首页 > 最新文献

Neurotoxicity Research最新文献

英文 中文
FABP3 Induces Mitochondrial Autophagy to Promote Neuronal Cell Apoptosis in Brain Ischemia-Reperfusion Injury. FABP3 在脑缺血再灌注损伤中诱导线粒体自噬以促进神经细胞凋亡
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-07-15 DOI: 10.1007/s12640-024-00712-4
Fang-Fang Zhong, Bo Wei, Guo-Xiang Bao, Yi-Ping Lou, Ming-Er Wei, Xin-Yue Wang, Xiao Xiao, Jin-Jin Tian

This study elucidates the molecular mechanisms by which FABP3 regulates neuronal apoptosis via mitochondrial autophagy in the context of cerebral ischemia-reperfusion (I/R). Employing a transient mouse model of middle cerebral artery occlusion (MCAO) established using the filament method, brain tissue samples were procured from I/R mice. High-throughput transcriptome sequencing on the Illumina CN500 platform was performed to identify differentially expressed mRNAs. Critical genes were selected by intersecting I/R-related genes from the GeneCards database with the differentially expressed mRNAs. The in vivo mechanism was explored by infecting I/R mice with lentivirus. Brain tissue injury, infarct volume ratio in the ischemic penumbra, neurologic deficits, behavioral abilities, neuronal apoptosis, apoptotic factors, inflammatory factors, and lipid peroxidation markers were assessed using H&E staining, TTC staining, Longa scoring, rotation experiments, immunofluorescence staining, and Western blot. For in vitro validation, an OGD/R model was established using primary neuron cells. Cell viability, apoptosis rate, mitochondrial oxidative stress, morphology, autophagosome formation, membrane potential, LC3 protein levels, and colocalization of autophagosomes and mitochondria were evaluated using MTT assay, LDH release assay, flow cytometry, ROS/MDA/GSH-Px measurement, transmission electron microscopy, MitoTracker staining, JC-1 method, Western blot, and immunofluorescence staining. FABP3 was identified as a critical gene in I/R through integrated transcriptome sequencing and bioinformatics analysis. In vivo experiments revealed that FABP3 silencing mitigated brain tissue damage, reduced infarct volume ratio, improved neurologic deficits, restored behavioral abilities, and attenuated neuronal apoptosis, inflammation, and mitochondrial oxidative stress in I/R mice. In vitro experiments demonstrated that FABP3 silencing restored OGD/R cell viability, reduced neuronal apoptosis, and decreased mitochondrial oxidative stress. Moreover, FABP3 induced mitochondrial autophagy through ROS, which was inhibited by the free radical scavenger NAC. Blocking mitochondrial autophagy with sh-ATG5 lentivirus confirmed that FABP3 induces mitochondrial dysfunction and neuronal apoptosis by activating mitochondrial autophagy. In conclusion, FABP3 activates mitochondrial autophagy through ROS, leading to mitochondrial dysfunction and neuronal apoptosis, thereby promoting cerebral ischemia-reperfusion injury.

本研究阐明了FABP3在脑缺血再灌注(I/R)情况下通过线粒体自噬调控神经细胞凋亡的分子机制。利用丝状法建立的大脑中动脉闭塞(MCAO)瞬时小鼠模型,从I/R小鼠身上获取脑组织样本。在 Illumina CN500 平台上进行了高通量转录组测序,以鉴定差异表达的 mRNA。通过将 GeneCards 数据库中与 I/R 相关的基因与差异表达的 mRNA 进行交叉,筛选出关键基因。通过用慢病毒感染I/R小鼠来探索体内机制。使用 H&E 染色、TTC 染色、Longa 评分、旋转实验、免疫荧光染色和 Western 印迹法评估了脑组织损伤、缺血半影的梗死体积比、神经功能缺损、行为能力、神经元凋亡、凋亡因子、炎症因子和脂质过氧化标记物。为了进行体外验证,使用原代神经元细胞建立了 OGD/R 模型。使用 MTT 试验、LDH 释放试验、流式细胞术、ROS/MDA/GSH-Px 测量、透射电子显微镜、MitoTracker 染色、JC-1 法、Western 印迹和免疫荧光染色评估了细胞活力、凋亡率、线粒体氧化应激、形态、自噬体形成、膜电位、LC3 蛋白水平以及自噬体和线粒体的共定位。通过综合转录组测序和生物信息学分析,发现FABP3是I/R的关键基因。体内实验表明,FABP3沉默可减轻I/R小鼠脑组织损伤,降低梗死体积比,改善神经功能缺损,恢复行为能力,减轻神经元凋亡、炎症和线粒体氧化应激。体外实验表明,沉默 FABP3 可恢复 OGD/R 细胞的活力,减少神经元凋亡,降低线粒体氧化应激。此外,FABP3 通过 ROS 诱导线粒体自噬,而自由基清除剂 NAC 可抑制线粒体自噬。用 sh-ATG5 慢病毒阻断线粒体自噬证实了 FABP3 通过激活线粒体自噬诱导线粒体功能障碍和神经元凋亡。总之,FABP3通过ROS激活线粒体自噬,导致线粒体功能障碍和神经细胞凋亡,从而促进脑缺血再灌注损伤。
{"title":"FABP3 Induces Mitochondrial Autophagy to Promote Neuronal Cell Apoptosis in Brain Ischemia-Reperfusion Injury.","authors":"Fang-Fang Zhong, Bo Wei, Guo-Xiang Bao, Yi-Ping Lou, Ming-Er Wei, Xin-Yue Wang, Xiao Xiao, Jin-Jin Tian","doi":"10.1007/s12640-024-00712-4","DOIUrl":"10.1007/s12640-024-00712-4","url":null,"abstract":"<p><p>This study elucidates the molecular mechanisms by which FABP3 regulates neuronal apoptosis via mitochondrial autophagy in the context of cerebral ischemia-reperfusion (I/R). Employing a transient mouse model of middle cerebral artery occlusion (MCAO) established using the filament method, brain tissue samples were procured from I/R mice. High-throughput transcriptome sequencing on the Illumina CN500 platform was performed to identify differentially expressed mRNAs. Critical genes were selected by intersecting I/R-related genes from the GeneCards database with the differentially expressed mRNAs. The in vivo mechanism was explored by infecting I/R mice with lentivirus. Brain tissue injury, infarct volume ratio in the ischemic penumbra, neurologic deficits, behavioral abilities, neuronal apoptosis, apoptotic factors, inflammatory factors, and lipid peroxidation markers were assessed using H&E staining, TTC staining, Longa scoring, rotation experiments, immunofluorescence staining, and Western blot. For in vitro validation, an OGD/R model was established using primary neuron cells. Cell viability, apoptosis rate, mitochondrial oxidative stress, morphology, autophagosome formation, membrane potential, LC3 protein levels, and colocalization of autophagosomes and mitochondria were evaluated using MTT assay, LDH release assay, flow cytometry, ROS/MDA/GSH-Px measurement, transmission electron microscopy, MitoTracker staining, JC-1 method, Western blot, and immunofluorescence staining. FABP3 was identified as a critical gene in I/R through integrated transcriptome sequencing and bioinformatics analysis. In vivo experiments revealed that FABP3 silencing mitigated brain tissue damage, reduced infarct volume ratio, improved neurologic deficits, restored behavioral abilities, and attenuated neuronal apoptosis, inflammation, and mitochondrial oxidative stress in I/R mice. In vitro experiments demonstrated that FABP3 silencing restored OGD/R cell viability, reduced neuronal apoptosis, and decreased mitochondrial oxidative stress. Moreover, FABP3 induced mitochondrial autophagy through ROS, which was inhibited by the free radical scavenger NAC. Blocking mitochondrial autophagy with sh-ATG5 lentivirus confirmed that FABP3 induces mitochondrial dysfunction and neuronal apoptosis by activating mitochondrial autophagy. In conclusion, FABP3 activates mitochondrial autophagy through ROS, leading to mitochondrial dysfunction and neuronal apoptosis, thereby promoting cerebral ischemia-reperfusion injury.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"42 4","pages":"35"},"PeriodicalIF":2.9,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141616934","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
Cerebral White Matter Alterations Associated With Oligodendrocyte Vulnerability in Organic Acidurias: Insights in Glutaric Aciduria Type I. 有机酸尿症患者大脑白质改变与少突胶质细胞脆弱性有关:戊二酸尿症 I 型的启示。
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-07-04 DOI: 10.1007/s12640-024-00710-6
Eugenia Isasi, Moacir Wajner, Juliana Avila Duarte, Silvia Olivera-Bravo

