Neurotoxicity Research最新文献

筛选
英文 中文
Restraint Stress Exacerbates Apoptosis in a 6-OHDA Animal Model of Parkinson Disease. 抑制应激加剧6-OHDA帕金森病动物模型的细胞凋亡
IF 3.7 3区 医学
Neurotoxicity Research Pub Date : 2023-04-01 DOI: 10.1007/s12640-022-00630-3
Sara El Idrissi, Nada Fath, Hind Ibork, Khalid Taghzouti, Meryem Alamy, Oualid Abboussi
{"title":"Restraint Stress Exacerbates Apoptosis in a 6-OHDA Animal Model of Parkinson Disease.","authors":"Sara El Idrissi,&nbsp;Nada Fath,&nbsp;Hind Ibork,&nbsp;Khalid Taghzouti,&nbsp;Meryem Alamy,&nbsp;Oualid Abboussi","doi":"10.1007/s12640-022-00630-3","DOIUrl":"https://doi.org/10.1007/s12640-022-00630-3","url":null,"abstract":"<p><p>Activation of the apoptotic pathway has been associated with promoting neuronal cell death in the pathophysiology of Parkinson disease (PD). Nonetheless, the mechanisms by which it may occur remain unclear. It has been suggested that stress-induced oxidation and potential apoptosis may play a major role in the progression of PD. Thus, in this study, we aimed to investigate the effect of subchronic restraint stress on striatal dopaminergic activity, iron, p53, caspase-3, and plasmatic acetylcholinesterase (AChE) levels in male Wistar rat model of PD induced by administration of 6-hydroxydopamine (6-OHDA) in the medial forebrain bundle (MFB). The obtained results showed that restraint stress exacerbates motor coordination deficits and anxiety in animals treated with 6-OHDA in comparison to animals receiving saline, and it had no effect on object recognition memory. On another hand, 6-OHDA decreased dopamine (DA) levels, increased iron accumulation, and induced overexpression of the pro-apoptotic factors caspase-3, p53, and AChE. More interestingly, post-lesion restraint stress exacerbated the expression of caspase-3 and AChE without affecting p53 expression. These findings suggest that subchronic stress may accentuate apoptosis and may contribute to DA neuronal loss in the striatal regions and possibly exacerbate the progression of PD.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"41 2","pages":"166-176"},"PeriodicalIF":3.7,"publicationDate":"2023-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9153378","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
Ioversol Induced Microglia Proinflammatory Activation and Oxidative Stress in Rats. Ioversol诱导大鼠小胶质细胞促炎激活和氧化应激。
IF 3.7 3区 医学
Neurotoxicity Research Pub Date : 2023-04-01 DOI: 10.1007/s12640-022-00629-w
Tao Li, Lili Zhao, Hong Fan, Zhiyang Chen, Ye Li, Meijuan Dang, Ziwei Lu, Jialiang Lu, Qiao Huang, Heying Wang, Shengxi Wu, Guilian Zhang, Fang Kuang
{"title":"Ioversol Induced Microglia Proinflammatory Activation and Oxidative Stress in Rats.","authors":"Tao Li,&nbsp;Lili Zhao,&nbsp;Hong Fan,&nbsp;Zhiyang Chen,&nbsp;Ye Li,&nbsp;Meijuan Dang,&nbsp;Ziwei Lu,&nbsp;Jialiang Lu,&nbsp;Qiao Huang,&nbsp;Heying Wang,&nbsp;Shengxi Wu,&nbsp;Guilian Zhang,&nbsp;Fang Kuang","doi":"10.1007/s12640-022-00629-w","DOIUrl":"https://doi.org/10.1007/s12640-022-00629-w","url":null,"abstract":"<p><p>Contrast-induced encephalopathy (CIE) following angiography, though not often and reversible, can in some cases lead to permanent neurological dysfunction. To identify how neuroinflammation is involved in CIE, we investigated microglia responses to a bolus injection of ioversol in the internal carotid artery (ICA) in rats. MicroCT scanning indicated that the injected ioversol was cleared from the rat's brain within 25 min. However, proinflammatory activated and significantly increased microglia were found in the rat occipital cortex at 1 day, and the number of blood vessel-associated microglia was still significantly higher at 3-day post-injection, compared with sham- and PBS-treated rats. Moreover, significantly upregulated malondialdehyde (MDA), downregulated superoxide dismutase (SOD) levels, and elevated proinflammatory cytokines were observed in the brain of rats treated with ioversol. Ioversol administration decreased cell