首页 > 最新文献

Neurochemical Research最新文献

英文 中文
Sinomenine Modifies Parkinson’s Disease Through Nrf2 Activation 青藤碱通过Nrf2激活改变帕金森病
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-18 DOI: 10.1007/s11064-025-04557-x
Lingling Zhu, Lin Lin, Zhenyu Yang, Hongxia Tang, Jinping Wu, Zeyi Kang, Yi Feng, Binbin Zheng, Qirou Hu, Shanshan Wang, Cuimin Liu, Zhengli Jiang, Suzhi Liu, Gang Wu

Background

Disease-modifying treatments for Parkinson’s disease (PD) are urgently needed, with the Nrf2/ARE pathway a promising target. This study aims to explore the effects of sinomenine on PD and Nrf2/ARE activation.

Methods

6-OHDA-treated Parkinsonian SH-SY5Y cells and rats were used. Apoptosis and cell viability were measured using flow cytometry and CCK-8 assays. Nrf2 and its downstream proteins were assessed by Western blotting, while ROS levels were detected with fluorescent dyes. Nrf2 silencing via shRNA evaluated sinomenine’s dependence on Nrf2 activation. In vivo, behavioral changes, tyrosine hydroxylase (TH) levels and malondialdehyde (MDA) levels were measured. Microglial inflammation was analyzed by measuring TNF-α and IL-1β expression and cytoskeleton analysis. Nrf2 nuclear translocation was verified by Western blotting and molecular docking was performed.

Results

Sinomenine reduced apoptosis and ROS, improved cell viability, upregulated Nrf2 and antioxidant enzyme expression. The protective effects against apoptosis were abolished by Nrf2 silencing. In PD animals, sinomenine improved motor deficits, enhanced Nrf2, GCLC, GCLM, NQO1, and HO-1 expression, decreased MDA levels, increased TH levels in the striatum and maintained count of dopaminergic neurons in substantia nigra. Additionally, it suppressed TNF-α and IL-1β levels in brain tissue and blood, preserving normal microglial morphology and reducing neuroinflammation. Sinomenine promoted Nrf2 nuclear translocation and showed high Keap1 affinity in docking.

Conclusions

Sinomenine activates the Nrf2/ARE pathway, mitigating oxidative stress and inflammation in PD models, possibly through Keap1 binding and Nrf2 nuclear translocation. These findings suggest sinomenine may serve as a potential disease-modifying therapy for Parkinson’s disease, pending further clinical validation.

目前迫切需要改善帕金森病(PD)的治疗方法,而Nrf2/ are通路是一个有希望的靶点。本研究旨在探讨青藤碱对PD和Nrf2/ARE激活的影响。方法采用ohda处理的帕金森SH-SY5Y细胞和大鼠。采用流式细胞术和CCK-8检测细胞凋亡和细胞活力。Western blotting检测Nrf2及其下游蛋白,荧光染料检测ROS水平。通过shRNA沉默Nrf2评估青藤碱对Nrf2激活的依赖性。在体内,测定行为变化、酪氨酸羟化酶(TH)水平和丙二醛(MDA)水平。通过检测TNF-α和IL-1β的表达和细胞骨架分析来分析小胶质细胞炎症。Western blotting验证Nrf2核易位,并进行分子对接。结果青藤碱可减少细胞凋亡和ROS,提高细胞活力,上调Nrf2和抗氧化酶的表达。Nrf2沉默可消除对细胞凋亡的保护作用。在PD动物中,青叶碱改善运动缺陷,增强Nrf2、GCLC、GCLM、NQO1和HO-1表达,降低纹状体MDA水平,增加TH水平,维持黑质多巴胺能神经元数量。此外,它还能抑制脑组织和血液中TNF-α和IL-1β的水平,保持正常的小胶质细胞形态,减轻神经炎症。青葱碱促进Nrf2核易位,在对接过程中表现出较高的Keap1亲和力。结论青叶碱激活Nrf2/ARE通路,减轻PD模型的氧化应激和炎症,可能通过Keap1结合和Nrf2核易位实现。这些发现表明青藤碱可能作为帕金森病的潜在疾病改善疗法,有待进一步的临床验证。
{"title":"Sinomenine Modifies Parkinson’s Disease Through Nrf2 Activation","authors":"Lingling Zhu,&nbsp;Lin Lin,&nbsp;Zhenyu Yang,&nbsp;Hongxia Tang,&nbsp;Jinping Wu,&nbsp;Zeyi Kang,&nbsp;Yi Feng,&nbsp;Binbin Zheng,&nbsp;Qirou Hu,&nbsp;Shanshan Wang,&nbsp;Cuimin Liu,&nbsp;Zhengli Jiang,&nbsp;Suzhi Liu,&nbsp;Gang Wu","doi":"10.1007/s11064-025-04557-x","DOIUrl":"10.1007/s11064-025-04557-x","url":null,"abstract":"<div><h3>Background</h3><p>Disease-modifying treatments for Parkinson’s disease (PD) are urgently needed, with the Nrf2/ARE pathway a promising target. This study aims to explore the effects of sinomenine on PD and Nrf2/ARE activation.</p><h3>Methods</h3><p>6-OHDA-treated Parkinsonian SH-SY5Y cells and rats were used. Apoptosis and cell viability were measured using flow cytometry and CCK-8 assays. Nrf2 and its downstream proteins were assessed by Western blotting, while ROS levels were detected with fluorescent dyes. Nrf2 silencing via shRNA evaluated sinomenine’s dependence on Nrf2 activation. In vivo, behavioral changes, tyrosine hydroxylase (TH) levels and malondialdehyde (MDA) levels were measured. Microglial inflammation was analyzed by measuring TNF-α and IL-1β expression and cytoskeleton analysis. Nrf2 nuclear translocation was verified by Western blotting and molecular docking was performed.</p><h3>Results</h3><p>Sinomenine reduced apoptosis and ROS, improved cell viability, upregulated Nrf2 and antioxidant enzyme expression. The protective effects against apoptosis were abolished by Nrf2 silencing. In PD animals, sinomenine improved motor deficits, enhanced Nrf2, GCLC, GCLM, NQO1, and HO-1 expression, decreased MDA levels, increased TH levels in the striatum and maintained count of dopaminergic neurons in substantia nigra. Additionally, it suppressed TNF-α and IL-1β levels in brain tissue and blood, preserving normal microglial morphology and reducing neuroinflammation. Sinomenine promoted Nrf2 nuclear translocation and showed high Keap1 affinity in docking.</p><h3>Conclusions</h3><p>Sinomenine activates the Nrf2/ARE pathway, mitigating oxidative stress and inflammation in PD models, possibly through Keap1 binding and Nrf2 nuclear translocation. These findings suggest sinomenine may serve as a potential disease-modifying therapy for Parkinson’s disease, pending further clinical validation.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 5","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145073926","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
Special Collection of Neurochemical Research Volume 50, 2025 in Remembrance of Prof. Arne Schousboe 神经化学研究特别集第50卷,2025年纪念阿恩·斯库伯教授
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-12 DOI: 10.1007/s11064-025-04526-4
Arne Schousboe, Henry Sershen
{"title":"Special Collection of Neurochemical Research Volume 50, 2025 in Remembrance of Prof. Arne Schousboe","authors":"Arne Schousboe,&nbsp;Henry Sershen","doi":"10.1007/s11064-025-04526-4","DOIUrl":"10.1007/s11064-025-04526-4","url":null,"abstract":"","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 5","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145037229","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
Riboflavin (Vitamin B2) Accumulation Modulates Neuronal Cellular Homeostasis in Typical Brain Development and Cerebral Palsy 核黄素(维生素B2)积累调节典型脑发育和脑瘫的神经元细胞稳态
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-12 DOI: 10.1007/s11064-025-04552-2
Eulália Rebeca Silva-Araújo, Eduardo Padrón-Hernández, Ana Elisa Toscano, Osmar Henrique dos Santos Júnior, Joaci Pereira dos Santos Júnior, Henrique José Cavalcanti Bezerra Gouveia, Adriana Soares de Carvalho, Janaína Viana de Melo, Luiz Alberto Reis Mattos-Júnior, Raul Manhães-de-Castro