The white matter is an important constituent of the central nervous system, containing axons, oligodendrocytes, and its progenitor cells, astrocytes, and microglial cells. Oligodendrocytes are central for myelin synthesis, the insulating envelope that protects axons and allows normal neural conduction. Both, oligodendrocytes and myelin, are highly vulnerable to toxic factors in many neurodevelopmental and neurodegenerative disorders associated with disturbances of myelination. Here we review the main alterations in oligodendrocytes and myelin observed in some organic acidurias/acidemias, which correspond to inherited neurometabolic disorders biochemically characterized by accumulation of potentially neurotoxic organic acids and their derivatives. The yet incompletely understood mechanisms underlying the high vulnerability of OLs and/or myelin in glutaric acidemia type I, the most prototypical cerebral organic aciduria, are particularly discussed.

白质是中枢神经系统的重要组成部分,包含轴突、少突胶质细胞及其祖细胞、星形胶质细胞和小胶质细胞。少突胶质细胞是髓鞘合成的核心,而髓鞘是保护轴突的绝缘包膜,可实现正常的神经传导。在许多与髓鞘化紊乱有关的神经发育和神经退行性疾病中,少突胶质细胞和髓鞘都极易受到毒性因素的影响。在此,我们回顾了在一些有机酸病/贫血症中观察到的少突胶质细胞和髓鞘的主要变化,这些病症与遗传性神经代谢紊乱相对应,其生物化学特征是潜在神经毒性有机酸及其衍生物的积累。本文特别讨论了戊二酸血症 I 型(最典型的脑有机酸尿症)中的有机酸和/或髓鞘极易受到损伤的机制,但人们对这一机制的理解尚不完全。
{"title":"Cerebral White Matter Alterations Associated With Oligodendrocyte Vulnerability in Organic Acidurias: Insights in Glutaric Aciduria Type I.","authors":"Eugenia Isasi, Moacir Wajner, Juliana Avila Duarte, Silvia Olivera-Bravo","doi":"10.1007/s12640-024-00710-6","DOIUrl":"10.1007/s12640-024-00710-6","url":null,"abstract":"<p><p>The white matter is an important constituent of the central nervous system, containing axons, oligodendrocytes, and its progenitor cells, astrocytes, and microglial cells. Oligodendrocytes are central for myelin synthesis, the insulating envelope that protects axons and allows normal neural conduction. Both, oligodendrocytes and myelin, are highly vulnerable to toxic factors in many neurodevelopmental and neurodegenerative disorders associated with disturbances of myelination. Here we review the main alterations in oligodendrocytes and myelin observed in some organic acidurias/acidemias, which correspond to inherited neurometabolic disorders biochemically characterized by accumulation of potentially neurotoxic organic acids and their derivatives. The yet incompletely understood mechanisms underlying the high vulnerability of OLs and/or myelin in glutaric acidemia type I, the most prototypical cerebral organic aciduria, are particularly discussed.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"42 4","pages":"33"},"PeriodicalIF":2.9,"publicationDate":"2024-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141498561","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 of Excitotoxicity, Oxidative Stress and Bioenergetics Disruption in the Neuropathology of Nonketotic Hyperglycinemia. 兴奋毒性、氧化应激和生物能破坏在非酮症性高血糖神经病理学中的作用
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-29 DOI: 10.1007/s12640-024-00711-5
Guilhian Leipnitz, Jaqueline Santana da Rosa, Moacir Wajner

Nonketotic hyperglycinemia (NKH) is an inherited disorder of amino acid metabolism biochemically characterized by the accumulation of glycine (Gly) predominantly in the brain. Affected patients usually manifest with neurological symptoms including hypotonia, seizures, epilepsy, lethargy, and coma, the pathophysiology of which is still not completely understood. Treatment is limited and based on lowering Gly levels aiming to reduce overstimulation of N-methyl-D-aspartate (NMDA) receptors. Mounting in vitro and in vivo animal and human evidence have recently suggested that excitotoxicity, oxidative stress, and bioenergetics disruption induced by Gly are relevant mechanisms involved in the neuropathology of NKH. This brief review gives emphasis to the deleterious effects of Gly in the brain of patients and animal models of NKH that may offer perspectives for the development of novel adjuvant treatments for this disorder.

