIV- 1 Tat proteins. These findings highlight TRIM13 and TRAF6 as potential targets for therapeutic intervention in the context of METH and HIV- 1 Tat protein-induced inflammatory responses and neurotoxic effects.

甲基苯丙胺(冰毒)滥用和艾滋病毒感染是全世界主要的公共卫生问题。虽然甲基苯丙胺和HIV- 1 Tat蛋白都可以诱导神经毒性和对神经系统的协同作用,但它们协同作用的机制尚不清楚。我们最近的研究表明,神经炎症在甲基安非他明和HIV- 1tat蛋白诱导的神经毒性中起重要作用,但其调控机制尚未明确。Tripartite Motif Containing 13 (TRIM13)是一种已知通过肿瘤坏死因子受体相关因子6 (TRAF6)泛素化调节炎症反应的蛋白。本研究探讨TRIM13和TRAF6在甲基安非他明和HIV- 1tat蛋白诱导的U- 87 MG细胞炎症反应中的作用。用2 mM甲基安非他明和/或100 nM HIV- 1tat蛋白处理U- 87 MG细胞。采用Western blot (WB)、免疫荧光(IF)和共免疫沉淀(Co-IP)实验来阐明TRIM13和TRAF6的作用。结果表明,甲基苯丙胺和HIV- 1tat蛋白可协同诱导U- 87mg细胞的炎症反应。此外,TRIM13的敲低显著增强了这种炎症反应,而TRAF6的抑制显著减弱了这种炎症反应。此外,该研究还发现TRIM13可以通过泛素化降解TRAF6。综上所述,本研究提示TRIM13调节TRAF6泛素化,可抑制METH和HIV- 1tat蛋白诱导的U- 87 MG细胞的炎症反应。这些发现强调TRIM13和TRAF6是在甲基甲氧胺和HIV- 1tat蛋白诱导的炎症反应和神经毒性作用的背景下进行治疗干预的潜在靶点。
{"title":"The Role and Mechanism of TRIM13 Regulation of TRAF6 Ubiquitination in the Synergy of Inflammatory Responses and Neurotoxicity Induced by METH and HIV- 1 Tat Protein in Astrocytes.","authors":"Yi Tan, Lin Miao, Chan Wang, Haowei Wang, Yi Li, Yizhen Huang, Hanxin Teng, Yunqing Tian, Genmeng Yang, Xiaofeng Zeng, Juan Li","doi":"10.1007/s12640-025-00743-5","DOIUrl":"10.1007/s12640-025-00743-5","url":null,"abstract":"<p><p>Methamphetamine (METH) abuse and HIV infection are major public health concerns worldwide. While both METH and HIV- 1 Tat proteins can induce neurotoxicity and synergistic effects on the nervous system, the mechanisms by which they act synergistically remain unclear. Our recent research shows that neuroinflammation plays an important role in neurotoxicity induced by METH and HIV- 1 Tat proteins, but the regulatory mechanism has not been clarified. Tripartite Motif Containing 13 (TRIM13) is a protein known to regulate the inflammatory response through ubiquitination of Tumor Necrosis Factor Receptor Associated Factor 6 (TRAF6). This study investigated the role of TRIM13 and TRAF6 in the inflammatory response of U- 87 MG cells induced by METH and HIV- 1 Tat proteins. U- 87 MG cells were treated with 2 mM METH and/or 100 nM HIV- 1 Tat protein. Western blot (WB), immunofluorescence (IF), and co-immunoprecipitation (Co-IP) experiments were employed to elucidate the role of TRIM13 and TRAF6. The results demonstrated that METH and HIV- 1 Tat protein could synergistically induce an inflammatory response in U- 87 MG cells. Furthermore, the knockdown of TRIM13 significantly enhanced this inflammatory response, while the inhibition of TRAF6 significantly weakened it. Additionally, the study revealed that TRIM13 could degrade TRAF6 via ubiquitination. In conclusion, this study suggests that TRIM13 regulates TRAF6 ubiquitination to dampen the inflammatory response of U- 87 MG cells induced by METH and HIV- 1 Tat proteins. These findings highlight TRIM13 and TRAF6 as potential targets for therapeutic intervention in the context of METH and HIV- 1 Tat protein-induced inflammatory responses and neurotoxic effects.