The developing brain requires high energy demands and metabolic efforts to regulate oxidative stress and myelination. Early insults cause mitochondrial dysfunction and compromise these pathways, potentially leading to cerebral palsy (CP), a severe and incurable neurological disorder that begins in childhood. Through a rodent preclinical study, we demonstrated that vitamin B2 (riboflavin), administered at a high dose (100 mg/kg), is accumulated in healthy (B2C) or paralytic (B2CP) brains and participates in neurodevelopment. Redox homeostasis was maintained in B2C through decreased malondialdehyde and carbonyls and increased glutathione-S-transferase activity. In B2CP rodents, there was a reduction in carbonyls and increased superoxide dismutase activity. Mitochondrial morphometric analysis suggests that riboflavin treatment increases biogenesis in controls and reduces mitochondrial deformation in CP. Ultrastructural analysis revealed increased myelin sheath thickness in B2C. Additionally, myelin figure formation and mitochondrial and axonal disintegration in CP were reduced by B2. Our evidence supports vitamin B2 accumulation as a beneficial mechanism to support energy homeostasis and mitochondrial demands that occur during typical neurodevelopment or in the face of CP.

发育中的大脑需要高能量需求和代谢努力来调节氧化应激和髓鞘形成。早期损伤会导致线粒体功能障碍,破坏这些通路,可能导致脑瘫(CP),这是一种始于儿童时期的严重且无法治愈的神经系统疾病。通过啮齿动物临床前研究,我们证明了高剂量(100 mg/kg)的维生素B2(核黄素)在健康(B2C)或瘫痪(B2CP)大脑中积累并参与神经发育。B2C通过降低丙二醛和羰基以及增加谷胱甘肽- s转移酶活性来维持氧化还原稳态。在B2CP啮齿动物中,羰基减少,超氧化物歧化酶活性增加。线粒体形态分析表明,核黄素治疗增加了对照组的生物发生,减少了CP的线粒体变形。超微结构分析显示,B2C的髓鞘厚度增加。此外,B2可减少CP的髓鞘图形成和线粒体和轴突解体。我们的证据支持维生素B2的积累是一个有益的机制,支持能量稳态和线粒体需求,发生在典型的神经发育或面对CP。
{"title":"Riboflavin (Vitamin B2) Accumulation Modulates Neuronal Cellular Homeostasis in Typical Brain Development and Cerebral Palsy","authors":"Eulália Rebeca Silva-Araújo,&nbsp;Eduardo Padrón-Hernández,&nbsp;Ana Elisa Toscano,&nbsp;Osmar Henrique dos Santos Júnior,&nbsp;Joaci Pereira dos Santos Júnior,&nbsp;Henrique José Cavalcanti Bezerra Gouveia,&nbsp;Adriana Soares de Carvalho,&nbsp;Janaína Viana de Melo,&nbsp;Luiz Alberto Reis Mattos-Júnior,&nbsp;Raul Manhães-de-Castro","doi":"10.1007/s11064-025-04552-2","DOIUrl":"10.1007/s11064-025-04552-2","url":null,"abstract":"<div><p>The developing brain requires high energy demands and metabolic efforts to regulate oxidative stress and myelination. Early insults cause mitochondrial dysfunction and compromise these pathways, potentially leading to cerebral palsy (CP), a severe and incurable neurological disorder that begins in childhood. Through a rodent preclinical study, we demonstrated that vitamin B2 (riboflavin), administered at a high dose (100 mg/kg), is accumulated in healthy (B2C) or paralytic (B2CP) brains and participates in neurodevelopment. Redox homeostasis was maintained in B2C through decreased malondialdehyde and carbonyls and increased glutathione-S-transferase activity. In B2CP rodents, there was a reduction in carbonyls and increased superoxide dismutase activity. Mitochondrial morphometric analysis suggests that riboflavin treatment increases biogenesis in controls and reduces mitochondrial deformation in CP. Ultrastructural analysis revealed increased myelin sheath thickness in B2C. Additionally, myelin figure formation and mitochondrial and axonal disintegration in CP were reduced by B2. Our evidence supports vitamin B2 accumulation as a beneficial mechanism to support energy homeostasis and mitochondrial demands that occur during typical neurodevelopment or in the face of CP.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 5","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145037227","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
Methylsulfonylmethane Mitigates Neurobehavioral Impairment, Oxidative Stress, Changes in Trace Elements and Clock Expression in Alcohol and Circadian-Disrupted C57BL/6J Mice Brain 甲基磺酰甲烷减轻酒精和昼夜节律紊乱的C57BL/6J小鼠大脑中神经行为损伤、氧化应激、微量元素和时钟表达的变化
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-12 DOI: 10.1007/s11064-025-04551-3
Indrani Paramasivan Latha Laxmi, Ramasamy Tamizhselvi