非酮症性高甘氨酸血症(NKH)是一种遗传性氨基酸代谢紊乱,其生化特征是甘氨酸(Gly)主要在大脑中蓄积。患者通常表现为神经系统症状,包括肌张力低下、抽搐、癫痫、嗜睡和昏迷,其病理生理学至今仍不完全清楚。治疗方法有限,主要是降低甘氨酸水平,以减少对 N-甲基-D-天冬氨酸(NMDA)受体的过度刺激。最近,越来越多的体外和体内动物和人体证据表明,Gly 诱导的兴奋毒性、氧化应激和生物能破坏是 NKH 神经病理学的相关机制。这篇简短的综述强调了 Gly 对 NKH 患者和动物模型大脑的有害影响,为开发治疗这种疾病的新型辅助疗法提供了前景。
{"title":"The Role of Excitotoxicity, Oxidative Stress and Bioenergetics Disruption in the Neuropathology of Nonketotic Hyperglycinemia.","authors":"Guilhian Leipnitz, Jaqueline Santana da Rosa, Moacir Wajner","doi":"10.1007/s12640-024-00711-5","DOIUrl":"10.1007/s12640-024-00711-5","url":null,"abstract":"<p><p>Nonketotic hyperglycinemia (NKH) is an inherited disorder of amino acid metabolism biochemically characterized by the accumulation of glycine (Gly) predominantly in the brain. Affected patients usually manifest with neurological symptoms including hypotonia, seizures, epilepsy, lethargy, and coma, the pathophysiology of which is still not completely understood. Treatment is limited and based on lowering Gly levels aiming to reduce overstimulation of N-methyl-D-aspartate (NMDA) receptors. Mounting in vitro and in vivo animal and human evidence have recently suggested that excitotoxicity, oxidative stress, and bioenergetics disruption induced by Gly are relevant mechanisms involved in the neuropathology of NKH. This brief review gives emphasis to the deleterious effects of Gly in the brain of patients and animal models of NKH that may offer perspectives for the development of novel adjuvant treatments for this disorder.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"42 4","pages":"32"},"PeriodicalIF":2.9,"publicationDate":"2024-06-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469703","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
FOXA1 Suppresses Endoplasmic Reticulum Stress, Oxidative Stress, and Neuronal Apoptosis in Parkinson's Disease by Activating PON2 Transcription. FOXA1 通过激活 PON2 转录抑制帕金森病的内质网应激、氧化应激和神经元凋亡
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-27 DOI: 10.1007/s12640-024-00709-z
Jiahui Liu, Yu Fan, Jinyu Chen, Meili Zhao, Changchun Jiang

Endoplasmic reticulum (ER) stress and oxidative stress (OS) are often related states in pathological conditions including Parkinson's disease (PD). This study investigates the role of anti-oxidant protein paraoxonase 2 (PON2) in ER stress and OS in PD, along with its regulatory molecule. PD was induced in C57BL/6 mice using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP) treatment and in SH-SY5Y cells using 1-methyl-4-phenylpyridinium. PON2 was found to be poorly expressed in the substantia nigra pars compacta (SNc) of PD mice, and its overexpression improved motor coordination of mice. Through the evaluation of tyrosine hydroxylase, dopamine transporter, reactive oxygen species (ROS), and C/EBP homologous protein (CHOP) levels and neuronal loss in mice, as well as the examination of CHOP, glucose-regulated protein 94 (GRP94), GRP78, caspase-12, sarco/endoplasmic reticulum calcium ATPase 2, malondialdehyde, and superoxide dismutase levels in SH-SY5Y cells, we observed that PON2 overexpression mitigated ER stress, OS, and neuronal apoptosis both in vivo and in vitro. Forkhead box A1 (FOXA1) was identified as a transcription factor binding to the PON2 promoter to activate its transcription. Upregulation of FOXA1 similarly protected against neuronal loss by alleviating ER stress and OS, while the protective roles were abrogated by additional PON2 silencing. In conclusion, this study demonstrates that FOXA1-mediated transcription of PON2 alleviates ER stress and OS, ultimately reducing neuronal apoptosis in PD.