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"43 2","pages":"21"},"PeriodicalIF":2.9,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143795818","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Associations among Angiotensin-Converting Enzyme, Neuroinflammation, and Cerebrospinal Fluid Biomarkers of Alzheimer's Disease in Non-Dementia Adults. 非痴呆成人阿尔茨海默病血管紧张素转换酶、神经炎症和脑脊液生物标志物之间的关系
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2025-04-04 DOI: 10.1007/s12640-025-00740-8
Lan-Yang Wang, Hao Hu, Ze-Hu Sheng, He-Ying Hu, Ya-Nan Ou, Fan Guo, Yang-Ke Zhu, Lan Tan

Recent studies have identified the angiotensin-converting enzyme (ACE) gene as a potential candidate influencing Alzheimer's disease (AD) risk. It is crucial to investigate the impact of ACE on AD pathology and its underlying mechanisms. A total of 450 non-demented participants from the Alzheimer's disease Neuroimaging Initiative (ADNI) with data on cerebrospinal fluid (CSF) ACE, AD core biomarkers and inflammation-related biomarkers were included. Multiple linear regression was used to assess the associations among CSF ACE, AD core biomarkers and inflammation-related biomarkers. And we used the mediation models to investigate the potential mechanisms through which ACE influenced AD pathology. The results of multiple linear regression were shown that CSF ACE was significantly correlated with CSF Aβ42, P-tau, T-tau (all P < 0.001), and inflammation-related biomarkers (soluble triggering receptor expressed on myeloid cells 2 [sTREM2], progranulin [PGRN], glial fibrillary acidic protein [GFAP], transforming growth factor [TGF]-β1, TGF-β2, TGF-β3, tumor necrosis factor [TNF]-R1, TNF-R2, TNF-α, interleukin [IL]-21, IL-6, IL-7, IL-9, IL-10, IL-12p40, vascular cell adhesion molecule-1 [VCAM-1], and intercellular adhesion molecule-1 [ICAM-1]) (all P < 0.05). In addition, the mediation analysis results showed that the association of CSF ACE and inflammation-related biomarkers (sTREM2, PGRN, TGF-β1, TGF-β2, TNFR1, IL-6, IL-7, IL-9, and VCAM-1) mediated the correlation of CSF Aβ42 with P-tau. Our findings show that CSF ACE and neuroinflammation are correlated and that their correlation mediates the link between Aβ pathology and P-tau. This suggests ACE may play a significant role in the progression from Aβ pathology to tau pathology.