Alcohol consumption can affect the brain due to an elevation in oxidative stress and inflammation. Alcoholism influences the brain homeostasis and is often associated with cognitive, emotional and behavioral changes. Excessive alcohol intake can lead to poor sleep quality, and individuals with alcohol use disorder generally develop insomnia. Alcohol use affects the expression of clock genes, altering the physiological and immune function regulated by the biological clock, and hence elevating the production of reactive oxygen species. This study aimed to explore the alterations in neurobehavioral function associated with circadian disturbance and alcohol exposure, and to understand the potential therapeutic effect of methylsulfonylmethane on alcohol-administered and circadian-disrupted C57BL/6J mice. The sleep cycle of the mice was disturbed by 10 h light/10 h dark exposure, and 25% w/v alcohol was administered to the mice intragastrically. The concentration of trace elements in the mouse brain was measured using Inductively Coupled Mass Spectrometry (ICP-MS). An increase in anxiety-like behavior was observed in the mice exposed to alcohol and the circadian-disrupted groups. The administration of the drug significantly increased the expression of core clock genes in the alcohol and circadian-disrupted group. The levels of calcium and iron were increased in the MSM-administered mice, reversing the effects of circadian disruption and the alcohol-exposed group of mice. Our findings suggest that alcohol and circadian disturbance impact neurological function through alterations in immune homeostasis. MSM treatment can improve the expression of clock genes in alcohol and circadian-disrupted conditions, highlighting its neuroprotective potential by reducing inflammation.

由于氧化应激和炎症的升高,饮酒会影响大脑。酒精中毒影响大脑的内稳态,通常与认知、情绪和行为的改变有关。过量饮酒会导致睡眠质量差,有酒精使用障碍的人通常会患上失眠。饮酒影响生物钟基因的表达,改变生物钟调节的生理和免疫功能,从而提高活性氧的产生。本研究旨在探讨与昼夜节律紊乱和酒精暴露相关的神经行为功能改变,并了解甲基磺酰甲烷对酒精给药和昼夜节律紊乱的C57BL/6J小鼠的潜在治疗作用。10 h光照/10 h黑暗暴露干扰小鼠睡眠周期,小鼠灌胃25% w/v酒精。采用电感耦合质谱法(ICP-MS)测定小鼠脑内微量元素浓度。在暴露于酒精和昼夜节律紊乱组的小鼠中,观察到焦虑样行为的增加。在酒精和昼夜节律紊乱组中,给药显著增加了核心时钟基因的表达。服用msm的小鼠体内的钙和铁含量增加,逆转了昼夜节律紊乱和酒精暴露组小鼠的影响。我们的研究结果表明,酒精和昼夜节律紊乱通过改变免疫稳态影响神经功能。男同性恋者治疗可以改善酒精和昼夜节律紊乱情况下生物钟基因的表达,通过减少炎症突出其神经保护潜力。
{"title":"Methylsulfonylmethane Mitigates Neurobehavioral Impairment, Oxidative Stress, Changes in Trace Elements and Clock Expression in Alcohol and Circadian-Disrupted C57BL/6J Mice Brain","authors":"Indrani Paramasivan Latha Laxmi,&nbsp;Ramasamy Tamizhselvi","doi":"10.1007/s11064-025-04551-3","DOIUrl":"10.1007/s11064-025-04551-3","url":null,"abstract":"<div><p>Alcohol consumption can affect the brain due to an elevation in oxidative stress and inflammation. Alcoholism influences the brain homeostasis and is often associated with cognitive, emotional and behavioral changes. Excessive alcohol intake can lead to poor sleep quality, and individuals with alcohol use disorder generally develop insomnia. Alcohol use affects the expression of clock genes, altering the physiological and immune function regulated by the biological clock, and hence elevating the production of reactive oxygen species. This study aimed to explore the alterations in neurobehavioral function associated with circadian disturbance and alcohol exposure, and to understand the potential therapeutic effect of methylsulfonylmethane on alcohol-administered and circadian-disrupted C57BL/6J mice. The sleep cycle of the mice was disturbed by 10 h light/10 h dark exposure, and 25% w/v alcohol was administered to the mice intragastrically. The concentration of trace elements in the mouse brain was measured using Inductively Coupled Mass Spectrometry (ICP-MS). An increase in anxiety-like behavior was observed in the mice exposed to alcohol and the circadian-disrupted groups. The administration of the drug significantly increased the expression of core clock genes in the alcohol and circadian-disrupted group. The levels of calcium and iron were increased in the MSM-administered mice, reversing the effects of circadian disruption and the alcohol-exposed group of mice. Our findings suggest that alcohol and circadian disturbance impact neurological function through alterations in immune homeostasis. MSM treatment can improve the expression of clock genes in alcohol and circadian-disrupted conditions, highlighting its neuroprotective potential by reducing inflammation.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 5","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145037228","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
Cholesterol-Lowering Treatment Suppresses Neuromuscular Transmission Via Presynaptic Mechanism at the Mouse Diaphragm Muscle 降胆固醇治疗通过小鼠膈肌突触前机制抑制神经肌肉传递
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-11 DOI: 10.1007/s11064-025-04550-4
Guzel F. Zakyrjanova, Andrei N. Tsentsevitsky, Valeriya A. Matigorova, Nikita S. Fedorov, Julia G. Odnoshivkina, Guzel V. Sibgatullina, Eva A. Kapliukhina, Arthur R. Giniatullin, Arthur N. Khaziev, Artem I. Malomouzh, Yuri V. Gogolev, Alexey M Petrov

Statins are widely prescribed and effective cholesterol-lowering drugs for the therapy of cerebrovascular and cardiovascular disorders. The main side effects limiting statin use are muscle-related adverse events, including weakness and myopathy. The precise mechanisms of statin-induced muscle damage remain to be elucidated. Possible alterations in neuromuscular transmission might contribute to the statin side effects. Here, we studied the action of one-month treatment with atorvastatin, the most prescribed statin, on the functioning of neuromuscular junctions and related processes in the mouse diaphragm. We found that atorvastatin treatment decreases evoked acetylcholine (ACh) release and involvement of synaptic vesicles in exocytosis during intense nerve activation, as well as recovery of ACh release after tetanic stimulation. This was accompanied by increased immunolabeling of synapsin 1, a protein retaining synaptic vesicles in a non-active pool, and decreased non-quantal ACh release under resting conditions. Additionally, atorvastatin administration decreased perimeters of postsynaptic ACh receptor clusters without signs of muscle denervation. Diaphragm contractile responses to phrenic nerve stimulation at moderate-to-high frequencies and peak inspiratory flow, an indicator of diaphragm function in vivo, were decreased in atorvastatin-treated mice, whereas diaphragm contractions elicited by direct stimulation of muscle fibers were unchanged. Thus, atorvastatin treatment caused a decline in evoked ACh release and synaptic vesicle recruitment into neurotransmission that could lead to a reduction of diaphragm contractile responses to phrenic nerve activity and peak inspiratory flow. These alterations, in combination with decreased non-quantal ACh release and neuromuscular junction size, may contribute to statin-associated muscle symptoms.