内质网(ER)应激和氧化应激(OS)往往是包括帕金森病(PD)在内的病理状态中的相关状态。本研究探讨了抗氧化蛋白对氧合酶2(PON2)及其调控分子在帕金森病ER应激和OS中的作用。用1-甲基-4-苯基-1,2,3,6-四氢吡啶盐酸盐(MPTP)处理C57BL/6小鼠,用1-甲基-4-苯基吡啶鎓诱导SH-SY5Y细胞。研究发现,PON2在帕金森病小鼠黑质(SNc)中的表达量很低,而过量表达PON2可改善小鼠的运动协调能力。通过评估小鼠体内酪氨酸羟化酶、多巴胺转运体、活性氧(ROS)、C/EBP同源蛋白(CHOP)的水平和神经元缺失情况,以及CHOP、葡萄糖调节蛋白94(GRP94)、GRP78、caspase-12我们观察到,在 SH-SY5Y 细胞中过表达 PON2 可减轻体内和体外的 ER 应激、OS 和神经元凋亡。研究发现,叉头盒 A1(FOXA1)是与 PON2 启动子结合以激活其转录的转录因子。FOXA1的上调同样能通过减轻ER应激和OS来防止神经元缺失,而额外的PON2沉默则会削弱其保护作用。总之,这项研究证明,FOXA1介导的PON2转录可减轻ER应激和OS,最终减少帕金森病中神经元的凋亡。
{"title":"FOXA1 Suppresses Endoplasmic Reticulum Stress, Oxidative Stress, and Neuronal Apoptosis in Parkinson's Disease by Activating PON2 Transcription.","authors":"Jiahui Liu, Yu Fan, Jinyu Chen, Meili Zhao, Changchun Jiang","doi":"10.1007/s12640-024-00709-z","DOIUrl":"10.1007/s12640-024-00709-z","url":null,"abstract":"<p><p>Endoplasmic reticulum (ER) stress and oxidative stress (OS) are often related states in pathological conditions including Parkinson's disease (PD). This study investigates the role of anti-oxidant protein paraoxonase 2 (PON2) in ER stress and OS in PD, along with its regulatory molecule. PD was induced in C57BL/6 mice using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP) treatment and in SH-SY5Y cells using 1-methyl-4-phenylpyridinium. PON2 was found to be poorly expressed in the substantia nigra pars compacta (SNc) of PD mice, and its overexpression improved motor coordination of mice. Through the evaluation of tyrosine hydroxylase, dopamine transporter, reactive oxygen species (ROS), and C/EBP homologous protein (CHOP) levels and neuronal loss in mice, as well as the examination of CHOP, glucose-regulated protein 94 (GRP94), GRP78, caspase-12, sarco/endoplasmic reticulum calcium ATPase 2, malondialdehyde, and superoxide dismutase levels in SH-SY5Y cells, we observed that PON2 overexpression mitigated ER stress, OS, and neuronal apoptosis both in vivo and in vitro. Forkhead box A1 (FOXA1) was identified as a transcription factor binding to the PON2 promoter to activate its transcription. Upregulation of FOXA1 similarly protected against neuronal loss by alleviating ER stress and OS, while the protective roles were abrogated by additional PON2 silencing. In conclusion, this study demonstrates that FOXA1-mediated transcription of PON2 alleviates ER stress and OS, ultimately reducing neuronal apoptosis in PD.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"42 4","pages":"31"},"PeriodicalIF":2.9,"publicationDate":"2024-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141458396","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
PEG300 Protects Mitochondrial Function By Upregulating PGC-1α to Delay Central Nervous System Oxygen Toxicity in Mice. PEG300 通过上调 PGC-1α 来保护线粒体功能,从而延缓小鼠中枢神经系统的氧毒性。
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-17 DOI: 10.1007/s12640-024-00708-0
Xin Li, Yue Shen, Dan Li, Kun Zhang, Jia Liu, Lu Yao, Jun Yang, Jiao Qian

Central nervous system oxygen toxicity (CNS-OT) is a complication of hyperbaric oxygen (HBO) treatment, with limited prevention and treatment options available. In this study, we aimed to explore the effect of polyethylene glycol 300 (PEG300) on CNS-OT and underlying mechanisms. Motor and cognitive functions of mice in normobaric conditions were evaluated by Morris water maze, passive active avoidance, and rotarod tests. HBO was applied at 6 atmospheres absolute (ATA) for 30 min after drug administration. The latency period of convulsion in mice was recorded, and hippocampal tissues were extracted for biochemical experiments. Our experimental results showed that PEG300 extended the convulsion latencies in CNS-OT mice, reduced oxidative stress and inflammation levels in hippocampal tissues. Furthermore, PEG300 preserved mitochondrial integrity and maintained mitochondrial membrane potential in hippocampal tissue by upregulating Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha (PGC-1α). This protective effect was enhanced following the administration of ZLN005, an agonist of PGC-1a. Hence, our study suggests that PEG300 might exert protective effects by upregulating PGC-1α expression and preserving mitochondrial health, offering promising prospects for CNS-OT treatment.

中枢神经系统氧毒性(CNS-OT)是高压氧(HBO)治疗的一种并发症,目前可用的预防和治疗方案有限。本研究旨在探讨聚乙二醇 300(PEG300)对中枢神经系统氧毒性的影响及其内在机制。通过莫里斯水迷宫、被动主动回避和旋转木马测试评估了常压条件下小鼠的运动和认知功能。给药后在 6 个绝对大气压(ATA)下使用 HBO 30 分钟。记录小鼠抽搐的潜伏期,并提取海马组织进行生化实验。实验结果表明,PEG300 延长了 CNS-OT 小鼠的抽搐潜伏期,降低了海马组织中的氧化应激和炎症水平。此外,PEG300 还能通过上调过氧化物酶体增殖激活受体伽马辅激活剂 1-α (PGC-1α)来保护线粒体的完整性并维持海马组织中线粒体的膜电位。在服用 PGC-1a 激动剂 ZLN005 后,这种保护作用得到加强。因此,我们的研究表明,PEG300 可通过上调 PGC-1α 的表达和保护线粒体的健康来发挥保护作用,为中枢神经系统-OT 的治疗提供了广阔的前景。
{"title":"PEG300 Protects Mitochondrial Function By Upregulating PGC-1α to Delay Central Nervous System Oxygen Toxicity in Mice.","authors":"Xin Li, Yue Shen, Dan Li, Kun Zhang, Jia Liu, Lu Yao, Jun Yang, Jiao Qian","doi":"10.1007/s12640-024-00708-0","DOIUrl":"10.1007/s12640-024-00708-0","url":null,"abstract":"<p><p>Central nervous system oxygen toxicity (CNS-OT) is a complication of hyperbaric oxygen (HBO) treatment, with limited prevention and treatment options available. In this study, we aimed to explore the effect of polyethylene glycol 300 (PEG300) on CNS-OT and underlying mechanisms. Motor and cognitive functions of mice in normobaric conditions were evaluated by Morris water maze, passive active avoidance, and rotarod tests. HBO was applied at 6 atmospheres absolute (ATA) for 30 min after drug administration. The latency period of convulsion in mice was recorded, and hippocampal tissues were extracted for biochemical experiments. Our experimental results showed that PEG300 extended the convulsion latencies in CNS-OT mice, reduced oxidative stress and inflammation levels in hippocampal tissues. Furthermore, PEG300 preserved mitochondrial integrity and maintained mitochondrial membrane potential in hippocampal tissue by upregulating Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha (PGC-1α). This protective effect was enhanced following the administration of ZLN005, an agonist of PGC-1a. Hence, our study suggests that PEG300 might exert protective effects by upregulating PGC-1α expression and preserving mitochondrial health, offering promising prospects for CNS-OT treatment.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"42 4","pages":"30"},"PeriodicalIF":2.9,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141331504","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
Assessment of Hippocampal-Related Behavioral Changes in Adolescent Rats of both Sexes Following Voluntary Intermittent Ethanol Intake and Noise Exposure: A Putative Underlying Mechanism and Implementation of a Non-pharmacological Preventive Strategy. 评估自愿间歇摄入乙醇和暴露于噪声后青春期雌雄大鼠海马相关行为的变化:推测的基本机制和非药物预防策略的实施。
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-10 DOI: 10.1007/s12640-024-00707-1
G E Buján, L D'Alessio, H A Serra, L R Guelman, S J Molina