最近的研究已经确定血管紧张素转换酶(ACE)基因是影响阿尔茨海默病(AD)风险的潜在候选基因。研究ACE对AD病理的影响及其潜在机制至关重要。共有450名来自阿尔茨海默病神经影像学倡议(ADNI)的非痴呆参与者,其脑脊液(CSF) ACE、AD核心生物标志物和炎症相关生物标志物的数据被纳入研究。采用多元线性回归评估脑脊液ACE、AD核心生物标志物和炎症相关生物标志物之间的相关性。我们使用中介模型来探讨ACE影响AD病理的潜在机制。多元线性回归结果显示,脑脊液ACE与脑脊液Aβ42、P-tau、T-tau显著相关(P 42均与P-tau相关)。我们的研究结果表明脑脊液ACE和神经炎症是相关的,它们的相关性介导了Aβ病理和P-tau之间的联系。这表明ACE可能在从a β病理到tau病理的进展中起重要作用。
{"title":"Associations among Angiotensin-Converting Enzyme, Neuroinflammation, and Cerebrospinal Fluid Biomarkers of Alzheimer's Disease in Non-Dementia Adults.","authors":"Lan-Yang Wang, Hao Hu, Ze-Hu Sheng, He-Ying Hu, Ya-Nan Ou, Fan Guo, Yang-Ke Zhu, Lan Tan","doi":"10.1007/s12640-025-00740-8","DOIUrl":"10.1007/s12640-025-00740-8","url":null,"abstract":"<p><p>Recent studies have identified the angiotensin-converting enzyme (ACE) gene as a potential candidate influencing Alzheimer's disease (AD) risk. It is crucial to investigate the impact of ACE on AD pathology and its underlying mechanisms. A total of 450 non-demented participants from the Alzheimer's disease Neuroimaging Initiative (ADNI) with data on cerebrospinal fluid (CSF) ACE, AD core biomarkers and inflammation-related biomarkers were included. Multiple linear regression was used to assess the associations among CSF ACE, AD core biomarkers and inflammation-related biomarkers. And we used the mediation models to investigate the potential mechanisms through which ACE influenced AD pathology. The results of multiple linear regression were shown that CSF ACE was significantly correlated with CSF Aβ<sub>42</sub>, P-tau, T-tau (all P < 0.001), and inflammation-related biomarkers (soluble triggering receptor expressed on myeloid cells 2 [sTREM2], progranulin [PGRN], glial fibrillary acidic protein [GFAP], transforming growth factor [TGF]-β1, TGF-β2, TGF-β3, tumor necrosis factor [TNF]-R1, TNF-R2, TNF-α, interleukin [IL]-21, IL-6, IL-7, IL-9, IL-10, IL-12p40, vascular cell adhesion molecule-1 [VCAM-1], and intercellular adhesion molecule-1 [ICAM-1]) (all P < 0.05). In addition, the mediation analysis results showed that the association of CSF ACE and inflammation-related biomarkers (sTREM2, PGRN, TGF-β1, TGF-β2, TNFR1, IL-6, IL-7, IL-9, and VCAM-1) mediated the correlation of CSF Aβ<sub>42</sub> with P-tau. Our findings show that CSF ACE and neuroinflammation are correlated and that their correlation mediates the link between Aβ pathology and P-tau. This suggests ACE may play a significant role in the progression from Aβ pathology to tau pathology.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"43 2","pages":"20"},"PeriodicalIF":2.9,"publicationDate":"2025-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143788375","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exploring the potential role of microtubule associated proteins-2 in the pathogenesis of HIV associated neurocognitive disorders. 探讨微管相关蛋白-2在HIV相关神经认知障碍发病机制中的潜在作用。
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2025-04-03 DOI: 10.1007/s12640-025-00739-1
Melanie K Becher, Valeria Avdoshina, Lee A Campbell, Italo Mocchetti

HIV-associated neurocognitive disorder (HAND) persists in people living with HIV (PLWH) despite antiretroviral therapy. HAND is characterized by synapto-dendritic damage, yet the cause of this pathology is still under investigation. Various viral proteins, including the envelope protein gp120, have been proposed to be the leading neurotoxic agents underlying HIV-mediated neuronal degeneration. Gp120 has been shown to bind to neuronal microtubules (MTs) and impair their functions. The dynamic properties of MTs are modulated by microtubule-associated proteins (MAP), including MAP2, which is particularly abundant in dendrites. This review article explores how gp120 could be altering the function of the neuronal cytoskeleton by affecting MAP2. These effects may serve as a causal link between viral proteins and HAND pathology.