他汀类药物被广泛用于治疗脑血管和心血管疾病,是一种有效的降胆固醇药物。限制他汀类药物使用的主要副作用是与肌肉有关的不良事件,包括无力和肌病。他汀类药物引起肌肉损伤的确切机制仍有待阐明。神经肌肉传导的可能改变可能导致他汀类药物的副作用。在这里,我们研究了阿托伐他汀(最常用的他汀类药物)治疗一个月对小鼠膈肌神经肌肉连接和相关过程功能的作用。我们发现,阿托伐他汀治疗减少了神经强烈激活时乙酰胆碱(ACh)的释放和突触囊泡参与胞吐,以及破伤风刺激后ACh释放的恢复。这伴随着突触蛋白1(一种在非活性池中保留突触囊泡的蛋白)免疫标记的增加,以及静息条件下非定量乙酰胆碱释放的减少。此外,阿托伐他汀可减少突触后ACh受体簇的周长,且无肌肉去神经支配的迹象。在阿托伐他汀治疗的小鼠中,膈神经在中高频率刺激下的膈肌收缩反应和呼气流量(体内膈肌功能的一个指标)减弱,而直接刺激肌纤维引起的膈肌收缩没有变化。因此,阿托伐他汀治疗导致诱发的乙酰胆碱释放和突触囊泡募集到神经传递的减少,这可能导致膈神经活动和吸气流量峰值的膈肌收缩反应减少。这些改变,加上非定量乙酰胆碱释放和神经肌肉连接处大小的减少,可能导致他汀类药物相关的肌肉症状。
{"title":"Cholesterol-Lowering Treatment Suppresses Neuromuscular Transmission Via Presynaptic Mechanism at the Mouse Diaphragm Muscle","authors":"Guzel F. Zakyrjanova,&nbsp;Andrei N. Tsentsevitsky,&nbsp;Valeriya A. Matigorova,&nbsp;Nikita S. Fedorov,&nbsp;Julia G. Odnoshivkina,&nbsp;Guzel V. Sibgatullina,&nbsp;Eva A. Kapliukhina,&nbsp;Arthur R. Giniatullin,&nbsp;Arthur N. Khaziev,&nbsp;Artem I. Malomouzh,&nbsp;Yuri V. Gogolev,&nbsp;Alexey M Petrov","doi":"10.1007/s11064-025-04550-4","DOIUrl":"10.1007/s11064-025-04550-4","url":null,"abstract":"<div><p>Statins are widely prescribed and effective cholesterol-lowering drugs for the therapy of cerebrovascular and cardiovascular disorders. The main side effects limiting statin use are muscle-related adverse events, including weakness and myopathy. The precise mechanisms of statin-induced muscle damage remain to be elucidated. Possible alterations in neuromuscular transmission might contribute to the statin side effects. Here, we studied the action of one-month treatment with atorvastatin, the most prescribed statin, on the functioning of neuromuscular junctions and related processes in the mouse diaphragm. We found that atorvastatin treatment decreases evoked acetylcholine (ACh) release and involvement of synaptic vesicles in exocytosis during intense nerve activation, as well as recovery of ACh release after tetanic stimulation. This was accompanied by increased immunolabeling of synapsin 1, a protein retaining synaptic vesicles in a non-active pool, and decreased non-quantal ACh release under resting conditions. Additionally, atorvastatin administration decreased perimeters of postsynaptic ACh receptor clusters without signs of muscle denervation. Diaphragm contractile responses to phrenic nerve stimulation at moderate-to-high frequencies and peak inspiratory flow, an indicator of diaphragm function in vivo, were decreased in atorvastatin-treated mice, whereas diaphragm contractions elicited by direct stimulation of muscle fibers were unchanged. Thus, atorvastatin treatment caused a decline in evoked ACh release and synaptic vesicle recruitment into neurotransmission that could lead to a reduction of diaphragm contractile responses to phrenic nerve activity and peak inspiratory flow. These alterations, in combination with decreased non-quantal ACh release and neuromuscular junction size, may contribute to statin-associated muscle symptoms.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 5","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145028412","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
Silencing PVT1 Alleviates Sevoflurane Anesthesia-Induced Oxidative Stress and Cognitive Dysfunction by Regulating miR-486-5p 沉默PVT1通过调节miR-486-5p减轻七氟醚麻醉诱导的氧化应激和认知功能障碍
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-11 DOI: 10.1007/s11064-025-04537-1
Jing Qian, Xiaoxiao Dai, Zhaoxuan Li

To investigate the role and mechanism of long non-coding RNA PVT1 in sevoflurane-induced oxidative stress and cognitive dysfunction. The expression level of PVT1 and the mRNA expressions of Caspase-3, Bax, and Bcl2 were detected by RT-qPCR. Cell viability and apoptosis rate were evaluated by MTT assay and flow cytometry, respectively. The levels of malondialdehyde (MDA), reactive oxygen species (ROS), and superoxide dismutase (SOD) were determined using commercial kits. The cognitive function of rats was assessed by Morris water maze (MWM) test. Online databases were used to predict the microRNAs (miRNAs) targeted by PVT1, and dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were performed to verify the targeted binding relationship. PVT1 levels were significantly upregulated in hippocampal tissues of rats and HT22 cells treated with sevoflurane. Silencing of PVT1 effectively alleviated sevoflurane-induced cell apoptosis, oxidative stress, and cognitive dysfunction. Mechanistic studies showed that PVT1 targeted miR-486-5p. In sevoflurane-treated hippocampal tissues of rats and HT22 cells, inhibition of miR-486-5p counteracted the protective effects of PVT1 silencing, leading to increased cell apoptosis, exacerbated oxidative stress, and deteriorated cognitive dysfunction. PVT1 silencing mitigates oxidative stress response and cognitive dysfunction by targeting miR-486-5p, providing a novel research perspective for the treatment of sevoflurane-induced nerve injury.

