首页 > 最新文献

Neurochemical Research最新文献

英文 中文
Myrtenal Ameliorates Ischemic Brain Injury Diabetic and Non-Diabetic Rats 桃金娘酸改善糖尿病和非糖尿病大鼠缺血性脑损伤。
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-17 DOI: 10.1007/s11064-025-04629-y
Engin Korkmaz, Asiye Beytur, Yavuz Erden, Kevser Tanbek, Çiğdem Tekin, Suat Tekin

Ischemic stroke (IS) is a leading cause of death and permanent disability worldwide. Diabetes is a major risk factor for IS and independently increases mortality. This study investigated the neuroprotective effects of Myrtenal (Myrt) in a rat model of IS under both diabetic and non-diabetic conditions. Sprague Dawley rats received Myrt (40 mg/kg, intraperitoneally) for 28 days before undergoing 60-minute middle cerebral artery occlusion followed by 24 h of reperfusion. Neurological outcomes were assessed using behavioral tests, infarct volume was measured by TTC staining, and biochemical analyses evaluated oxidative stress (MDA, SOD, CAT, GSH-Px) and inflammatory markers (NLRP3, TNF-α, IL-6, IL-1β). Western blotting was performed to examine BDNF/TrkB, p-PI3K/p-Akt signaling, and apoptosis-related proteins (Caspase-3, Bcl-2, Bax). IS impaired neurological function and increased infarct size, apoptosis, inflammation, and lipid peroxidation, while reducing antioxidant enzymes and BDNF/TrkB and p-PI3K/p-Akt levels (p < 0.05). These pathological changes were more severe in diabetic rats. Pretreatment with Myrt significantly ameliorated these effects in both diabetic and non-diabetic groups (p < 0.05). These findings suggest that Myrt exerts neuroprotective effects against IS by suppressing inflammation, oxidative stress, and apoptosis, possibly through modulation of BDNF/TrkB and p-PI3K/p-Akt pathways. These findings indicate that Myrt may possess neuroprotective potential in IS under both hyperglycemic and normoglycemic conditions.

缺血性中风(IS)是世界范围内导致死亡和永久性残疾的主要原因。糖尿病是is的主要危险因素,并独立增加死亡率。本研究探讨了桃金娘醛(Myrt)在糖尿病和非糖尿病大鼠IS模型中的神经保护作用。Sprague Dawley大鼠接受Myrt (40 mg/kg,腹腔注射)28天,然后进行大脑中动脉闭塞60分钟,再灌注24小时。通过行为测试评估神经系统预后,TTC染色测量梗死体积,生化分析评估氧化应激(MDA、SOD、CAT、GSH-Px)和炎症标志物(NLRP3、TNF-α、IL-6、IL-1β)。Western blotting检测BDNF/TrkB、p-PI3K/p-Akt信号和凋亡相关蛋白(Caspase-3、Bcl-2、Bax)。IS损害神经功能,增加梗死面积、细胞凋亡、炎症和脂质过氧化,同时降低抗氧化酶和BDNF/TrkB和p- pi3k /p- akt水平(p
{"title":"Myrtenal Ameliorates Ischemic Brain Injury Diabetic and Non-Diabetic Rats","authors":"Engin Korkmaz,&nbsp;Asiye Beytur,&nbsp;Yavuz Erden,&nbsp;Kevser Tanbek,&nbsp;Çiğdem Tekin,&nbsp;Suat Tekin","doi":"10.1007/s11064-025-04629-y","DOIUrl":"10.1007/s11064-025-04629-y","url":null,"abstract":"<div><p>Ischemic stroke (IS) is a leading cause of death and permanent disability worldwide. Diabetes is a major risk factor for IS and independently increases mortality. This study investigated the neuroprotective effects of Myrtenal (Myrt) in a rat model of IS under both diabetic and non-diabetic conditions. <i>Sprague Dawley</i> rats received Myrt (40 mg/kg, intraperitoneally) for 28 days before undergoing 60-minute middle cerebral artery occlusion followed by 24 h of reperfusion. Neurological outcomes were assessed using behavioral tests, infarct volume was measured by TTC staining, and biochemical analyses evaluated oxidative stress (MDA, SOD, CAT, GSH-Px) and inflammatory markers (NLRP3, TNF-α, IL-6, IL-1β). Western blotting was performed to examine BDNF/TrkB, p-PI3K/p-Akt signaling, and apoptosis-related proteins (Caspase-3, Bcl-2, Bax). IS impaired neurological function and increased infarct size, apoptosis, inflammation, and lipid peroxidation, while reducing antioxidant enzymes and BDNF/TrkB and p-PI3K/p-Akt levels (<i>p</i> &lt; 0.05). These pathological changes were more severe in diabetic rats. Pretreatment with Myrt significantly ameliorated these effects in both diabetic and non-diabetic groups (<i>p</i> &lt; 0.05). These findings suggest that Myrt exerts neuroprotective effects against IS by suppressing inflammation, oxidative stress, and apoptosis, possibly through modulation of BDNF/TrkB and p-PI3K/p-Akt pathways. These findings indicate that Myrt may possess neuroprotective potential in IS under both hyperglycemic and normoglycemic conditions.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"51 1","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145766772","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
NEXN-AS1 Predicts the Occurrence of Post-Stroke Cognitive Impairment and Alleviates Inflammation and Oxidative Stress by Targeting the miR-92a-3p/NRF1 Axis NEXN-AS1通过靶向miR-92a-3p/NRF1轴预测脑卒中后认知障碍的发生并缓解炎症和氧化应激:NEXN-AS1减轻PSCI的炎症和氧化应激
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-16 DOI: 10.1007/s11064-025-04626-1
Kai Wang, Lu Yang, Chuanzhou Zhang, Xiaomin Xing, Zhiguo Su, Ping Gao, Bing Han