尽管抗逆转录病毒治疗,HIV相关神经认知障碍(HAND)仍然存在于HIV感染者(PLWH)中。HAND的特点是突触-树突损伤,但这种病理的原因仍在研究中。包括包膜蛋白gp120在内的各种病毒蛋白被认为是hiv介导的神经元变性的主要神经毒性因子。Gp120已被证明与神经元微管(MTs)结合并损害其功能。MTs的动态特性是由微管相关蛋白(MAP)调控的,其中包括在树突中特别丰富的MAP2。这篇综述文章探讨了gp120如何通过影响MAP2来改变神经元细胞骨架的功能。这些影响可能是病毒蛋白与HAND病理之间的因果关系。
{"title":"Exploring the potential role of microtubule associated proteins-2 in the pathogenesis of HIV associated neurocognitive disorders.","authors":"Melanie K Becher, Valeria Avdoshina, Lee A Campbell, Italo Mocchetti","doi":"10.1007/s12640-025-00739-1","DOIUrl":"10.1007/s12640-025-00739-1","url":null,"abstract":"<p><p>HIV-associated neurocognitive disorder (HAND) persists in people living with HIV (PLWH) despite antiretroviral therapy. HAND is characterized by synapto-dendritic damage, yet the cause of this pathology is still under investigation. Various viral proteins, including the envelope protein gp120, have been proposed to be the leading neurotoxic agents underlying HIV-mediated neuronal degeneration. Gp120 has been shown to bind to neuronal microtubules (MTs) and impair their functions. The dynamic properties of MTs are modulated by microtubule-associated proteins (MAP), including MAP2, which is particularly abundant in dendrites. This review article explores how gp120 could be altering the function of the neuronal cytoskeleton by affecting MAP2. These effects may serve as a causal link between viral proteins and HAND pathology.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"43 2","pages":"19"},"PeriodicalIF":2.9,"publicationDate":"2025-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143772353","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Kynurenine Pathway in Epilepsy: Unraveling Its Role in Glutamate Excitotoxicity, GABAergic Dysregulation, Neuroinflammation, and Mitochondrial Dysfunction. 癫痫中的犬尿氨酸通路:揭示犬尿氨酸通路在谷氨酸兴奋毒性、GABA能失调、神经炎症和线粒体功能障碍中的作用。
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2025-03-28 DOI: 10.1007/s12640-025-00738-2
Manpreet Kaur, Pratyush Porel, Royal Patel, Khadga Raj Aran

Epilepsy is a chronic noncommunicable neurological disorder characterized by recurrent seizures and ranks as the seventh most prevalent neurological disease globally. According to the Global Burden of Disease report, 3.40 billion people were affected by epilepsy in 2021. The pathophysiology of epilepsy states that a disturbed balance between excitatory and inhibitory signaling at the synaptic level, which can cause seizure activity, is similar across epilepsies and includes mitochondrial dysfunction, neuroinflammation, and kynurenine metabolites such as kynurenic acid and quinolinic acid. The kynurenine pathway (KP) is the major metabolic pathway in which tryptophan (TRP) is the key precursor which is further converted into a variety of neuroactive substances that can have both neurotoxic metabolites (Quinolinic acid) and neuroprotective metabolites such as kynurenic acid, and picolinic acid. KP plays a significant role in the brain such as the metabolism of TRP, the production of metabolites, and its impact on aging. However, higher concentrations of kynurenine and its metabolites, such as quinolinic acid may increase the frequency and intensity of seizures, and dysregulation of the KP has been linked to the pathophysiology of epilepsy. Concurrently, glutamate and GABA signaling is altered by neuroinflammatory processes linked to epilepsy, which results in excitotoxic neuronal damage. This review aims to provide novel therapeutic strategies that might improve the prognosis of individuals with epilepsy and related disorders by elucidating the mechanisms underlying KP dysregulation in these circumstances. To develop targeted therapies for CNS disorders characterized by inflammation and seizures, it is essential to understand how kynurenine metabolites both promote and prevent excitotoxicity.