viability of primarily cultured microglia and induced significant proinflammatory activation. Furthermore, ioversol remarkably upregulated astrocytic aquaporin (AQP) 4 expression in the rats brain, and transwell cultures showed significantly enhanced microglia migrating to ioversol-treated endothelial cells. Immediate injection of edaravone dexborneol, a novel antioxidative drug, after ioversol injection effectively rescued ioversol-induced neuroinflammation. Together, these findings suggest that ioversol induced neuroinflammation and oxidative stress in the brain via microglia activation in a direct and indirect manner, which might contribute to the pathogenesis of CIE.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"41 2","pages":"149-165"},"PeriodicalIF":3.7,"publicationDate":"2023-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9151321","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
Neuroprotective and Immunomodulatory Effects of Probiotics in a Rat Model of Parkinson's Disease. 益生菌在帕金森病大鼠模型中的神经保护和免疫调节作用。
IF 3.7 3区 医学
Neurotoxicity Research Pub Date : 2023-04-01 DOI: 10.1007/s12640-022-00627-y
Irving Parra, Isabel Martínez, Lizbeth Vásquez-Celaya, Jose L Gongora-Alfaro, Yousef Tizabi, Liliana Mendieta
{"title":"Neuroprotective and Immunomodulatory Effects of Probiotics in a Rat Model of Parkinson's Disease.","authors":"Irving Parra,&nbsp;Isabel Martínez,&nbsp;Lizbeth Vásquez-Celaya,&nbsp;Jose L Gongora-Alfaro,&nbsp;Yousef Tizabi,&nbsp;Liliana Mendieta","doi":"10.1007/s12640-022-00627-y","DOIUrl":"https://doi.org/10.1007/s12640-022-00627-y","url":null,"abstract":"<p><p>It is now well recognized that a bidirectional relationship between gut microbiota and the brain, referred to as the gut-brain axis, plays a prominent role in maintaining homeostasis and that a disruption in this axis can result in neuroinflammatory response and neurological disorders such as Parkinson's disease (PD). The protective action of probiotics such as Bifidobacterium animalis ssp. lactis Bb12 and Lactobacillus rhamnosus GG in various animal models of PD has been reported. Therefore, in this study, we used an inflammatory model of PD to assess the effects of a combination of these two probiotics (Microbiot<sup>®</sup>) on motor behavior as well as on the response of microglia, including microglia morphology, to gain a better understanding of their mechanism of action. Microbiot<sup>®</sup> (300 µL) was administered orally once daily for 15 days in a lipopolysaccharide-induced PD model using male Wistar rats. Although LPS-induced motor asymmetry in cylinder test was not affected by Microbiot<sup>®</sup>, impairment of motor coordination in the narrow-beam test was significantly reduced by this probiotic. Moreover, Microbiot<sup>®</sup> treatment reduced microglial activation suggesting an anti-inflammatory effect. While further mechanistic investigation of Microbiot<sup>®</sup> in neurodegenerative diseases is warranted, our results support the potential utility of probiotics in PD.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"41 2","pages":"187-200"},"PeriodicalIF":3.7,"publicationDate":"2023-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9154115","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: 1
L-2-Hydroxyglutaric Acid Administration to Neonatal Rats Elicits Marked Neurochemical Alterations and Long-Term Neurobehavioral Disabilities Mediated by Oxidative Stress. l -2-羟基戊二酸给药引起氧化应激介导的神经化学改变和长期神经行为障碍。
IF 3.7 3区 医学
Neurotoxicity Research Pub Date : 2023-04-01 DOI: 10.1007/s12640-022-00625-0
Rafael Teixeira Ribeiro, Andrey Vinícios Soares Carvalho, Rafael Palavro, Luz Elena Durán-Carabali, Ângela Beatris Zemniaçak, Alexandre Umpierrez Amaral, Carlos Alexandre Netto, Moacir Wajner