Post-stroke cognitive impairment (PSCI) is a prevalent cerebrovascular condition resulting from ischemic stroke. This study aimed to determine the expression levels of NEXN-AS1 in PSCI, evaluate its clinical significance, and further uncover the molecular mechanisms through which it contributes to the initiation and progression of PSCI. The quantification of NEXN-AS1, miR-92a-3p, and NRF1 expression was performed using qRT-PCR. The diagnostic utility of serum NEXN-AS1 was assessed through ROC analysis. Risk factors associated with cognitive impairment following stroke were identified using both univariate and multivariate logistic regression. A cellular model of cognitive dysfunction was established via oxygen-glucose deprivation/reperfusion (OGD/R). The PSCI animal model was established through the Middle cerebral artery occlusion (MCAO) surgery. Inflammatory status was determined by measuring cytokine levels, including IL-6, IL-1β, and IL-10, while oxidative stress was evaluated by quantifying ROS, MDA, and CAT. In stroke patients, NEXN-AS1 expression was notably downregulated and further decreased in cases with PSCI. It served as a reliable biomarker for distinguishing stroke patients from healthy individuals and PSCI from post-stroke cognitive normality (PSCN) groups. Upregulation of NEXN-AS1 in BV2 cells following OGD/R stimulation led to increased proliferation, decreased inflammatory response, and reduced oxidative stress. Moreover, miR-92a-3p expression reversed the protective effects of NEXN-AS1 under OGD/R conditions. Overexpression of NEXN-AS1 alleviated cognitive dysfunction, inflammatory response and oxidative stress in PSCI rats, while overexpression of miR-92a-3p counteracted the protective effect of NEXN-AS1 on PSCI rats. Further analysis identified NRF1 as a downstream target of miR-92a-3p. NEXN-AS1 exerts protective effect against ischemic brain injury in both in vitro and in vivo models by regulating miR-92a-3p. Therefore, NEXN-AS1 may predict the occurrence of PSCI, and NEXN-AS1 may contribute to PSCI pathogenesis via regulation of the miR-92a-3p/NRF1 axis.