癫痫是一种以反复发作为特征的慢性非传染性神经系统疾病,是全球第七大神经系统疾病。根据《全球疾病负担报告》,2021 年将有 34 亿人受到癫痫的影响。癫痫的病理生理学认为,突触水平的兴奋信号和抑制信号之间的平衡失调可导致癫痫发作活动,不同癫痫的病理生理学相似,包括线粒体功能障碍、神经炎症以及犬尿氨酸代谢物(如犬尿酸和喹啉酸)。犬尿氨酸途径(KP)是主要的代谢途径,其中色氨酸(TRP)是关键的前体物质,可进一步转化为多种神经活性物质,这些物质既有神经毒性代谢物(喹啉酸),也有神经保护性代谢物,如犬尿氨酸和吡啶甲酸。KP 在大脑中发挥着重要作用,如 TRP 的代谢、代谢产物的产生以及对衰老的影响。然而,较高浓度的犬尿氨酸及其代谢物(如喹啉酸)可能会增加癫痫发作的频率和强度,KP 的失调与癫痫的病理生理学有关。与此同时,与癫痫有关的神经炎症过程会改变谷氨酸和 GABA 信号转导,从而导致兴奋毒性神经元损伤。本综述旨在通过阐明这些情况下 KP 失调的内在机制,提供可能改善癫痫和相关疾病患者预后的新型治疗策略。要开发针对以炎症和癫痫发作为特征的中枢神经系统疾病的靶向疗法,就必须了解犬尿氨酸代谢物是如何促进和预防兴奋毒性的。
{"title":"Kynurenine Pathway in Epilepsy: Unraveling Its Role in Glutamate Excitotoxicity, GABAergic Dysregulation, Neuroinflammation, and Mitochondrial Dysfunction.","authors":"Manpreet Kaur, Pratyush Porel, Royal Patel, Khadga Raj Aran","doi":"10.1007/s12640-025-00738-2","DOIUrl":"10.1007/s12640-025-00738-2","url":null,"abstract":"<p><p>Epilepsy is a chronic noncommunicable neurological disorder characterized by recurrent seizures and ranks as the seventh most prevalent neurological disease globally. According to the Global Burden of Disease report, 3.40 billion people were affected by epilepsy in 2021. The pathophysiology of epilepsy states that a disturbed balance between excitatory and inhibitory signaling at the synaptic level, which can cause seizure activity, is similar across epilepsies and includes mitochondrial dysfunction, neuroinflammation, and kynurenine metabolites such as kynurenic acid and quinolinic acid. The kynurenine pathway (KP) is the major metabolic pathway in which tryptophan (TRP) is the key precursor which is further converted into a variety of neuroactive substances that can have both neurotoxic metabolites (Quinolinic acid) and neuroprotective metabolites such as kynurenic acid, and picolinic acid. KP plays a significant role in the brain such as the metabolism of TRP, the production of metabolites, and its impact on aging. However, higher concentrations of kynurenine and its metabolites, such as quinolinic acid may increase the frequency and intensity of seizures, and dysregulation of the KP has been linked to the pathophysiology of epilepsy. Concurrently, glutamate and GABA signaling is altered by neuroinflammatory processes linked to epilepsy, which results in excitotoxic neuronal damage. This review aims to provide novel therapeutic strategies that might improve the prognosis of individuals with epilepsy and related disorders by elucidating the mechanisms underlying KP dysregulation in these circumstances. To develop targeted therapies for CNS disorders characterized by inflammation and seizures, it is essential to understand how kynurenine metabolites both promote and prevent excitotoxicity.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"43 2","pages":"18"},"PeriodicalIF":2.9,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143736069","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mechanistic Analysis of Decabromodiphenyl Ether-Induced Neurotoxicity in Humans Using Network Toxicology and Molecular Docking. 基于网络毒理学和分子对接的十溴联苯醚致人神经毒性机理分析。
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2025-03-24 DOI: 10.1007/s12640-025-00741-7
Fuat Karakuş, Burak Kuzu