Ethanol (EtOH) intake and noise exposure are particularly concerning among human adolescents because the potential to harm brain. Unfortunately, putative underlying mechanisms remain to be elucidated. Moreover, implementing non-pharmacological strategies, such as enriched environments (EE), would be pertinent in the field of neuroprotection. This study aims to explore possible underlying triggering mechanism of hippocampus-dependent behaviors in adolescent animals of both sexes following ethanol intake, noise exposure, or a combination of both, as well as the impact of EE. Adolescent Wistar rats of both sexes were subjected to an intermittent voluntary EtOH intake paradigm for one week. A subgroup of animals was exposed to white noise for two hours after the last session of EtOH intake. Some animals of both groups were housed in EE cages. Hippocampal-dependent behavioral assessment and hippocampal oxidative state evaluation were performed. Results show that different hippocampal-dependent behavioral alterations might be induced in animals of both sexes after EtOH intake and sequential noise exposure, that in some cases are sex-specific. Moreover, hippocampal oxidative imbalance seems to be one of the potential underlying mechanisms. Additionally, most behavioral and oxidative alterations were prevented by EE. These findings suggest that two frequently found environmental agents may impact behavior and oxidative pathways in both sexes in an animal model. In addition, EE resulted a partially effective neuroprotective strategy. Therefore, it could be suggested that the implementation of a non-pharmacological approach might also potentially provide neuroprotective advantages against other challenges. Finally, considering its potential for translational human benefit might be worth.

青少年摄入乙醇(EtOH)和接触噪音尤其令人担忧,因为这有可能对大脑造成伤害。遗憾的是,其潜在机制仍有待阐明。此外,在神经保护领域,实施非药物策略(如丰富的环境(EE))也很有意义。本研究旨在探讨摄入乙醇、暴露于噪音或二者结合后,可能引发青少年雌雄动物海马依赖行为的潜在机制,以及 EE 的影响。对青春期Wistar雌雄大鼠进行为期一周的间歇性自愿摄入乙醇范例研究。在最后一次摄入乙醇后,一组动物暴露于白噪声两小时。两组中都有一些动物被安置在EE笼中。进行了海马依赖行为评估和海马氧化状态评估。结果表明,在摄入乙醇和连续接触噪声后,雌雄动物可能会诱发不同的海马依赖性行为改变,在某些情况下具有性别特异性。此外,海马氧化失衡似乎是潜在的潜在机制之一。此外,大多数行为和氧化改变都能被 EE 所阻止。这些发现表明,在动物模型中,两种常见的环境因素可能会影响两性的行为和氧化途径。此外,EE 还产生了部分有效的神经保护策略。因此,可以认为采用非药物方法也有可能在应对其他挑战时提供神经保护优势。最后,还值得考虑其转化为人类利益的潜力。
{"title":"Assessment of Hippocampal-Related Behavioral Changes in Adolescent Rats of both Sexes Following Voluntary Intermittent Ethanol Intake and Noise Exposure: A Putative Underlying Mechanism and Implementation of a Non-pharmacological Preventive Strategy.","authors":"G E Buján, L D'Alessio, H A Serra, L R Guelman, S J Molina","doi":"10.1007/s12640-024-00707-1","DOIUrl":"10.1007/s12640-024-00707-1","url":null,"abstract":"<p><p>Ethanol (EtOH) intake and noise exposure are particularly concerning among human adolescents because the potential to harm brain. Unfortunately, putative underlying mechanisms remain to be elucidated. Moreover, implementing non-pharmacological strategies, such as enriched environments (EE), would be pertinent in the field of neuroprotection. This study aims to explore possible underlying triggering mechanism of hippocampus-dependent behaviors in adolescent animals of both sexes following ethanol intake, noise exposure, or a combination of both, as well as the impact of EE. Adolescent Wistar rats of both sexes were subjected to an intermittent voluntary EtOH intake paradigm for one week. A subgroup of animals was exposed to white noise for two hours after the last session of EtOH intake. Some animals of both groups were housed in EE cages. Hippocampal-dependent behavioral assessment and hippocampal oxidative state evaluation were performed. Results show that different hippocampal-dependent behavioral alterations might be induced in animals of both sexes after EtOH intake and sequential noise exposure, that in some cases are sex-specific. Moreover, hippocampal oxidative imbalance seems to be one of the potential underlying mechanisms. Additionally, most behavioral and oxidative alterations were prevented by EE. These findings suggest that two frequently found environmental agents may impact behavior and oxidative pathways in both sexes in an animal model. In addition, EE resulted a partially effective neuroprotective strategy. Therefore, it could be suggested that the implementation of a non-pharmacological approach might also potentially provide neuroprotective advantages against other challenges. Finally, considering its potential for translational human benefit might be worth.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"42 3","pages":"29"},"PeriodicalIF":2.9,"publicationDate":"2024-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141296483","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
Rotenone Induces a Neuropathological Phenotype in Cholinergic-like Neurons Resembling Parkinson's Disease Dementia (PDD). 罗替农诱导胆碱能样神经元出现类似帕金森氏症痴呆(PDD)的神经病理学表型
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-06 DOI: 10.1007/s12640-024-00705-3
Daniela Giraldo-Berrio, Miguel Mendivil-Perez, Carlos Velez-Pardo, Marlene Jimenez-Del-Rio