脑卒中后认知障碍(PSCI)是缺血性脑卒中后常见的脑血管疾病。本研究旨在确定NEXN-AS1在PSCI中的表达水平,评估其临床意义,并进一步揭示其参与PSCI发生和进展的分子机制。采用qRT-PCR定量测定NEXN-AS1、miR-92a-3p和NRF1的表达。通过ROC分析评估血清NEXN-AS1的诊断价值。使用单变量和多变量逻辑回归确定与脑卒中后认知障碍相关的危险因素。通过氧糖剥夺/再灌注(OGD/R)建立认知功能障碍的细胞模型。通过大脑中动脉闭塞术(MCAO)建立PSCI动物模型。通过测量细胞因子水平(包括IL-6、IL-1β和IL-10)来确定炎症状态,通过量化ROS、MDA和CAT来评估氧化应激。在脑卒中患者中,NEXN-AS1的表达明显下调,在PSCI患者中进一步降低。它是区分脑卒中患者与健康个体以及PSCI与脑卒中后认知正常(PSCN)组的可靠生物标志物。OGD/R刺激后BV2细胞中NEXN-AS1的上调导致增殖增加,炎症反应降低,氧化应激降低。此外,在OGD/R条件下,miR-92a-3p的表达逆转了NEXN-AS1的保护作用。过表达NEXN-AS1可减轻PSCI大鼠的认知功能障碍、炎症反应和氧化应激,而过表达miR-92a-3p可抵消NEXN-AS1对PSCI大鼠的保护作用。进一步分析发现NRF1是miR-92a-3p的下游靶点。NEXN-AS1通过调节miR-92a-3p在体外和体内模型中对缺血性脑损伤均有保护作用。因此,NEXN-AS1可能预测PSCI的发生,并且NEXN-AS1可能通过调控miR-92a-3p/NRF1轴参与PSCI的发病。
{"title":"NEXN-AS1 Predicts the Occurrence of Post-Stroke Cognitive Impairment and Alleviates Inflammation and Oxidative Stress by Targeting the miR-92a-3p/NRF1 Axis","authors":"Kai Wang,&nbsp;Lu Yang,&nbsp;Chuanzhou Zhang,&nbsp;Xiaomin Xing,&nbsp;Zhiguo Su,&nbsp;Ping Gao,&nbsp;Bing Han","doi":"10.1007/s11064-025-04626-1","DOIUrl":"10.1007/s11064-025-04626-1","url":null,"abstract":"<div><p>Post-stroke cognitive impairment (PSCI) is a prevalent cerebrovascular condition resulting from ischemic stroke. This study aimed to determine the expression levels of NEXN-AS1 in PSCI, evaluate its clinical significance, and further uncover the molecular mechanisms through which it contributes to the initiation and progression of PSCI. The quantification of NEXN-AS1, miR-92a-3p, and NRF1 expression was performed using qRT-PCR. The diagnostic utility of serum NEXN-AS1 was assessed through ROC analysis. Risk factors associated with cognitive impairment following stroke were identified using both univariate and multivariate logistic regression. A cellular model of cognitive dysfunction was established via oxygen-glucose deprivation/reperfusion (OGD/R). The PSCI animal model was established through the Middle cerebral artery occlusion (MCAO) surgery. Inflammatory status was determined by measuring cytokine levels, including IL-6, IL-1β, and IL-10, while oxidative stress was evaluated by quantifying ROS, MDA, and CAT. In stroke patients, NEXN-AS1 expression was notably downregulated and further decreased in cases with PSCI. It served as a reliable biomarker for distinguishing stroke patients from healthy individuals and PSCI from post-stroke cognitive normality (PSCN) groups. Upregulation of NEXN-AS1 in BV2 cells following OGD/R stimulation led to increased proliferation, decreased inflammatory response, and reduced oxidative stress. Moreover, miR-92a-3p expression reversed the protective effects of NEXN-AS1 under OGD/R conditions. Overexpression of NEXN-AS1 alleviated cognitive dysfunction, inflammatory response and oxidative stress in PSCI rats, while overexpression of miR-92a-3p counteracted the protective effect of NEXN-AS1 on PSCI rats. Further analysis identified NRF1 as a downstream target of miR-92a-3p. NEXN-AS1 exerts protective effect against ischemic brain injury in both in vitro and in vivo models by regulating miR-92a-3p. Therefore, NEXN-AS1 may predict the occurrence of PSCI, and NEXN-AS1 may contribute to PSCI pathogenesis via regulation of the miR-92a-3p/NRF1 axis.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"51 1","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145761913","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
PRMT5 Aggravates Parkinson’s Disease Progression Through the Inhibition of Neuronal Autophagy Through DKK1 PRMT5通过DKK1抑制神经元自噬加速帕金森病进展
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-11 DOI: 10.1007/s11064-025-04625-2
Houjun Zhou, Hao Fan, Hui Bian, Yanghong Zou, Ailan Pang, Xin Geng

Parkinson’s disease (PD) is the second most common neurodegenerative disease worldwide and severely affects the physical and mental health of patients. The protein arginine methyltransferase 5 (PRMT5) has been shown to be associated with neuronal degeneration in PD, but its specific mechanism of mediating PD remains unclear. The purpose of this study was to investigate the role of PRMT5 in PD and its potential mechanism. PD models in rats and MN9D cells were induced by 6-hydroxydopamine (6-OHDA). Key genes and proteins were identified through real-time quantitative polymerase chain reaction (RT‒qPCR), Western blotting, and immunofluorescence staining; apoptosis levels were measured using flow cytometry; autophagosome formation was observed via monodansylcadaverine (MDC) staining; and neuronal damage in PD rats was evaluated using hematoxylin‒eosin (H&E) and Nissl staining. In this study, we found that PRMT5 levels were elevated in the peripheral blood of PD patients and in 6-OHDA-induced rat brain tissue and MN9D cells and that the expression of PRMT5 was positively correlated with the level of α-Syn. After PRMT5 was knocked down, α-Syn levels in PD rats decreased, neuronal damage was inhibited, and motor disorders improved. In addition, knockdown of PRMT5 promoted 6-OHDA-induced MN9D cell proliferation, inhibited apoptosis, and upregulated autophagy. Mechanistically, PRMT5 inhibits the activation of the Wnt/β-catenin signaling pathway through H3R8me2s modification to stabilize the expression of DKK1, thus inhibiting neuronal autophagy and promoting the development of PD. Our study suggests that PRMT5 may be a potential intervention target for improving PD progression.