Commercial decabromodiphenyl ether (c-decaBDE) is a widely used additive flame retardant in textiles and plastics. This formulation predominantly consists of the congener BDE-209, with trace amounts of other brominated diphenyl ether congeners, such as nonabromodiphenyl ether and octabromodiphenyl ether. Recognized as a persistent organic pollutant due to its potential for long-range environmental transport, c-decaBDE poses significant environmental threats and serious human health risks, including endocrine, reproductive, developmental, and neurotoxic effects. The mechanisms underlying its neurotoxicity remain largely undefined. This study investigates the neurotoxic effects of BDE-209 in humans through network toxicology, multi-level bioinformatics approaches, and molecular docking analyses. Prediction results indicate that BDE-209 can cross the blood-brain barrier, entering the central nervous system and inducing neurotoxic effects. A comprehensive analysis has identified 294 potential targets linked to the neurotoxicity induced by BDE-209. Gene-gene interaction and pathway enrichment analyses revealed significant associations related to cellular responses to chemical stress and synaptic transmission. Further investigation of protein-protein interactions, combined with centrality analysis, identified 14 hub targets, including CaMK-II alpha, PSD-95, GluR-1, and GluN2B, as key proteins in this process. Molecular docking results indicate that BDE-209 exhibits a stronger binding affinity to GluN2B, a subunit of the N-methyl-D-aspartate (NMDA) receptors, compared to other key targets. These findings suggest that BDE-209 may disrupt the function of GluN2B-containing NMDA receptors, potentially leading to their inhibition. Such inhibition could result in reduced excitatory neurotransmission, impairing synaptic potentiation and plasticity, and ultimately contributing to neurotoxicity.

商用十溴联苯醚(c-decaBDE)是一种广泛应用于纺织品和塑料的阻燃剂。该制剂主要由同系物BDE-209和微量其他溴化二苯醚同系物组成,如非溴二苯醚和八溴二苯醚。十溴二苯醚是一种持久性有机污染物,具有远距离环境迁移的潜力,对环境构成重大威胁,对人类健康构成严重风险,包括内分泌、生殖、发育和神经毒性影响。其神经毒性的机制在很大程度上仍未明确。本研究通过网络毒理学、多层次生物信息学方法和分子对接分析研究了BDE-209对人类的神经毒性作用。预测结果表明,BDE-209可穿过血脑屏障,进入中枢神经系统,引起神经毒性作用。一项综合分析已经确定了294个与BDE-209引起的神经毒性有关的潜在靶点。基因-基因相互作用和通路富集分析显示,细胞对化学应激和突触传递的反应存在显著关联。进一步研究蛋白-蛋白相互作用,结合中心性分析,确定了14个枢纽靶点,包括CaMK-II α, PSD-95, GluR-1和GluN2B,是这一过程的关键蛋白。分子对接结果表明,与其他关键靶点相比,BDE-209与n -甲基- d -天冬氨酸(NMDA)受体亚基GluN2B具有更强的结合亲和力。这些发现表明,BDE-209可能破坏glun2b含NMDA受体的功能,可能导致其抑制。这种抑制可能导致兴奋性神经传递减少,损害突触增强和可塑性,最终导致神经毒性。
{"title":"Mechanistic Analysis of Decabromodiphenyl Ether-Induced Neurotoxicity in Humans Using Network Toxicology and Molecular Docking.","authors":"Fuat Karakuş, Burak Kuzu","doi":"10.1007/s12640-025-00741-7","DOIUrl":"10.1007/s12640-025-00741-7","url":null,"abstract":"<p><p>Commercial decabromodiphenyl ether (c-decaBDE) is a widely used additive flame retardant in textiles and plastics. This formulation predominantly consists of the congener BDE-209, with trace amounts of other brominated diphenyl ether congeners, such as nonabromodiphenyl ether and octabromodiphenyl ether. Recognized as a persistent organic pollutant due to its potential for long-range environmental transport, c-decaBDE poses significant environmental threats and serious human health risks, including endocrine, reproductive, developmental, and neurotoxic effects. The mechanisms underlying its neurotoxicity remain largely undefined. This study investigates the neurotoxic effects of BDE-209 in humans through network toxicology, multi-level bioinformatics approaches, and molecular docking analyses. Prediction results indicate that BDE-209 can cross the blood-brain barrier, entering the central nervous system and inducing neurotoxic effects. A comprehensive analysis has identified 294 potential targets linked to the neurotoxicity induced by BDE-209. Gene-gene