{"title":"L-2-Hydroxyglutaric Acid Administration to Neonatal Rats Elicits Marked Neurochemical Alterations and Long-Term Neurobehavioral Disabilities Mediated by Oxidative Stress.","authors":"Rafael Teixeira Ribeiro,&nbsp;Andrey Vinícios Soares Carvalho,&nbsp;Rafael Palavro,&nbsp;Luz Elena Durán-Carabali,&nbsp;Ângela Beatris Zemniaçak,&nbsp;Alexandre Umpierrez Amaral,&nbsp;Carlos Alexandre Netto,&nbsp;Moacir Wajner","doi":"10.1007/s12640-022-00625-0","DOIUrl":"https://doi.org/10.1007/s12640-022-00625-0","url":null,"abstract":"<p><p>L-2-Hydroxyglutaric aciduria (L-2-HGA) is an inherited neurometabolic disorder caused by deficient activity of L-2-hydroxyglutarate dehydrogenase. L-2-Hydroxyglutaric acid (L-2-HG) accumulation in the brain and biological fluids is the biochemical hallmark of this disease. Patients present exclusively neurological symptoms and brain abnormalities, particularly in the cerebral cortex, basal ganglia, and cerebellum. Since the pathogenesis of this disorder is still poorly established, we investigated the short-lived effects of an intracerebroventricular injection of L-2-HG to neonatal rats on redox homeostasis in the cerebellum, which is mostly affected in this disorder. We also determined immunohistochemical landmarks of neuronal viability (NeuN), astrogliosis (S100B and GFAP), microglia activation (Iba1), and myelination (MBP and CNPase) in the cerebral cortex and striatum following L-2-HG administration. Finally, the neuromotor development and cognitive abilities were examined. L-2-HG elicited oxidative stress in the cerebellum 6 h after its injection, which was verified by increased reactive oxygen species production, lipid oxidative damage, and altered antioxidant defenses (decreased concentrations of reduced glutathione and increased glutathione peroxidase and superoxide dismutase activities). L-2-HG also decreased the content of NeuN, MBP, and CNPase, and increased S100B, GFAP, and Iba1 in the cerebral cortex and striatum at postnatal days 15 and 75, implying long-standing neuronal loss, demyelination, astrocyte reactivity, and increased inflammatory response, respectively. Finally, L-2-HG administration caused a delay in neuromotor development and a deficit of cognition in adult animals. Importantly, the antioxidant melatonin prevented L-2-HG-induced deleterious neurochemical, immunohistochemical, and behavioral effects, indicating that oxidative stress may be central to the pathogenesis of brain damage in L-2-HGA.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"41 2","pages":"119-140"},"PeriodicalIF":3.7,"publicationDate":"2023-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9207323","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
CircUBE3B High Expression Participates in Sevoflurane-Induced Human Hippocampal Neuron Injury via Targeting miR-326 and Regulating MYD88 Expression. cirbe3b高表达通过靶向miR-326调控MYD88表达参与七氟醚诱导的人海马神经元损伤
IF 3.7 3区 医学
Neurotoxicity Research Pub Date : 2023-02-01 DOI: 10.1007/s12640-022-00617-0
Xinye Qian, Shanshan Zheng, Yingfang Yu
{"title":"CircUBE3B High Expression Participates in Sevoflurane-Induced Human Hippocampal Neuron Injury via Targeting miR-326 and Regulating MYD88 Expression.","authors":"Xinye Qian,&nbsp;Shanshan Zheng,&nbsp;Yingfang Yu","doi":"10.1007/s12640-022-00617-0","DOIUrl":"https://doi.org/10.1007/s12640-022-00617-0","url":null,"abstract":"<p><p>The clinical application of Sevoflurane (Sevo) brings about non-negligible neuron injury, leading to postoperative cognitive dysfunction (POCD). However, related pathogenesis is complex and not fully established. We aimed to disclose the role of circRNA UBE3B (circUBE3B) in neuron injury induced by Sevo. Cell viability and apoptosis were determined by CCK-8 and flow cytometry experiments. Inflammation production was monitored by ELISA. The expression of circUBE3B, miR-326, and myeloid differentiation factor 88 (MYD88) mRNA was assessed by quantitative real-time PCR (qPCR). Apoptosis-associated markers and MYD88 protein were quantified by western blot. The putative binding site between miR-326 and circUBE3B or MYD88 was verified by a dual-luciferase reporter experiment, and their binding was validated by a