帕金森病(PD)是全球第二大常见的神经退行性疾病,严重影响患者的身心健康。蛋白精氨酸甲基转移酶5 (PRMT5)已被证明与PD的神经元变性有关,但其介导PD的具体机制尚不清楚。本研究旨在探讨PRMT5在帕金森病中的作用及其可能的机制。6-羟基多巴胺(6-OHDA)诱导大鼠PD模型和MN9D细胞。通过实时定量聚合酶链反应(RT-qPCR)、Western blotting和免疫荧光染色鉴定关键基因和蛋白;流式细胞术检测细胞凋亡水平;MDC染色观察自噬体形成;采用苏木精-伊红(H&E)和尼氏染色评价PD大鼠神经元损伤。本研究发现PD患者外周血、6- ohda诱导的大鼠脑组织及MN9D细胞中PRMT5水平升高,且PRMT5的表达与α-Syn水平呈正相关。敲除PRMT5后,PD大鼠α-Syn水平降低,神经元损伤受到抑制,运动障碍得到改善。此外,PRMT5敲低可促进6- ohda诱导的MN9D细胞增殖,抑制凋亡,上调自噬。机制上,PRMT5通过修饰H3R8me2s抑制Wnt/β-catenin信号通路的激活,稳定DKK1的表达,从而抑制神经元自噬,促进PD的发展。我们的研究表明,PRMT5可能是改善PD进展的潜在干预目标。
{"title":"PRMT5 Aggravates Parkinson’s Disease Progression Through the Inhibition of Neuronal Autophagy Through DKK1","authors":"Houjun Zhou,&nbsp;Hao Fan,&nbsp;Hui Bian,&nbsp;Yanghong Zou,&nbsp;Ailan Pang,&nbsp;Xin Geng","doi":"10.1007/s11064-025-04625-2","DOIUrl":"10.1007/s11064-025-04625-2","url":null,"abstract":"<div><p>Parkinson’s disease (PD) is the second most common neurodegenerative disease worldwide and severely affects the physical and mental health of patients. The protein arginine methyltransferase 5 (PRMT5) has been shown to be associated with neuronal degeneration in PD, but its specific mechanism of mediating PD remains unclear. The purpose of this study was to investigate the role of PRMT5 in PD and its potential mechanism. PD models in rats and MN9D cells were induced by 6-hydroxydopamine (6-OHDA). Key genes and proteins were identified through real-time quantitative polymerase chain reaction (RT‒qPCR), Western blotting, and immunofluorescence staining; apoptosis levels were measured using flow cytometry; autophagosome formation was observed via monodansylcadaverine (MDC) staining; and neuronal damage in PD rats was evaluated using hematoxylin‒eosin (H&amp;E) and Nissl staining. In this study, we found that PRMT5 levels were elevated in the peripheral blood of PD patients and in 6-OHDA-induced rat brain tissue and MN9D cells and that the expression of PRMT5 was positively correlated with the level of α-Syn. After PRMT5 was knocked down, α-Syn levels in PD rats decreased, neuronal damage was inhibited, and motor disorders improved. In addition, knockdown of PRMT5 promoted 6-OHDA-induced MN9D cell proliferation, inhibited apoptosis, and upregulated autophagy. Mechanistically, PRMT5 inhibits the activation of the Wnt/β-catenin signaling pathway through H3R8me2s modification to stabilize the expression of DKK1, thus inhibiting neuronal autophagy and promoting the development of PD. Our study suggests that PRMT5 may be a potential intervention target for improving PD progression.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"51 1","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145720382","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 Insular Cortex and Prefrontal Cortex in the Pathogenesis of Fibromyalgia: Biochemical and Electrophysiological Rodent Study 岛叶皮质和前额叶皮质在纤维肌痛发病机制中的作用:啮齿动物的生化和电生理研究。
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-11 DOI: 10.1007/s11064-025-04617-2
Ahmed F. Abouelnaga, Abdelaziz M. Hussein, Marwa Abass, Mena Z. Shafiek, Hala F. Zaki, Ahmed F. Mohamed, Weam W. Ibrahim

Neuroimaging implicates the medial prefrontal cortex (mPFC) and insula in fibromyalgia (FM), but synaptic and network mechanisms are unclear. To verify mPFC hypersynapticity in a reserpine-induced FM model and test whether anterior insular deep brain stimulation (DBS) restores mPFC oscillations and pain behavior. Thirty-six male Sprague–Dawley rats received reserpine (1 mg·kg⁻¹ i.p., days 1–3) or vehicle. For Objective 1, mPFC tissue was analyzed by ELISA for glutamate, γ-aminobutyric acid (GABA), C-FOS, nerve growth factor (NGF), synaptophysin, and postsynaptic density protein-95 (PSD-95). For Objective 2, rats were assigned to control-sham, control-DBS, FM-sham, or FM-DBS. Monopolar DBS (130 Hz, 60 µs, 100 µA; 15 min·day⁻¹ for 3 days) targeted the anterior insula; local field potentials were recorded from mPFC, and thermal nociception was assessed by tail immersion and hot-plate tests. Reserpine increased glutamate, C-FOS, NGF, synaptophysin, and PSD-95 and reduced GABA (all p < 0.001), confirming hypersynapticity. Insular DBS increased delta-band normalized ratios (NR) in FM and controls (p < 0.0001), normalized FM-associated theta reductions, decreased alpha/beta NR in controls, and suppressed elevated gamma NR in FM (p < 0.0001). DBS increased withdrawal latencies in FM, indicating improved pain thresholds (p < 0.001). Reserpine induces biochemical hypersynapticity in mPFC, and brief anterior insular DBS rebalances mPFC oscillations and alleviates hyperalgesia. Insular DBS may correct cortical network dysfunction in FM.