探讨长链非编码RNA PVT1在七氟醚诱导氧化应激和认知功能障碍中的作用及机制。RT-qPCR检测PVT1表达水平及Caspase-3、Bax、Bcl2 mRNA表达水平。分别用MTT法和流式细胞术检测细胞活力和凋亡率。丙二醛(MDA)、活性氧(ROS)和超氧化物歧化酶(SOD)水平采用商用试剂盒测定。采用Morris水迷宫(Morris water maze, MWM)测试大鼠认知功能。利用在线数据库预测PVT1靶向的microRNAs (miRNAs),并通过双荧光素酶报告基因实验和RNA免疫沉淀(RIP)实验验证PVT1靶向结合关系。七氟醚处理大鼠海马组织和HT22细胞中PVT1水平显著上调。PVT1沉默可有效缓解七氟醚诱导的细胞凋亡、氧化应激和认知功能障碍。机制研究表明PVT1靶向miR-486-5p。在七氟烷处理的大鼠海马组织和HT22细胞中,miR-486-5p的抑制抵消了PVT1沉默的保护作用,导致细胞凋亡增加,氧化应激加剧,认知功能障碍恶化。PVT1沉默通过靶向miR-486-5p减轻氧化应激反应和认知功能障碍,为七氟醚诱导的神经损伤的治疗提供了新的研究视角。
{"title":"Silencing PVT1 Alleviates Sevoflurane Anesthesia-Induced Oxidative Stress and Cognitive Dysfunction by Regulating miR-486-5p","authors":"Jing Qian,&nbsp;Xiaoxiao Dai,&nbsp;Zhaoxuan Li","doi":"10.1007/s11064-025-04537-1","DOIUrl":"10.1007/s11064-025-04537-1","url":null,"abstract":"<div><p>To investigate the role and mechanism of long non-coding RNA PVT1 in sevoflurane-induced oxidative stress and cognitive dysfunction. The expression level of PVT1 and the mRNA expressions of Caspase-3, Bax, and Bcl2 were detected by RT-qPCR. Cell viability and apoptosis rate were evaluated by MTT assay and flow cytometry, respectively. The levels of malondialdehyde (MDA), reactive oxygen species (ROS), and superoxide dismutase (SOD) were determined using commercial kits. The cognitive function of rats was assessed by Morris water maze (MWM) test. Online databases were used to predict the microRNAs (miRNAs) targeted by PVT1, and dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were performed to verify the targeted binding relationship. PVT1 levels were significantly upregulated in hippocampal tissues of rats and HT22 cells treated with sevoflurane. Silencing of PVT1 effectively alleviated sevoflurane-induced cell apoptosis, oxidative stress, and cognitive dysfunction. Mechanistic studies showed that PVT1 targeted miR-486-5p. In sevoflurane-treated hippocampal tissues of rats and HT22 cells, inhibition of miR-486-5p counteracted the protective effects of PVT1 silencing, leading to increased cell apoptosis, exacerbated oxidative stress, and deteriorated cognitive dysfunction. PVT1 silencing mitigates oxidative stress response and cognitive dysfunction by targeting miR-486-5p, providing a novel research perspective for the treatment of sevoflurane-induced nerve injury.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 5","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145028411","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
Electroacupuncture Ameliorates Neuroinflammatory Injury in CPSP Rats by Inhibiting the LncRNA MEG3-Mediated Wnt/β-Catenin Signaling Pathway 电针通过抑制LncRNA meg3介导的Wnt/β-Catenin信号通路改善CPSP大鼠神经炎症损伤
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-11 DOI: 10.1007/s11064-025-04547-z
Guihua Tian, Meiyue Wang, Ke He, Xinyi Li, Yang Wu, Huifeng Hao, Fan Zhang, Youxiang Su, Junyi Long, Yi Lin, Zhihao Shang, Liangqing Huang

Electroacupuncture (EA) therapy has been shown to significantly alleviate central poststroke pain (CPSP). However, current research on the mechanisms by which EA relieves CPSP is insufficient. This study explored the role of EA in ameliorating central nervous system inflammation in CPSP rats.The CPSP rat model was established by injecting collagenase IV into the right ventral posterolateral nucleus of the thalamus (VPL). The treatment group was treated with 15 Hz and 2 mA continuous wave EA every other day for a total of 8 sessions. The lncRNA MEG3 (MEG3) was knocked down or overexpressed by adeno-associated virus delivery in vivo in the rat brain. Pain thresholds were measured to assess the hypersensitivity of the rats to pain. Immunofluorescence, Nissl staining and enzyme-linked immunosorbent assay (ELISA) were used to assess the levels of MEG3 and glial fibrillary acidic protein (GFAP) in VPL brain tissue, neuronal injury, and the levels of substance P (SP), TNF-α, IL-1β and IL-6 in VPL brain tissue and serum, respectively. The levels of MEG3, Wnt3a, β-catenin and GFAP in VPL brain tissue were assessed by qRT‒PCR or Western blotting. EA inhibits the expression of MEG3 and neuroinflammatory injury in the VPL brain tissue of CPSP rats, ameliorating hyperalgesia symptoms in CPSP rats. The overexpression of MEG3 weakened the inhibitory effect of EA on the Wnt/β-catenin pathway in the VPL region of the brain, exacerbating pain hypersensitivity and neuroinflammatory damage in the brain hemorrhage regions of CPSP rats. Suppressing the expression of MEG3 in the VPL brain tissue of CPSP rats produced a therapeutic effect similar to that of EA intervention. EA could alleviate neuroinflammation and reduce pain in CPSP rats by suppressing the expression of MEG3. EA could regulate the Wnt/β-catenin signaling pathway via MEG3.