interaction and pathway enrichment analyses revealed significant associations related to cellular responses to chemical stress and synaptic transmission. Further investigation of protein-protein interactions, combined with centrality analysis, identified 14 hub targets, including CaMK-II alpha, PSD-95, GluR-1, and GluN2B, as key proteins in this process. Molecular docking results indicate that BDE-209 exhibits a stronger binding affinity to GluN2B, a subunit of the N-methyl-D-aspartate (NMDA) receptors, compared to other key targets. These findings suggest that BDE-209 may disrupt the function of GluN2B-containing NMDA receptors, potentially leading to their inhibition. Such inhibition could result in reduced excitatory neurotransmission, impairing synaptic potentiation and plasticity, and ultimately contributing to neurotoxicity.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"43 2","pages":"17"},"PeriodicalIF":2.9,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11930881/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143692814","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Oxytetracycline and its Non-Antibiotic Derivative DOT Protect Midbrain Dopamine Neurons from Iron-Driven Oxidative Damage. 土霉素及其非抗生素衍生物DOT保护中脑多巴胺神经元免受铁驱动的氧化损伤。
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2025-03-22 DOI: 10.1007/s12640-025-00742-6
Thaís Antonia Alves Fernandes, Aurore Tourville, Ismaila Ciss, Rafaela Ribeiro Silva, Bianca Andretto de Mattos, Maurício Dos Santos Pereira, Maxime Oblaza, Jean-Michel Brunel, Laurent Ferrié, Rita Raisman-Vozari, Bruno Figadère, Elaine Del-Bel, Patrick Pierre Michel

This study aimed to investigate the neuroprotective potential of the tetracycline (TC) antibiotic oxytetracycline (OT) and its non-antibiotic derivative 4-dedimethylamino 12a-deoxy-oxytetracycline (DOT), in experimental conditions that mimic the gradual loss of dopamine (DA) neurons in Parkinson's disease (PD). Specifically, we established a model system of mouse midbrain cultures where DA neurons progressively die when exposed to an iron-containing medium. We found that OT (EC50 = 0.25µM) and DOT (EC50 = 0.34µM) efficiently protected DA neurons from degeneration, with these effects observable until advanced stages of neurodegeneration. The reference antibiotic TC doxycycline (DOX) also exhibited protective effects in this context. Importantly, DA neurons rescued by OT, DOT, and DOX retained their capacity to accumulate and release DA, indicating full functional integrity. Additionally, molecules with iron-chelating properties (apotransferrin, desferoxamine), as well as inhibitors of lipid peroxidation and ferroptosis (Trolox, Liproxstatin-1), could replicate the rescue of DA neurons provided by OT, DOT, and DOX. Live-cell imaging studies showed that test TCs and other neuroprotective molecules prevented the emission of intracellular reactive oxygen species and the associated disruption of the mitochondrial membrane potential. However, neither OT, DOT, nor DOX could protect DA neurons from selective mitochondrial poisoning by 1-methyl-4-phenylpyridinium. This suggests that test TCs may be protective against iron-mediated damage through a mechanism not directly involving mitochondria. Overall, we demonstrate that OT and DOT possess promising properties that could be useful for combating PD neurodegeneration. However, the absence of antimicrobial activity makes DOT a better candidate drug compared to its parent compound OT.