pull-down assay. Sevo treatment weakened cell viability and promoted cell apoptosis and inflammatory response. CircUBE3B expression was elevated in Sevo-treated neurons. Sevo-induced neuron injury was alleviated by circUBE3B downregulation but aggravated by circUBE3B overexpression. MiR-326 was targeted by circUBE3B, and miR-326 inhibition recovered neuron injury that was repressed by circUBE3B absence in Sevo-treated neurons. MiR-326 interacted with MYD88. MiR-326 enrichment attenuated Sevo-induced neuron injury, while these effects were reversed by MYD88 overexpression. CircUBE3B dysregulation was involved in Sevo-induced human hippocampal neuron injury via targeting the miR-326/MYD88 network.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"41 1","pages":"16-28"},"PeriodicalIF":3.7,"publicationDate":"2023-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10755929","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: 1
Sevoflurane Preconditioning Downregulates GRIA1 Expression to Attenuate Cerebral Ischemia-Reperfusion-Induced Neuronal Injury. 七氟醚预处理下调 GRIA1 表达以减轻脑缺血再灌注诱导的神经元损伤
IF 2.9 3区 医学
Neurotoxicity Research Pub Date : 2023-02-01 Epub Date: 2023-01-03 DOI: 10.1007/s12640-022-00620-5
Ye Li, Zhi Liang, Shuyan Lei, Xiaoning Wu, Tao Yuan, Kai Ma, Kui Chi
{"title":"Sevoflurane Preconditioning Downregulates GRIA1 Expression to Attenuate Cerebral Ischemia-Reperfusion-Induced Neuronal Injury.","authors":"Ye Li, Zhi Liang, Shuyan Lei, Xiaoning Wu, Tao Yuan, Kai Ma, Kui Chi","doi":"10.1007/s12640-022-00620-5","DOIUrl":"10.1007/s12640-022-00620-5","url":null,"abstract":"<p><p>Cerebral ischemia/reperfusion (I/R) injury is the main cause of death following trauma. The neuroprotective effect of sevoflurane (Sev) has been implicated in cerebral I/R injury. However, the mechanisms remain elusive. In this study, we aimed to explore its function in PC12 exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) and in rats challenged with I/R. Sev pretreatment reduced the damage of PC12 cells after OGD/R treatment. Moreover, Sev pretreatment ameliorated neurobehavioral deficits induced by I/R treatment, reduced brain infarct volume, and decreased apoptosis of neurons in hippocampal tissues. Sev pretreatment reduced the surface expression of glutamate receptor 1 (GRIA1) in neurons, while GRIA1 reduced the neuroprotective effects of Sev pretreatment in vitro and in vivo. There was no difference in the surface expression of GRIA2 in rats with I/R and PC12 cells exposed to OGD/R. The ratio of GRIA1/GRIA2 surface expression was reduced, and calcium permeable-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (CP-AMPAR) was blocked by Sev. Together, Sev might exert beneficial effects on cerebral I/R-induced neuronal injury through inhibiting the surface expression of GRIA1 and blocking CP-AMPAR.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"41 1","pages":"29-40"},"PeriodicalIF":2.9,"publicationDate":"2023-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10829345","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
Sodium Para-Aminosalicylic Acid Modulates Autophagy to Lessen Lead-Induced Neurodegeneration in Rat Cortex. 对氨基水杨酸钠调节自噬减轻铅诱导的大鼠皮层神经变性。
IF 3.7 3区 医学
Neurotoxicity Research Pub Date : 2023-02-01 DOI: 10.1007/s12640-022-00615-2
Lei-Lei Wang, Xiao-Juan Zhu, Yuan-Yuan Fang, Yan Li, Yue-Song Zhao, Cui-Liu Gan, Jing-Jing Luo, Shi-Yan Ou, Michael Aschner, Yue-Ming Jiang