Parkinson's disease with dementia (PDD) is a neurological disorder that clinically and neuropathologically overlaps with Parkinson's disease (PD) and Alzheimer's disease (AD). Although it is assumed that alpha-synuclein ( α -Syn), amyloid beta (A β ), and the protein Tau might synergistically induce cholinergic neuronal degeneration, presently the pathological mechanism of PDD remains unclear. Therefore, it is essential to delve into the cellular and molecular aspects of this neurological entity to identify potential targets for prevention and treatment strategies. Cholinergic-like neurons (ChLNs) were exposed to rotenone (ROT, 10 μ M) for 24 h. ROT provokes loss of Δ Ψ m , generation of reactive oxygen species (ROS), phosphorylation of leucine-rich repeated kinase 2 (LRRK2 at Ser935) concomitantly with phosphorylation of α -synuclein ( α -Syn, Ser129), induces accumulation of intracellular A β (iA β ), oxidized DJ-1 (Cys106), as well as phosphorylation of TAU (Ser202/Thr205), increases the phosphorylation of c-JUN (Ser63/Ser73), and increases expression of proapoptotic proteins TP53, PUMA, and cleaved caspase 3 (CC3) in ChLNs. These neuropathological features resemble those reproduced in presenilin 1 (PSEN1) E280A ChLNs. Interestingly, anti-oxidant and anti-amyloid cannabidiol (CBD), JNK inhibitor SP600125 (SP), TP53 inhibitor pifithrin- α (PFT), and LRRK2 kinase inhibitor PF-06447475 (PF475) significantly diminish ROT-induced oxidative stress (OS), proteinaceous, and cell death markers in ChLNs compared to naïve ChLNs. In conclusion, ROT induces p- α -Syn, iA β , p-Tau, and cell death in ChLNs, recapitulating the neuropathology findings in PDD. Our report provides an excellent in vitro model to test for potential therapeutic strategies against PDD. Our data suggest that ROT induces a neuropathologic phenotype in ChLNs similar to that caused by the mutation PSEN1 E280A.

帕金森病伴痴呆(PDD)是一种神经系统疾病,在临床和神经病理学上与帕金森病(PD)和阿尔茨海默病(AD)重叠。尽管人们认为α-突触核蛋白(α -Syn)、淀粉样蛋白β(A β)和蛋白质Tau可能会协同诱导胆碱能神经元变性,但目前PDD的病理机制仍不清楚。因此,有必要深入研究这种神经系统疾病的细胞和分子方面,以确定潜在的预防和治疗策略靶点。将胆碱能样神经元(ChLNs)暴露于鱼藤酮(ROT,10 μ M)中 24 小时。ROT 可导致 Δ Ψ m 的丧失、活性氧(ROS)的生成、富亮氨酸重复激酶 2(LRRK2)在 Ser935 处的磷酸化以及 α -synuclein ( α -Syn, Ser129) 的磷酸化,并诱导细胞内 A β(iA β)的积累、氧化的 DJ-1(Cys106)以及 TAU(Ser202/Thr205)的磷酸化,增加 c-JUN(Ser63/Ser73)的磷酸化,并增加 ChLNs 中促凋亡蛋白 TP53、PUMA 和裂解的 Caspase 3(CC3)的表达。这些神经病理学特征与预激蛋白 1(PSEN1)E280A ChLNs 中再现的特征相似。有趣的是,与天真 ChLNs 相比,抗氧化剂和抗淀粉样蛋白大麻二酚(CBD)、JNK 抑制剂 SP600125(SP)、TP53 抑制剂 pifithrin- α(PFT)和 LRRK2 激酶抑制剂 PF-06447475 (PF475)能显著减少 ROT 在 ChLNs 中诱导的氧化应激(OS)、蛋白质和细胞死亡标记物。总之,ROT 可诱导 ChLNs 中 p-α -Syn、iA β、p-Tau 和细胞死亡,再现了 PDD 的神经病理学发现。我们的报告为测试潜在的 PDD 治疗策略提供了一个极好的体外模型。我们的数据表明,ROT 在 ChLNs 中诱导的神经病理学表型与 PSEN1 E280A 突变引起的表型相似。
{"title":"Rotenone Induces a Neuropathological Phenotype in Cholinergic-like Neurons Resembling Parkinson's Disease Dementia (PDD).","authors":"Daniela Giraldo-Berrio, Miguel Mendivil-Perez, Carlos Velez-Pardo, Marlene Jimenez-Del-Rio","doi":"10.1007/s12640-024-00705-3","DOIUrl":"10.1007/s12640-024-00705-3","url":null,"abstract":"<p><p>Parkinson's disease with dementia (PDD) is a neurological disorder that clinically and neuropathologically overlaps with Parkinson's disease (PD) and Alzheimer's disease (AD). Although it is assumed that alpha-synuclein ( <math><mi>α</mi></math> -Syn), amyloid beta (A <math><mi>β</mi></math> ), and the protein Tau might synergistically induce cholinergic neuronal degeneration, presently the pathological mechanism of PDD remains unclear. Therefore, it is essential to delve into the cellular and molecular aspects of this neurological entity to identify potential targets for prevention and treatment strategies. Cholinergic-like neurons (ChLNs) were exposed to rotenone (ROT, 10 <math><mi>μ</mi></math> M) for 24 h. ROT provokes loss of <math><mrow><mi>Δ</mi> <mrow><mi>Ψ</mi> <mi>m</mi></mrow> </mrow> </math> , generation of reactive oxygen species (ROS), phosphorylation of leucine-rich repeated kinase 2 (LRRK2 at Ser<sup>935</sup>) concomitantly with phosphorylation of <math><mi>α</mi></math> -synuclein ( <math><mi>α</mi></math> -Syn, Ser<sup>129</sup>), induces accumulation of intracellular A <math><mi>β</mi></math> (iA <math><mi>β</mi></math> ), oxidized DJ-1 (Cys<sup>106</sup>), as well as phosphorylation of TAU (Ser<sup>202</sup>/Thr<sup>205</sup>), increases the phosphorylation of c-JUN (Ser<sup>63</sup>/Ser<sup>73</sup>), and increases expression of proapoptotic proteins TP53, PUMA, and cleaved caspase 3 (CC3) in ChLNs. These neuropathological features resemble those reproduced in presenilin 1 (PSEN1) E280A ChLNs. Interestingly, anti-oxidant and anti-amyloid cannabidiol (CBD), JNK inhibitor SP600125 (SP), TP53 inhibitor pifithrin- <math><mi>α</mi></math> (PFT), and LRRK2 kinase inhibitor PF-06447475 (PF475) significantly diminish ROT-induced oxidative stress (OS), proteinaceous, and cell death markers in ChLNs compared to naïve ChLNs. In conclusion, ROT induces p- <math><mi>α</mi></math> -Syn, iA <math><mi>β</mi></math> , p-Tau, and cell death in ChLNs, recapitulating the neuropathology findings in PDD. Our report provides an excellent in vitro model to test for potential therapeutic strategies against PDD. Our data suggest that ROT induces a neuropathologic phenotype in ChLNs similar to that caused by the mutation PSEN1 E280A.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"42 3","pages":"28"},"PeriodicalIF":2.9,"publicationDate":"2024-06-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11156752/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141262426","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
Fer-1 Protects against Isoflurane-Induced Ferroptosis in Astrocytes and Cognitive Impairment in Neonatal Mice. Fer-1 保护新生小鼠免受异氟醚诱导的星形胶质细胞铁突变和认知障碍的影响
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-05-31 DOI: 10.1007/s12640-024-00706-2
Peng Zhang, Xiaotong Shi, Danyi He, Yu Hu, Yongchao Zhang, Youyi Zhao, Sanxing Ma, Shuhui Cao, Meiting Zhai, Ze Fan