电针(EA)治疗已被证明可以显著减轻中枢性脑卒中后疼痛(CPSP)。然而,目前对EA缓解CPSP的机制研究不足。本研究探讨EA对CPSP大鼠中枢神经系统炎症的改善作用。通过向丘脑右腹侧后外侧核(VPL)注射胶原酶IV建立CPSP大鼠模型。治疗组采用15 Hz、2 mA连续波EA治疗,每隔一天进行一次,共8次。lncRNA MEG3 (MEG3)在大鼠脑内通过腺相关病毒的体内传递被敲低或过表达。测量疼痛阈值以评估大鼠对疼痛的超敏反应。采用免疫荧光法、尼氏染色法和酶联免疫吸附法(ELISA)检测VPL脑组织、神经元损伤组织中MEG3和胶质原纤维酸性蛋白(GFAP)水平,以及VPL脑组织和血清中P物质(SP)、TNF-α、IL-1β和IL-6水平。采用qRT-PCR或Western blotting检测VPL脑组织中MEG3、Wnt3a、β-catenin、GFAP水平。EA可抑制CPSP大鼠VPL脑组织MEG3表达及神经炎症损伤,改善CPSP大鼠痛觉过敏症状。MEG3过表达减弱了EA对脑VPL区Wnt/β-catenin通路的抑制作用,加重了CPSP大鼠脑出血区疼痛超敏反应和神经炎症损伤。抑制CPSP大鼠VPL脑组织中MEG3的表达可产生与EA干预相似的治疗效果。EA可通过抑制MEG3的表达,减轻CPSP大鼠的神经炎症和疼痛。EA可通过MEG3调控Wnt/β-catenin信号通路。
{"title":"Electroacupuncture Ameliorates Neuroinflammatory Injury in CPSP Rats by Inhibiting the LncRNA MEG3-Mediated Wnt/β-Catenin Signaling Pathway","authors":"Guihua Tian,&nbsp;Meiyue Wang,&nbsp;Ke He,&nbsp;Xinyi Li,&nbsp;Yang Wu,&nbsp;Huifeng Hao,&nbsp;Fan Zhang,&nbsp;Youxiang Su,&nbsp;Junyi Long,&nbsp;Yi Lin,&nbsp;Zhihao Shang,&nbsp;Liangqing Huang","doi":"10.1007/s11064-025-04547-z","DOIUrl":"10.1007/s11064-025-04547-z","url":null,"abstract":"<div><p>Electroacupuncture (EA) therapy has been shown to significantly alleviate central poststroke pain (CPSP). However, current research on the mechanisms by which EA relieves CPSP is insufficient. This study explored the role of EA in ameliorating central nervous system inflammation in CPSP rats.The CPSP rat model was established by injecting collagenase IV into the right ventral posterolateral nucleus of the thalamus (VPL). The treatment group was treated with 15 Hz and 2 mA continuous wave EA every other day for a total of 8 sessions. The lncRNA MEG3 (MEG3) was knocked down or overexpressed by adeno-associated virus delivery in vivo in the rat brain. Pain thresholds were measured to assess the hypersensitivity of the rats to pain. Immunofluorescence, Nissl staining and enzyme-linked immunosorbent assay (ELISA) were used to assess the levels of <i>MEG3</i> and glial fibrillary acidic protein (GFAP) in VPL brain tissue, neuronal injury, and the levels of substance P (SP), TNF-α, IL-1β and IL-6 in VPL brain tissue and serum, respectively. The levels of <i>MEG3</i>, Wnt3a, β-catenin and GFAP in VPL brain tissue were assessed by qRT‒PCR or Western blotting. EA inhibits the expression of <i>MEG3</i> and neuroinflammatory injury in the VPL brain tissue of CPSP rats, ameliorating hyperalgesia symptoms in CPSP rats. The overexpression of <i>MEG3</i> weakened the inhibitory effect of EA on the Wnt/β-catenin pathway in the VPL region of the brain, exacerbating pain hypersensitivity and neuroinflammatory damage in the brain hemorrhage regions of CPSP rats. Suppressing the expression of <i>MEG3</i> in the VPL brain tissue of CPSP rats produced a therapeutic effect similar to that of EA intervention. EA could alleviate neuroinflammation and reduce pain in CPSP rats by suppressing the expression of <i>MEG3</i>. EA could regulate the Wnt/β-catenin signaling pathway via <i>MEG3</i>.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 5","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://link.springer.com/content/pdf/10.1007/s11064-025-04547-z.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145028413","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
Salidroside Attenuates Cerebral Ischemia-Reperfusion Injury via ERβ/BNIP3-Mediated Mitochondrial Autophagy Activation in a Rat Model 红红草苷通过ERβ/ bnip3介导的线粒体自噬激活减轻大鼠模型脑缺血再灌注损伤
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-11 DOI: 10.1007/s11064-025-04535-3
Xing Rong, Peipei Lu, Yu Li, Hongxiang Wang, Yuanjia Yue, Huimin Wang, Zhao Ji, Lin Jiang

This study aimed to assess the neuroprotective effects of salidroside (SAL) on cerebral ischemia-reperfusion injury (CIRI) in a rat model and to elucidate the underlying mechanisms, with a focus on the role of estrogen receptor beta (ERβ) and BCL2 interacting protein 3 (BNIP3)-mediated mitochondrial autophagy as potential therapeutic targets in ischemic stroke. A total of 165 female Sprague-Dawley rats were randomly assigned into 11 groups (n = 15 per group). One group served as the control. The remaining animals underwent bilateral ovariectomy and were subsequently allocated into the following groups: ovariectomy-only, middle cerebral artery occlusion/reperfusion (MCAO/R), estradiol control, ERβ inhibitor, two inhibitor arms (inhibitor-only and inhibitor-plus-SAL), three SAL treatment groups (low, mediummitochondrial division, high dose), and a positive control (edaravone). All groups, except the control and ovariectomy-only groups, were subjected to MCAO for one hour followed by 24 h of reperfusion. Neurological function, cerebral infarct volume, blood-brain barrier (BBB) permeability, and brain water content were evaluated. Histopathological alterations were assessed, and transmission electron microscopy was employed to detect autophagosomes. Western blot analysis was performed to quantify protein expression levels of ERβ, BNIP3, NIP3-like protein X, and microtubule-associated protein 1 A/1B-light chain 3. Administration of SAL and edaravone significantly reduced neurological impairment, infarct volume, BBB disruption, and cerebral edema in the MCAO/R model. SAL treatment upregulated ERβ and BNIP3 expression and enhanced mitochondrial autophagy-associated protein levels. These effects were attenuated by the use of ERβ and mitochondrial division inhibitors, indicating a mechanistic link between SAL-mediated neuroprotection and activation of the ERβ/BNIP3 signaling axis. SAL exerts a neuroprotective effect against CIRI in rats, primarily through activation of ERβ and enhancement of BNIP3-mediated mitochondrial autophagy. These findings suggest that modulation of the ERβ/BNIP3 pathway may represent a promising therapeutic approach for ischemic stroke.