{"title":"Sodium Para-Aminosalicylic Acid Modulates Autophagy to Lessen Lead-Induced Neurodegeneration in Rat Cortex.","authors":"Lei-Lei Wang,&nbsp;Xiao-Juan Zhu,&nbsp;Yuan-Yuan Fang,&nbsp;Yan Li,&nbsp;Yue-Song Zhao,&nbsp;Cui-Liu Gan,&nbsp;Jing-Jing Luo,&nbsp;Shi-Yan Ou,&nbsp;Michael Aschner,&nbsp;Yue-Ming Jiang","doi":"10.1007/s12640-022-00615-2","DOIUrl":"https://doi.org/10.1007/s12640-022-00615-2","url":null,"abstract":"<p><p>Lead (Pb) is a common heavy metal contaminant in the environment, and it may perturb autophagy and cause neurodegeneration. Although sodium para-aminosalicylic (PAS-Na) has been shown to protect the brain from lead-induced toxicity, the mechanisms associated with its efficacy have yet to be fully understood. In this study, we evaluated the efficacy of PAS-Na in attenuating the neurotoxic effects of lead, as well as the specific mechanisms that mediate such protection. Lead exposure resulted in weight loss and injury to the liver and kidney, and PAS-Na had a protective effect against this damage. Both short-term and subchronic lead exposure impaired learning ability, and this effect was reversed by PAS-Na intervention. Lead exposure also perturbed autophagic processes through the modulation of autophagy-related factors. Short-term lead exposure downregulated LC3 and beclin1 and upregulated the expression of p62; subchronic lead exposure upregulated the expression of LC3, beclin1, and P62. It follows that PAS-Na had an antagonistic effect on the activation of the above autophagy-related factors. Overall, our novel findings suggest that PAS-Na can protect the rat cortex from lead-induced toxicity by regulating autophagic processes. (1) Short-term lead exposure inhibits autophagy, whereas subchronic lead exposure promotes autophagy. (2) PAS-NA ameliorated the abnormal process of lead-induced autophagy, which had a protective effect on the cerebral cortex.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"41 1","pages":"1-15"},"PeriodicalIF":3.7,"publicationDate":"2023-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10771191","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: 1
Aryl Hydrocarbon Receptor in Glia Cells: A Plausible Glutamatergic Neurotransmission Orchestrator. 神经胶质细胞中的芳基烃受体:一种似是而非的谷氨酸神经传递协调者。
IF 3.7 3区 医学
Neurotoxicity Research Pub Date : 2023-02-01 DOI: 10.1007/s12640-022-00623-2
Janisse Silva-Parra, Cristina Sandu, Marie-Paule Felder-Schmittbuhl, Luisa C Hernández-Kelly, Arturo Ortega
{"title":"Aryl Hydrocarbon Receptor in Glia Cells: A Plausible Glutamatergic Neurotransmission Orchestrator.","authors":"Janisse Silva-Parra,&nbsp;Cristina Sandu,&nbsp;Marie-Paule Felder-Schmittbuhl,&nbsp;Luisa C Hernández-Kelly,&nbsp;Arturo Ortega","doi":"10.1007/s12640-022-00623-2","DOIUrl":"https://doi.org/10.1007/s12640-022-00623-2","url":null,"abstract":"<p><p>Glutamate is the major excitatory amino acid in the vertebrate brain. Glutamatergic signaling is involved in most of the central nervous system functions. Its main components, namely receptors, ion channels, and transporters, are tightly regulated at the transcriptional, translational, and post-translational levels through a diverse array of extracellular signals, such as food, light, and neuroactive molecules. An exquisite and well-coordinated glial/neuronal bidirectional communication is required for proper excitatory amino acid signal transactions. Biochemical shuttles such as the glutamate/glutamine and the astrocyte-neuronal lactate represent the fundamental involvement of glial cells in glutamatergic transmission. In fact, the disruption of any of these coordinated biochemical intercellular cascades leads to an excitotoxic insult that underlies some aspects of most of the neurodegenerative diseases characterized thus far. In this contribution, we provide a comprehensive summary of the involvement of the Aryl hydrocarbon receptor, a ligand-dependent transcription factor in the gene expression regulation of glial glutamate transporters. These receptors might serve as potential targets for the development of novel strategies for the treatment of neurodegenerative diseases.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"41 1","pages":"103-117"},"PeriodicalIF":3.7,"publicationDate":"2023-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10772002","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
Catalpol Mitigates Alzheimer's Disease Progression by Promoting the Expression of Neural Stem Cell Exosomes Released miR-138-5p. 梓醇通过促进神经干细胞外泌体释放的miR-138-5p的表达来缓解阿尔茨海默病的进展