Graphical Abstract

Injection of Reserpine into rats decrease the pain threshold by hot plate and tail immersion tests indicating development of fibromyalgia (FM). On the other hand, deep brain stimulation (DBS) for the anterior insular cortex (dorsal anterior insular cortex, AID) reset the fast rhythms of LFPs into slow LFPs in the cingulate cortex (Cg1) region of the medial prefrontal cortex (mPFC) as well as increase the pain threshold

神经影像学提示纤维肌痛(FM)与内侧前额叶皮质(mPFC)和脑岛有关,但突触和网络机制尚不清楚。在利血平诱导的FM模型中验证mPFC高突触性,并测试前岛脑深部刺激(DBS)是否能恢复mPFC振荡和疼痛行为。36只雄性Sprague-Dawley大鼠接受利血平(1 mg·kg, 1-3天)或载药。目的1,采用ELISA法分析mPFC组织中谷氨酸、γ-氨基丁酸(GABA)、C-FOS、神经生长因子(NGF)、突触素和突触后密度蛋白-95 (PSD-95)的含量。对于目标2,大鼠被分配到对照-sham,对照- dbs, FM-sham或FM-DBS。单极DBS (130 Hz, 60µs, 100µA, 15分钟·天⁻¹,持续3天)针对脑岛前部;从mPFC记录局部场电位,并通过尾部浸泡和热板试验评估热伤害感受。利血平增加谷氨酸、C-FOS、NGF、突触素和PSD-95,降低GABA(均p
{"title":"The Role of Insular Cortex and Prefrontal Cortex in the Pathogenesis of Fibromyalgia: Biochemical and Electrophysiological Rodent Study","authors":"Ahmed F. Abouelnaga,&nbsp;Abdelaziz M. Hussein,&nbsp;Marwa Abass,&nbsp;Mena Z. Shafiek,&nbsp;Hala F. Zaki,&nbsp;Ahmed F. Mohamed,&nbsp;Weam W. Ibrahim","doi":"10.1007/s11064-025-04617-2","DOIUrl":"10.1007/s11064-025-04617-2","url":null,"abstract":"<div><p>Neuroimaging implicates the medial prefrontal cortex (mPFC) and insula in fibromyalgia (FM), but synaptic and network mechanisms are unclear. To verify mPFC hypersynapticity in a reserpine-induced FM model and test whether anterior insular deep brain stimulation (DBS) restores mPFC oscillations and pain behavior. Thirty-six male Sprague–Dawley rats received reserpine (1 mg·kg⁻¹ i.p., days 1–3) or vehicle. For Objective 1, mPFC tissue was analyzed by ELISA for glutamate, γ-aminobutyric acid (GABA), C-FOS, nerve growth factor (NGF), synaptophysin, and postsynaptic density protein-95 (PSD-95). For Objective 2, rats were assigned to control-sham, control-DBS, FM-sham, or FM-DBS. Monopolar DBS (130 Hz, 60 µs, 100 µA; 15 min·day⁻¹ for 3 days) targeted the anterior insula; local field potentials were recorded from mPFC, and thermal nociception was assessed by tail immersion and hot-plate tests. Reserpine increased glutamate, C-FOS, NGF, synaptophysin, and PSD-95 and reduced GABA (all <i>p</i> &lt; 0.001), confirming hypersynapticity. Insular DBS increased delta-band normalized ratios (NR) in FM and controls (<i>p</i> &lt; 0.0001), normalized FM-associated theta reductions, decreased alpha/beta NR in controls, and suppressed elevated gamma NR in FM (<i>p</i> &lt; 0.0001). DBS increased withdrawal latencies in FM, indicating improved pain thresholds (<i>p</i> &lt; 0.001). Reserpine induces biochemical hypersynapticity in mPFC, and brief anterior insular DBS rebalances mPFC oscillations and alleviates hyperalgesia. Insular DBS may correct cortical network dysfunction in FM.</p><h3>Graphical Abstract</h3><div><figure><div><div><picture><source><img></source></picture></div><div><p>Injection of Reserpine into rats decrease the pain threshold by hot plate and tail immersion tests indicating development of fibromyalgia (FM). On the other hand, deep brain stimulation (DBS) for the anterior insular cortex (dorsal anterior insular cortex, AID) reset the fast rhythms of LFPs into slow LFPs in the cingulate cortex (Cg1) region of the medial prefrontal cortex (mPFC) as well as increase the pain threshold</p></div></div></figure></div></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"51 1","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://link.springer.com/content/pdf/10.1007/s11064-025-04617-2.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145720296","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
Saracatinib Promotes Hippocampal Myelin Regeneration and Oligodendrocyte Precursor Cell Maturation by Inhibiting the NOTCH1 Signaling Pathway in Epileptic Mice Saracatinib通过抑制癫痫小鼠NOTCH1信号通路促进海马髓鞘再生和少突胶质前体细胞成熟。
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-09 DOI: 10.1007/s11064-025-04621-6
Xinming Luo, Wenyue Wu, Baiqi Yu, Sisi Chen, Jing Zhao, Jun Min, Xunhu Gu