Early and prolonged exposure to anesthetic agents could cause neurodevelopmental disorders in children. Astrocytes, heavily outnumber neurons in the brain, are crucial regulators of synaptic formation and function during development. However, how general anesthetics act on astrocytes and the impact on cognition are still unclear. In this study, we investigated the role of ferroptosis and GPX4, a major hydroperoxide scavenger playing a pivotal role in suppressing the process of ferroptosis, and their underlying mechanism in isoflurane-induced cytotoxicity in astrocytes and cognitive impairment. Our results showed that early 6 h isoflurane anesthesia induced cognitive impairment in mice. Ferroptosis-relative genes and metabolic changes were involved in the pathological process of isoflurane-induced cytotoxicity in astrocytes. The level of GPX4 was decreased while the expression of 4-HNE and generation of ROS were elevated after isoflurane exposure. Selectively blocking ferroptosis with Fer-1 attenuated the abovementioned cytotoxicity in astrocytes, paralleling with the reverse of the changes in GPX4, ROS and 4-HNE secondary to isoflurane anesthesia. Fer-1 attenuated the cognitive impairment induced by prolonged isoflurane exposure. Thus, ferroptosis conduced towards isoflurane-induced cytotoxicity in astrocytes via suppressing GPX4 and promoting lipid peroxidation. Fer-1 was expected to be an underlying intervention for the neurotoxicity induced by isoflurane in the developing brain, and to alleviate cognitive impairment in neonates.