本研究旨在研究四环素(TC)抗生素土霉素(OT)及其非抗生素衍生物 4-二甲氨基 12a-脱氧土霉素(DOT)在模拟帕金森病(PD)中多巴胺(DA)神经元逐渐丧失的实验条件下的神经保护潜力。具体来说,我们建立了一个小鼠中脑培养模型系统,在该系统中,当DA神经元暴露于含铁培养基时会逐渐死亡。我们发现,OT(EC50 = 0.25µM)和 DOT(EC50 = 0.34µM)能有效保护 DA 神经元免于变性,这些效果直到神经变性的晚期都能观察到。在这种情况下,参考抗生素多西环素(TC DOX)也表现出保护作用。重要的是,经 OT、DOT 和 DOX 拯救的 DA 神经元保留了积聚和释放 DA 的能力,表明其功能完整。此外,具有铁螯合特性的分子(apotransferrin、desferoxamine)以及脂质过氧化和铁跃迁抑制剂(Trolox、Liproxstatin-1)也能复制 OT、DOT 和 DOX 对 DA 神经元的拯救作用。活细胞成像研究表明,TCs 和其他神经保护分子能阻止细胞内活性氧的释放和线粒体膜电位的破坏。然而,OT、DOT和DOX都不能保护DA神经元免受1-甲基-4-苯基吡啶鎓的选择性线粒体中毒。这表明,测试 TC 可能通过一种不直接涉及线粒体的机制来保护神经元免受铁介导的损伤。总之,我们证明了 OT 和 DOT 具有可用于防治帕金森病神经变性的良好特性。然而,与母体化合物 OT 相比,没有抗菌活性的 DOT 更适合作为候选药物。
{"title":"Oxytetracycline and its Non-Antibiotic Derivative DOT Protect Midbrain Dopamine Neurons from Iron-Driven Oxidative Damage.","authors":"Thaís Antonia Alves Fernandes, Aurore Tourville, Ismaila Ciss, Rafaela Ribeiro Silva, Bianca Andretto de Mattos, Maurício Dos Santos Pereira, Maxime Oblaza, Jean-Michel Brunel, Laurent Ferrié, Rita Raisman-Vozari, Bruno Figadère, Elaine Del-Bel, Patrick Pierre Michel","doi":"10.1007/s12640-025-00742-6","DOIUrl":"10.1007/s12640-025-00742-6","url":null,"abstract":"<p><p>This study aimed to investigate the neuroprotective potential of the tetracycline (TC) antibiotic oxytetracycline (OT) and its non-antibiotic derivative 4-dedimethylamino 12a-deoxy-oxytetracycline (DOT), in experimental conditions that mimic the gradual loss of dopamine (DA) neurons in Parkinson's disease (PD). Specifically, we established a model system of mouse midbrain cultures where DA neurons progressively die when exposed to an iron-containing medium. We found that OT (EC<sub>50</sub> = 0.25µM) and DOT (EC<sub>50</sub> = 0.34µM) efficiently protected DA neurons from degeneration, with these effects observable until advanced stages of neurodegeneration. The reference antibiotic TC doxycycline (DOX) also exhibited protective effects in this context. Importantly, DA neurons rescued by OT, DOT, and DOX retained their capacity to accumulate and release DA, indicating full functional integrity. Additionally, molecules with iron-chelating properties (apotransferrin, desferoxamine), as well as inhibitors of lipid peroxidation and ferroptosis (Trolox, Liproxstatin-1), could replicate the rescue of DA neurons provided by OT, DOT, and DOX. Live-cell imaging studies showed that test TCs and other neuroprotective molecules prevented the emission of intracellular reactive oxygen species and the associated disruption of the mitochondrial membrane potential. However, neither OT, DOT, nor DOX could protect DA neurons from selective mitochondrial poisoning by 1-methyl-4-phenylpyridinium. This suggests that test TCs may be protective against iron-mediated damage through a mechanism not directly involving mitochondria. Overall, we demonstrate that OT and DOT possess promising properties that could be useful for combating PD neurodegeneration. However, the absence of antimicrobial activity makes DOT a better candidate drug compared to its parent compound OT.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"43 2","pages":"16"},"PeriodicalIF":2.9,"publicationDate":"2025-03-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143676984","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","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
Neurotoxicity Research
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:604180095
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