Epilepsy is a common neurological disorder often accompanied by hippocampal myelin damage and impaired differentiation of oligodendrocyte precursor cells (OPCs). This study aimed to investigate the regulatory effects of Saracatinib on myelin regeneration and OPC maturation in a mouse model of epilepsy, as well as its underlying mechanisms, to provide new strategies for the treatment of epilepsy-related myelin damage. Changes in hippocampal myelin structure were observed using Fast Blue staining and transmission electron microscopy. Immunofluorescence was used to detect the number of oligodendrocyte precursor cell (OPCs, PDGFRα) and mature oligodendrocytes (ODCs, MBP+). Cell culture experiments verified the effects of Saracatinib on OPC differentiation, and SwissTargetPrediction and GSEA were used to predict its targets. Western blot and immunofluorescence further validated the role of the NOTCH1 signaling pathway in Saracatinib-mediated OPC differentiation. Saracatinib Reduced the Racine Score, Prolonged Seizure Latency, and Decreased Seizure Duration in an Epileptic Mouse Model. Epileptic model mice exhibited significant hippocampal myelin damage, characterized by thinning of myelin sheaths, reduced myelin quantity, and increased axonal exposure. Saracatinib treatment significantly improved myelin structure, restored myelin thickness and continuity, and alleviated axonal atrophy. Immunofluorescence showed that Saracatinib increased MBP expression and decreased PDGFRα expression, promoting the differentiation of OPCs into ODCs. Bioinformatics analysis and experimental validation demonstrated that Saracatinib promoted OPC maturation by inhibiting the NOTCH1 signaling pathway, and this effect could be reversed by the NOTCH1 agonist JAG1. Saracatinib significantly promotes hippocampal myelin regeneration and OPC maturation in epileptic mice by inhibiting the NOTCH1 signaling pathway, providing a potential molecular target and therapeutic strategy for the treatment of epilepsy-related myelin damage.

癫痫是一种常见的神经系统疾病,常伴有海马髓鞘损伤和少突胶质前体细胞(OPCs)分化受损。本研究旨在探讨Saracatinib对癫痫小鼠模型髓磷脂再生和OPC成熟的调控作用及其潜在机制,为癫痫相关髓磷脂损伤的治疗提供新的策略。用Fast Blue染色和透射电镜观察海马髓磷脂结构的变化。采用免疫荧光法检测少突胶质细胞前体细胞(OPCs, PDGFRα)和成熟少突胶质细胞(ODCs, MBP+)的数量。细胞培养实验验证了Saracatinib对OPC分化的影响,利用SwissTargetPrediction和GSEA预测其靶点。Western blot和免疫荧光进一步验证了NOTCH1信号通路在saracatinib介导的OPC分化中的作用。在癫痫小鼠模型中,萨拉卡替尼降低了拉辛评分,延长了癫痫发作潜伏期,缩短了癫痫发作持续时间。癫痫模型小鼠表现出明显的海马髓鞘损伤,其特征是髓鞘变薄,髓鞘数量减少,轴突暴露增加。Saracatinib治疗显著改善髓鞘结构,恢复髓鞘厚度和连续性,减轻轴突萎缩。免疫荧光显示萨拉卡替尼增加MBP表达,降低PDGFRα表达,促进OPCs向ODCs分化。生物信息学分析和实验验证表明,Saracatinib通过抑制NOTCH1信号通路促进OPC成熟,NOTCH1激动剂JAG1可逆转这一作用。Saracatinib通过抑制NOTCH1信号通路显著促进癫痫小鼠海马髓鞘再生和OPC成熟,为治疗癫痫相关髓鞘损伤提供了潜在的分子靶点和治疗策略。
{"title":"Saracatinib Promotes Hippocampal Myelin Regeneration and Oligodendrocyte Precursor Cell Maturation by Inhibiting the NOTCH1 Signaling Pathway in Epileptic Mice","authors":"Xinming Luo,&nbsp;Wenyue Wu,&nbsp;Baiqi Yu,&nbsp;Sisi Chen,&nbsp;Jing Zhao,&nbsp;Jun Min,&nbsp;Xunhu Gu","doi":"10.1007/s11064-025-04621-6","DOIUrl":"10.1007/s11064-025-04621-6","url":null,"abstract":"<div>\u0000 \u0000 <p>Epilepsy is a common neurological disorder often accompanied by hippocampal myelin damage and impaired differentiation of oligodendrocyte precursor cells (OPCs). This study aimed to investigate the regulatory effects of Saracatinib on myelin regeneration and OPC maturation in a mouse model of epilepsy, as well as its underlying mechanisms, to provide new strategies for the treatment of epilepsy-related myelin damage. Changes in hippocampal myelin structure were observed using Fast Blue staining and transmission electron microscopy. Immunofluorescence was used to detect the number of oligodendrocyte precursor cell (OPCs, PDGFRα) and mature oligodendrocytes (ODCs, MBP+). Cell culture experiments verified the effects of Saracatinib on OPC differentiation, and SwissTargetPrediction and GSEA were used to predict its targets. Western blot and immunofluorescence further validated the role of the NOTCH1 signaling pathway in Saracatinib-mediated OPC differentiation. Saracatinib Reduced the Racine Score, Prolonged Seizure Latency, and Decreased Seizure Duration in an Epileptic Mouse Model. Epileptic model mice exhibited significant hippocampal myelin damage, characterized by thinning of myelin sheaths, reduced myelin quantity, and increased axonal exposure. Saracatinib treatment significantly improved myelin structure, restored myelin thickness and continuity, and alleviated axonal atrophy. Immunofluorescence showed that Saracatinib increased MBP expression and decreased PDGFRα expression, promoting the differentiation of OPCs into ODCs. Bioinformatics analysis and experimental validation demonstrated that Saracatinib promoted OPC maturation by inhibiting the NOTCH1 signaling pathway, and this effect could be reversed by the NOTCH1 agonist JAG1. Saracatinib significantly promotes hippocampal myelin regeneration and OPC maturation in epileptic mice by inhibiting the NOTCH1 signaling pathway, providing a potential molecular target and therapeutic strategy for the treatment of epilepsy-related myelin damage.</p>\u0000 </div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"51 1","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145706749","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
Genistein Exerts Neuroprotective Effects in an Ouabain-Induced Model of Bipolar Disorder: Behavioral and Molecular Insights 染料木素在瓦巴因诱导的双相障碍模型中发挥神经保护作用:行为和分子的见解。
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-08 DOI: 10.1007/s11064-025-04597-3
Mariam T. Arafat, Heba R. Ghaiad, Eman M. Elbaz