本研究旨在评估红红草苷(SAL)对大鼠脑缺血再灌注损伤(CIRI)的神经保护作用,并阐明其潜在机制,重点研究雌激素受体β (ERβ)和BCL2相互作用蛋白3 (BNIP3)介导的线粒体自噬作为缺血性卒中潜在治疗靶点的作用。选取雌性Sprague-Dawley大鼠165只,随机分为11组,每组15只。其中一组作为对照组。其余动物进行双侧卵巢切除术,随后被分配到以下组:卵巢切除术,大脑中动脉闭塞/再灌注(MCAO/R),雌二醇对照组,ERβ抑制剂,两个抑制剂组(仅抑制剂和抑制剂加SAL),三个SAL治疗组(低,中线粒体分裂,高剂量)和阳性对照(依曲拉酮)。除对照组和单纯卵巢切除组外,其余各组均给予MCAO 1 h,再灌注24 h。评估神经功能、脑梗死体积、血脑屏障(BBB)通透性和脑含水量。评估组织病理学改变,并采用透射电镜检测自噬体。Western blot检测ERβ、BNIP3、nip3样蛋白X和微管相关蛋白1a / 1b轻链3的表达水平。在MCAO/R模型中,SAL和依达拉奉可显著降低神经功能损害、梗死体积、血脑屏障破坏和脑水肿。SAL处理上调ERβ和BNIP3表达,增强线粒体自噬相关蛋白水平。这些作用通过使用ERβ和线粒体分裂抑制剂而减弱,这表明sal介导的神经保护与ERβ/BNIP3信号轴的激活之间存在机制联系。SAL对大鼠CIRI具有神经保护作用,主要通过激活ERβ和增强bnip3介导的线粒体自噬。这些发现表明,调节ERβ/BNIP3通路可能是缺血性卒中的一种有希望的治疗方法。
{"title":"Salidroside Attenuates Cerebral Ischemia-Reperfusion Injury via ERβ/BNIP3-Mediated Mitochondrial Autophagy Activation in a Rat Model","authors":"Xing Rong,&nbsp;Peipei Lu,&nbsp;Yu Li,&nbsp;Hongxiang Wang,&nbsp;Yuanjia Yue,&nbsp;Huimin Wang,&nbsp;Zhao Ji,&nbsp;Lin Jiang","doi":"10.1007/s11064-025-04535-3","DOIUrl":"10.1007/s11064-025-04535-3","url":null,"abstract":"<div><p>This study aimed to assess the neuroprotective effects of salidroside (SAL) on cerebral ischemia-reperfusion injury (CIRI) in a rat model and to elucidate the underlying mechanisms, with a focus on the role of estrogen receptor beta (ERβ) and BCL2 interacting protein 3 (BNIP3)-mediated mitochondrial autophagy as potential therapeutic targets in ischemic stroke. A total of 165 female Sprague-Dawley rats were randomly assigned into 11 groups (<i>n</i> = 15 per group). One group served as the control. The remaining animals underwent bilateral ovariectomy and were subsequently allocated into the following groups: ovariectomy-only, middle cerebral artery occlusion/reperfusion (MCAO/R), estradiol control, ERβ inhibitor, two inhibitor arms (inhibitor-only and inhibitor-plus-SAL), three SAL treatment groups (low, mediummitochondrial division, high dose), and a positive control (edaravone). All groups, except the control and ovariectomy-only groups, were subjected to MCAO for one hour followed by 24 h of reperfusion. Neurological function, cerebral infarct volume, blood-brain barrier (BBB) permeability, and brain water content were evaluated. Histopathological alterations were assessed, and transmission electron microscopy was employed to detect autophagosomes. Western blot analysis was performed to quantify protein expression levels of ERβ, BNIP3, NIP3-like protein X, and microtubule-associated protein 1 A/1B-light chain 3. Administration of SAL and edaravone significantly reduced neurological impairment, infarct volume, BBB disruption, and cerebral edema in the MCAO/R model. SAL treatment upregulated ERβ and BNIP3 expression and enhanced mitochondrial autophagy-associated protein levels. These effects were attenuated by the use of ERβ and mitochondrial division inhibitors, indicating a mechanistic link between SAL-mediated neuroprotection and activation of the ERβ/BNIP3 signaling axis. SAL exerts a neuroprotective effect against CIRI in rats, primarily through activation of ERβ and enhancement of BNIP3-mediated mitochondrial autophagy. These findings suggest that modulation of the ERβ/BNIP3 pathway may represent a promising therapeutic approach for ischemic stroke.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 5","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145028414","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
Therapeutic Effect of Physical Activity in a Male Wistar Rat Model of Paraquat and Maneb-Induced Parkinson’s Disease 体育锻炼对雄性Wistar大鼠百草枯和马奈诱发帕金森病模型的治疗作用
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-10 DOI: 10.1007/s11064-025-04543-3
Youssef El Mekhlouf, Nezha Bouhaddou, Amal Dimaoui, Abdessamad Ittorahou, Ahmed Moussaif, Abdelghani Iddar, Mohammed El Mzibri, Abdellatif Bouayyadi, Abdelhalem Mesfioui

Parkinson’s disease (PD) is characterized by impairments in motor control following the degeneration of dopamine-producing neurons located in the substantia nigra pars compacta. Environmental pesticides such as Paraquat (PQ) and Maneb (MB) contribute to the onset of PD by inducing oxidative stress (OS). This study evaluated the therapeutic efficacy of moderate physical activity (PA) on both motor and non-motor symptoms in a Wistar rat model of Paraquat and Maneb (PQ/MB) induced PD. Thirty male Wistar rats were randomly divided into six groups: control, exercise (EX), PQ/MB, PQ/MB + L-dopa, PQ/MB + EX, and PQ/MB + EX + L-dopa. PD was induced via intraperitoneal (IP) injections of PQ (5 mg/kg) and MB (0.05 mg/kg) administered twice weekly for six weeks, followed by four weeks of moderate exercise in the designated groups. Motor and non-motor behaviors were then assessed, and OS markers were analyzed in the prefrontal cortex (PFC), striatum (ST), and hippocampus (HP). In our model, PQ/MB exposure induced characteristic PD symptoms, including motor dysfunction, anxiety, depression, and memory deficits. These symptoms were accompanied by elevated malondialdehyde (MDA) levels and reduced activity of antioxidant enzymes. However, moderate PA significantly improved several parameters: it enhanced coordination and balance, reduced anxiety and depressive-like behaviors, improved memory performance, attenuated lipid peroxidation, and increased antioxidant defense mechanisms, particularly the activity of catalase (CAT) and superoxide dismutase (SOD). These findings suggest that PA is a promising non-pharmacological therapeutic approach for the management of both motor and non-motor symptoms of PD.