IF 3.7 3区 医学
Neurotoxicity Research Pub Date : 2023-02-01 DOI: 10.1007/s12640-022-00626-z
Shengxi Meng, Huize Chen, Chunjun Deng, Zeyu Meng
{"title":"Catalpol Mitigates Alzheimer's Disease Progression by Promoting the Expression of Neural Stem Cell Exosomes Released miR-138-5p.","authors":"Shengxi Meng,&nbsp;Huize Chen,&nbsp;Chunjun Deng,&nbsp;Zeyu Meng","doi":"10.1007/s12640-022-00626-z","DOIUrl":"https://doi.org/10.1007/s12640-022-00626-z","url":null,"abstract":"<p><p>Alzheimer's disease (Alzheimer's disease, AD) is a neurodegenerative disease characterized by senile plaque deposition and neurofibrillary tangles. The pathogenesis of AD is complicated and the drugs used to treat AD are single-targeted drugs, which can only improve or alleviate the symptoms of patients, but cannot delay or prevent the progress of the disease. Because of its ability to act on multiple targets, multiple systems, multiple links, and multiple pathways, Chinese herbal compound prescriptions have shown unique advantages in the research and treatment of AD. Our previous study has demonstrated the protect role of the Chinese medicine Rehmannia in AD. However, the underlying mechanism remains unclear. In the present study, both in vitro and vivo experiments were employed, and we found Catalpol (Ca), the main extract of Rehmannia, could mitigate AD progression both in vitro and in vivo by promoting miR-138-5p level in neural stem cell secreted exosomes.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"41 1","pages":"41-56"},"PeriodicalIF":3.7,"publicationDate":"2023-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9944361/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10761522","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: 2
Therapeutic Molecular Insights into the Active Engagement of Cannabinoids in the Therapy of Parkinson's Disease: A Novel and Futuristic Approach. 治疗分子洞察大麻素在帕金森病治疗中的积极参与:一种新颖和未来的方法。
IF 3.7 3区 医学
Neurotoxicity Research Pub Date : 2023-02-01 DOI: 10.1007/s12640-022-00619-y
Vaibhav Jain, Tapan Behl, Aayush Sehgal, Sukhbir Singh, Neelam Sharma, Hafiz A Makeen, Mohammed Albratty, Abdulkarim M Meraya, Asim Najmi
{"title":"Therapeutic Molecular Insights into the Active Engagement of Cannabinoids in the Therapy of Parkinson's Disease: A Novel and Futuristic Approach.","authors":"Vaibhav Jain,&nbsp;Tapan Behl,&nbsp;Aayush Sehgal,&nbsp;Sukhbir Singh,&nbsp;Neelam Sharma,&nbsp;Hafiz A Makeen,&nbsp;Mohammed Albratty,&nbsp;Abdulkarim M Meraya,&nbsp;Asim Najmi","doi":"10.1007/s12640-022-00619-y","DOIUrl":"https://doi.org/10.1007/s12640-022-00619-y","url":null,"abstract":"<p><p>Parkinson's disease is a neurodegenerative disorder which is characterised mostly by loss of dopaminergic nerve cells throughout the nigral area mainly as a consequence of oxidative stress. Muscle stiffness, disorganised bodily responses, disturbed sleep, weariness, amnesia, and voice impairment are all symptoms of dopaminergic neuron degeneration and existing symptomatic treatments are important to arrest additional neuronal death. Some cannabinoids have recently been demonstrated as robust antioxidants that might protect the nerve cells from degeneration even when cannabinoid receptors are not triggered. Cannabinoids are likely to have property to slow or presumably cease the steady deterioration of the brain's dopaminergic systems, a condition for which there is now no treatment. The use of cannabinoids in combination with currently available drugs has the potential to introduce a radically new paradigm for treatment of Parkinson's disease, making it immensely useful in the treatment of such a debilitating illness.</p>","PeriodicalId":19193,"journal":{"name":"Neurotoxicity Research","volume":"41 1","pages":"85-102"},"PeriodicalIF":3.7,"publicationDate":"2023-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10744511","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: 2
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信