Bipolar disorder (BD) is a chronic and prevalent psychiatric disease that has been considered a leading cause of disability among psychiatric conditions. Taking into account that there is yet no satisfactory disease-modifying treatment, we investigated the effect of genistein on ouabain-induced BD in male C57BL/6 mice. Animals were categorized into control, genistein control, ouabain model, lithium (Li)-treated, and genistein-treated groups. BD was induced by bilateral intracerebroventricular injection of 0.625 nmol ouabain. Genistein (10 mg/kg/day) was orally administered for 2 weeks following a single dose of ouabain. Open field test, sucrose preference test, and forced swim test were performed. Na⁺/K⁺-ATPase activity was evaluated through measuring the hippocampal levels of phosphorylated epidermal growth factor receptor, proto-oncogene tyrosine-protein kinase, extracellular signal-regulated kinase, and cAMP response element-binding protein (p-CREB) by western blot analysis. The levels of brain-derived neurotrophic factor (BDNF), serotonin, oxidative stress, and inflammatory markers were quantified by ELISA. The BCL-2-associated X protein (BAX) to B-cell Lymphoma/Leukemia (BCL2) ratio was assessed by qRT-PCR. Genistein reduced manic and anxious behaviors during the manic phase and showed an antidepressant effect during the depression phase, all while maintaining an effective metabolic balance on body weight. Additionally, genistein increased serotonin, p-CREB, and BDNF levels while decreasing inflammation and apoptosis produced by ouabain. Furthermore, genistein restored the normal architecture in both hippocampal and cortical H&E-stained sections. Taken together, genistein was able to activate the Na⁺/K⁺-ATPase signalosome via a multifaceted mode of action, exerting a neuroprotective effect in an animal model of BD, promoting genistein as a therapeutic candidate for BD.