帕金森病(PD)的特征是位于黑质致密部产生多巴胺的神经元变性后的运动控制障碍。环境农药如百草枯(PQ)和百草枯(MB)通过诱导氧化应激(OS)参与PD的发病。本研究评估了适度运动(PA)对百草枯和马内布(PQ/MB)诱导的Wistar大鼠PD模型的运动和非运动症状的治疗效果。将30只雄性Wistar大鼠随机分为6组:对照组、运动组、PQ/MB组、PQ/MB +左旋多巴组、PQ/MB + EX组和PQ/MB + EX +左旋多巴组。通过腹腔注射PQ (5 mg/kg)和MB (0.05 mg/kg),每周两次,连续6周诱导PD,然后在指定组进行4周的适度运动。然后评估运动和非运动行为,并分析前额皮质(PFC)、纹状体(ST)和海马(HP)的OS标记。在我们的模型中,PQ/MB暴露诱发了PD的特征性症状,包括运动功能障碍、焦虑、抑郁和记忆缺陷。这些症状伴随着丙二醛(MDA)水平升高和抗氧化酶活性降低。然而,适度的PA显著改善了几个参数:增强协调和平衡,减少焦虑和抑郁样行为,改善记忆表现,减轻脂质过氧化,增加抗氧化防御机制,特别是过氧化氢酶(CAT)和超氧化物歧化酶(SOD)的活性。这些发现表明,PA是一种很有前途的非药物治疗方法,可用于治疗帕金森病的运动和非运动症状。
{"title":"Therapeutic Effect of Physical Activity in a Male Wistar Rat Model of Paraquat and Maneb-Induced Parkinson’s Disease","authors":"Youssef El Mekhlouf,&nbsp;Nezha Bouhaddou,&nbsp;Amal Dimaoui,&nbsp;Abdessamad Ittorahou,&nbsp;Ahmed Moussaif,&nbsp;Abdelghani Iddar,&nbsp;Mohammed El Mzibri,&nbsp;Abdellatif Bouayyadi,&nbsp;Abdelhalem Mesfioui","doi":"10.1007/s11064-025-04543-3","DOIUrl":"10.1007/s11064-025-04543-3","url":null,"abstract":"<div><p>Parkinson’s disease (PD) is characterized by impairments in motor control following the degeneration of dopamine-producing neurons located in the substantia nigra pars compacta. Environmental pesticides such as Paraquat (PQ) and Maneb (MB) contribute to the onset of PD by inducing oxidative stress (OS). This study evaluated the therapeutic efficacy of moderate physical activity (PA) on both motor and non-motor symptoms in a Wistar rat model of Paraquat and Maneb (PQ/MB) induced PD. Thirty male Wistar rats were randomly divided into six groups: control, exercise (EX), PQ/MB, PQ/MB + L-dopa, PQ/MB + EX, and PQ/MB + EX + L-dopa. PD was induced via intraperitoneal (IP) injections of PQ (5 mg/kg) and MB (0.05 mg/kg) administered twice weekly for six weeks, followed by four weeks of moderate exercise in the designated groups. Motor and non-motor behaviors were then assessed, and OS markers were analyzed in the prefrontal cortex (PFC), striatum (ST), and hippocampus (HP). In our model, PQ/MB exposure induced characteristic PD symptoms, including motor dysfunction, anxiety, depression, and memory deficits. These symptoms were accompanied by elevated malondialdehyde (MDA) levels and reduced activity of antioxidant enzymes. However, moderate PA significantly improved several parameters: it enhanced coordination and balance, reduced anxiety and depressive-like behaviors, improved memory performance, attenuated lipid peroxidation, and increased antioxidant defense mechanisms, particularly the activity of catalase (CAT) and superoxide dismutase (SOD). These findings suggest that PA is a promising non-pharmacological therapeutic approach for the management of both motor and non-motor symptoms of PD.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 5","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145021772","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
Astrocyte-Neuron Metabolic Synergies in Neurological Homeostasis and Disease 星形胶质细胞-神经元在神经稳态和疾病中的代谢协同作用
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-09 DOI: 10.1007/s11064-025-04548-y
Jiahao Dong, Zihan Gao, Mingrui Liu, Binglu Qian, Cheng Yuan, Hui Liu, Ni Rao, Yingjiao Liu

Metabolic synergy between astrocytes and neurons is key to maintaining normal brain function. As the main supporting cells in the brain, astrocytes work closely with neurons through intercellular metabolic synergy networks to jointly regulate energy metabolism, lipid metabolism, synaptic transmission, and cerebral blood flow. This important synergy is often disrupted in neurological diseases such as Alzheimer’s disease, Parkinson’s disease, and stroke. This study systematically explores the physiological basis of this intercellular collaboration and its dysfunctional manifestations in the aforementioned diseases, and provides detailed insights into how abnormalities in specific collaborative pathways (such as impaired lactate transport, disrupted glutamate cycling, or lipid processing defects) significantly contribute to disease progression. By elucidating the molecular mechanisms underlying these collaborative impairments, this study aims to identify potential therapeutic targets, with the core strategy being to restore these critical intercellular collaborative relationships to alleviate neurological diseases.

星形胶质细胞和神经元之间的代谢协同作用是维持正常脑功能的关键。星形胶质细胞作为脑内的主要支持细胞,通过细胞间代谢协同网络与神经元密切配合,共同调节能量代谢、脂质代谢、突触传递和脑血流。这种重要的协同作用在阿尔茨海默病、帕金森病和中风等神经系统疾病中经常被破坏。本研究系统地探讨了上述疾病中细胞间协同作用的生理基础及其功能失调表现,并提供了具体协同通路异常(如乳酸转运受损、谷氨酸循环中断或脂质加工缺陷)如何显著促进疾病进展的详细见解。通过阐明这些协同性损伤的分子机制,本研究旨在确定潜在的治疗靶点,其核心策略是恢复这些关键的细胞间协作关系,以减轻神经系统疾病。
{"title":"Astrocyte-Neuron Metabolic Synergies in Neurological Homeostasis and Disease","authors":"Jiahao Dong,&nbsp;Zihan Gao,&nbsp;Mingrui Liu,&nbsp;Binglu Qian,&nbsp;Cheng Yuan,&nbsp;Hui Liu,&nbsp;Ni Rao,&nbsp;Yingjiao Liu","doi":"10.1007/s11064-025-04548-y","DOIUrl":"10.1007/s11064-025-04548-y","url":null,"abstract":"<div><p>Metabolic synergy between astrocytes and neurons is key to maintaining normal brain function. As the main supporting cells in the brain, astrocytes work closely with neurons through intercellular metabolic synergy networks to jointly regulate energy metabolism, lipid metabolism, synaptic transmission, and cerebral blood flow. This important synergy is often disrupted in neurological diseases such as Alzheimer’s disease, Parkinson’s disease, and stroke. This study systematically explores the physiological basis of this intercellular collaboration and its dysfunctional manifestations in the aforementioned diseases, and provides detailed insights into how abnormalities in specific collaborative pathways (such as impaired lactate transport, disrupted glutamate cycling, or lipid processing defects) significantly contribute to disease progression. By elucidating the molecular mechanisms underlying these collaborative impairments, this study aims to identify potential therapeutic targets, with the core strategy being to restore these critical intercellular collaborative relationships to alleviate neurological diseases.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 5","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145011796","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
Neurochemical 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