过早和长时间接触麻醉剂可能会导致儿童神经发育障碍。星形胶质细胞在大脑中的数量远远超过神经元,是发育过程中突触形成和功能的重要调节器。然而,全身麻醉剂如何作用于星形胶质细胞及其对认知的影响仍不清楚。在这项研究中,我们研究了异氟醚诱导的星形胶质细胞细胞毒性和认知障碍中的铁蜕变和 GPX4(一种主要的过氧化氢清除剂,在抑制铁蜕变过程中起着关键作用)的作用及其内在机制。我们的研究结果表明,早期6 h异氟醚麻醉会诱发小鼠认知障碍。异氟醚诱导的星形胶质细胞细胞毒性的病理过程中涉及铁变态相关基因和代谢变化。暴露于异氟醚后,GPX4的水平下降,而4-HNE的表达和ROS的生成则升高。用 Fer-1 选择性阻断铁突变可减轻星形胶质细胞的上述细胞毒性,这与异氟醚麻醉后 GPX4、ROS 和 4-HNE 的变化相反。Fer-1 可减轻长时间暴露于异氟醚诱发的认知障碍。因此,铁变态反应通过抑制 GPX4 和促进脂质过氧化,对异氟醚诱导的星形胶质细胞细胞毒性起作用。铁-1有望成为异氟醚在发育中大脑诱导的神经毒性的基本干预措施,并减轻新生儿的认知障碍。
{"title":"Fer-1 Protects against Isoflurane-Induced Ferroptosis in Astrocytes and Cognitive Impairment in Neonatal Mice.","authors":"Peng Zhang, Xiaotong Shi, Danyi He, Yu Hu, Yongchao Zhang, Youyi Zhao, Sanxing Ma, Shuhui Cao, Meiting Zhai, Ze Fan","doi":"10.1007/s12640-024-00706-2","DOIUrl":"10.1007/s12640-024-00706-2","url":null,"abstract":"<p><p>Early and prolonged exposure to anesthetic agents could cause neurodevelopmental disorders in children. Astrocytes, heavily outnumber neurons in the brain, are crucial regulators of synaptic formation and function during development. However, how general anesthetics act on astrocytes and the impact on cognition are still unclear. In this study, we investigated the role of ferroptosis and GPX4, a major hydroperoxide scavenger playing a pivotal role in suppressing the process of ferroptosis, and their underlying mechanism in isoflurane-induced cytotoxicity in astrocytes and cognitive impairment. Our results showed that early 6 h isoflurane anesthesia induced cognitive impairment in mice. Ferroptosis-relative genes and metabolic changes were involved in the pathological process of isoflurane-induced cytotoxicity in astrocytes. The level of GPX4 was decreased while the expression of 4-HNE and generation of ROS were elevated after isoflurane exposure. Selectively blocking ferroptosis with Fer-1 attenuated the abovementioned cytotoxicity in astrocytes, paralleling with the reverse of the changes in GPX4, ROS and 4-HNE secondary to isoflurane anesthesia. Fer-1 attenuated the cognitive impairment induced by prolonged isoflurane exposure. Thus, ferroptosis conduced towards isoflurane-induced cytotoxicity in astrocytes via suppressing GPX4 and promoting lipid peroxidation. Fer-1 was expected to be an underlying intervention for the neurotoxicity induced by isoflurane in the developing brain, and to alleviate cognitive impairment in neonates.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"42 3","pages":"27"},"PeriodicalIF":2.9,"publicationDate":"2024-05-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141180237","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
Correction to: Connexin 43 Promotes Neurogenesis via Regulating Aquaporin-4 after Cerebral Ischemia. 更正:脑缺血后,Connexin 43 通过调节 Aquaporin-4 促进神经再生
IF 2.9 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-04-30 DOI: 10.1007/s12640-024-00701-7
Heling Chu, Jing Dong, Yuping Tang, Chuyi Huang, Qihao Guo
{"title":"Correction to: Connexin 43 Promotes Neurogenesis via Regulating Aquaporin-4 after Cerebral Ischemia.","authors":"Heling Chu, Jing Dong, Yuping Tang, Chuyi Huang, Qihao Guo","doi":"10.1007/s12640-024-00701-7","DOIUrl":"10.1007/s12640-024-00701-7","url":null,"abstract":"","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"42 3","pages":"26"},"PeriodicalIF":2.9,"publicationDate":"2024-04-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140869823","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
Hyperoxia and brain: the link between necessity and injury from a molecular perspective 高氧与大脑:从分子角度看必要性与损伤之间的联系
IF 3.7 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-04-15 DOI: 10.1007/s12640-024-00702-6
Richard Simon Machado, Khiany Mathias, Larissa Joaquim, Rafaella Willig de Quadros, Gislaine Tezza Rezin, Fabricia Petronilho

Oxygen (O2) supplementation is commonly used to treat hypoxia in patients with respiratory failure. However, indiscriminate use can lead to hyperoxia, a condition detrimental to living tissues, particularly the brain. The brain is sensitive to reactive oxygen species (ROS) and inflammation caused by high concentrations of O2, which can result in brain damage and mitochondrial dysfunction, common features of neurodegenerative disorders. Hyperoxia leads to increased production of ROS, causing oxidative stress, an imbalance between oxidants and antioxidants, which can damage tissues. The brain is particularly vulnerable to oxidative stress due to its lipid composition, high O2 consumption rate, and low levels of antioxidant enzymes. Moreover, hyperoxia can cause vasoconstriction and decreased O2 supply to the brain, posing a challenge to redox balance and neurodegenerative processes. Studies have shown that the severity of hyperoxia-induced brain damage varies with inspired O2 concentration and duration of exposure. Therefore, careful evaluation of the balance between benefits and risks of O2 supplementation, especially in clinical settings, is crucial.

补充氧气通常用于治疗呼吸衰竭患者的缺氧。然而,不加选择地使用氧气会导致高氧症,而高氧症是一种对生物组织(尤其是大脑)有害的疾病。大脑对高浓度氧气引起的活性氧(ROS)和炎症非常敏感,会导致脑损伤和线粒体功能障碍,这是神经退行性疾病的常见特征。高氧会导致 ROS 生成增加,造成氧化应激,即氧化剂和抗氧化剂之间的不平衡,从而损害组织。大脑由于其脂质成分、高氧气消耗率和低水平的抗氧化酶,特别容易受到氧化应激的影响。此外,高氧会导致血管收缩和大脑氧气供应减少,对氧化还原平衡和神经退行性过程构成挑战。研究表明,高氧诱导的脑损伤的严重程度随启发的氧气浓度和暴露时间的长短而变化。因此,仔细评估补充氧气的益处和风险之间的平衡至关重要,尤其是在临床环境中。
{"title":"Hyperoxia and brain: the link between necessity and injury from a molecular perspective","authors":"Richard Simon Machado, Khiany Mathias, Larissa Joaquim, Rafaella Willig de Quadros, Gislaine Tezza Rezin, Fabricia Petronilho","doi":"10.1007/s12640-024-00702-6","DOIUrl":"https://doi.org/10.1007/s12640-024-00702-6","url":null,"abstract":"<p>Oxygen (O<sub>2</sub>) supplementation is commonly used to treat hypoxia in patients with respiratory failure. However, indiscriminate use can lead to hyperoxia, a condition detrimental to living tissues, particularly the brain. The brain is sensitive to reactive oxygen species (ROS) and inflammation caused by high concentrations of O<sub>2</sub>, which can result in brain damage and mitochondrial dysfunction, common features of neurodegenerative disorders. Hyperoxia leads to increased production of ROS, causing oxidative stress, an imbalance between oxidants and antioxidants, which can damage tissues. The brain is particularly vulnerable to oxidative stress due to its lipid composition, high O<sub>2</sub> consumption rate, and low levels of antioxidant enzymes. Moreover, hyperoxia can cause vasoconstriction and decreased O<sub>2</sub> supply to the brain, posing a challenge to redox balance and neurodegenerative processes. Studies have shown that the severity of hyperoxia-induced brain damage varies with inspired O<sub>2</sub> concentration and duration of exposure. Therefore, careful evaluation of the balance between benefits and risks of O<sub>2</sub> supplementation, especially in clinical settings, is crucial.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"94 1","pages":""},"PeriodicalIF":3.7,"publicationDate":"2024-04-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140598751","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: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