Graphical Abstract

双相情感障碍(BD)是一种慢性和流行的精神疾病,被认为是精神疾病中致残的主要原因。考虑到目前还没有令人满意的疾病改善治疗方法,我们研究了染料木素对瓦阿卡因诱导的雄性C57BL/6小鼠BD的影响。将动物分为对照组、染料木素对照组、瓦巴因模型组、锂(Li)组和染料木素组。双侧脑室内注射0.625 nmol瓦巴因诱导BD。染料木素(10mg /kg/天)在单次给药瓦巴因后口服2周。进行大田试验、蔗糖偏好试验和强迫游泳试验。通过western blot检测海马磷酸化表皮生长因子受体、原癌基因酪氨酸蛋白激酶、细胞外信号调节激酶和cAMP反应元件结合蛋白(p-CREB)水平,评估Na + /K + - atp酶活性。采用酶联免疫吸附法(ELISA)定量测定脑源性神经营养因子(BDNF)、血清素、氧化应激和炎症标志物水平。采用qRT-PCR检测bcl -2相关X蛋白(BAX)与b细胞淋巴瘤/白血病(BCL2)的比值。染料木素在躁狂期减少躁狂和焦虑行为,在抑郁期表现出抗抑郁作用,同时保持有效的体重代谢平衡。此外,染料木素增加血清素、p-CREB和BDNF水平,同时减少瓦巴因引起的炎症和细胞凋亡。此外,染料木素恢复了海马和皮质h&e染色切片的正常结构。综上所述,染料木素能够通过多方面的作用模式激活Na + /K + - atp酶信号体,在BD动物模型中发挥神经保护作用,促进染料木素成为BD的治疗候选药物。
{"title":"Genistein Exerts Neuroprotective Effects in an Ouabain-Induced Model of Bipolar Disorder: Behavioral and Molecular Insights","authors":"Mariam T. Arafat,&nbsp;Heba R. Ghaiad,&nbsp;Eman M. Elbaz","doi":"10.1007/s11064-025-04597-3","DOIUrl":"10.1007/s11064-025-04597-3","url":null,"abstract":"<div><p>Bipolar disorder (BD) is a chronic and prevalent psychiatric disease that has been considered a leading cause of disability among psychiatric conditions. Taking into account that there is yet no satisfactory disease-modifying treatment, we investigated the effect of genistein on ouabain-induced BD in male C57BL/6 mice. Animals were categorized into control, genistein control, ouabain model, lithium (Li)-treated, and genistein-treated groups. BD was induced by bilateral intracerebroventricular injection of 0.625 nmol ouabain. Genistein (10 mg/kg/day) was orally administered for 2 weeks following a single dose of ouabain. Open field test, sucrose preference test, and forced swim test were performed. Na⁺/K⁺-ATPase activity was evaluated through measuring the hippocampal levels of phosphorylated epidermal growth factor receptor, proto-oncogene tyrosine-protein kinase, extracellular signal-regulated kinase, and cAMP response element-binding protein (p-CREB) by western blot analysis. The levels of brain-derived neurotrophic factor (BDNF), serotonin, oxidative stress, and inflammatory markers were quantified by ELISA. The BCL-2-associated X protein (BAX) to B-cell Lymphoma/Leukemia (BCL2) ratio was assessed by qRT-PCR. Genistein reduced manic and anxious behaviors during the manic phase and showed an antidepressant effect during the depression phase, all while maintaining an effective metabolic balance on body weight. Additionally, genistein increased serotonin, p-CREB, and BDNF levels while decreasing inflammation and apoptosis produced by ouabain. Furthermore, genistein restored the normal architecture in both hippocampal and cortical H&amp;E-stained sections. Taken together, genistein was able to activate the Na⁺/K⁺-ATPase signalosome via a multifaceted mode of action, exerting a neuroprotective effect in an animal model of BD, promoting genistein as a therapeutic candidate for BD.</p><h3>Graphical Abstract</h3><div><figure><div><div><picture><source><img></source></picture></div></div></figure></div></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"51 1","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12682918/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145699564","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
Targeting Liquid-Liquid Phase Separation and Autophagy in Alzheimer’s Disease: Insights into Molecular Mechanisms and Therapeutic Potential 针对阿尔茨海默病的液-液相分离和自噬:分子机制和治疗潜力的见解。
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-04 DOI: 10.1007/s11064-025-04623-4
Xiaopeng Li, Yan Liu, Hong Hu, Shenghong Li

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and the leading cause of dementia, marked by cognitive decline and memory loss. Its multifactorial etiology involves genetic, environmental, and cellular factors, with key pathological features including amyloid-beta (Aβ) plaques and tau tangles. Recent studies have highlighted the roles of liquid-liquid phase separation (LLPS) and autophagy in AD onset and progression. LLPS, an emerging biophysical phenomenon, facilitates protein aggregation and may contribute to early disease stages. Dysregulated autophagy results in the accumulation of toxic proteins, such as Aβ and tau, exacerbating neurodegeneration. This review explores the interplay between LLPS and autophagy in AD, a relationship often overlooked in the literature. It examines their biological mechanisms, synergistic effects on AD pathology, and potential therapeutic strategies. Additionally, we discuss the therapeutic potential of both natural and non-natural compounds in modulating LLPS and autophagy. While compounds like curcumin show promise, a comprehensive framework for their targeted use remains under development. This review provides theoretical support for the advancement of more precise AD therapies.

阿尔茨海默病(AD)是一种进行性神经退行性疾病,是痴呆症的主要原因,其特征是认知能力下降和记忆力丧失。其多因素病因涉及遗传、环境和细胞因素,主要病理特征包括淀粉样蛋白斑块和tau蛋白缠结。近年来的研究强调了液-液相分离(LLPS)和自噬在AD发生和发展中的作用。LLPS是一种新兴的生物物理现象,促进蛋白质聚集,可能有助于疾病的早期阶段。失调的自噬导致有毒蛋白的积累,如Aβ和tau,加剧神经变性。这篇综述探讨了AD中LLPS和自噬之间的相互作用,这一关系在文献中经常被忽视。它探讨了它们的生物学机制,对AD病理的协同作用,以及潜在的治疗策略。此外,我们讨论了天然和非天然化合物在调节LLPS和自噬方面的治疗潜力。虽然像姜黄素这样的化合物显示出希望,但针对其目标用途的综合框架仍在开发中。这一综述为更精确的阿尔茨海默病治疗提供了理论支持。
{"title":"Targeting Liquid-Liquid Phase Separation and Autophagy in Alzheimer’s Disease: Insights into Molecular Mechanisms and Therapeutic Potential","authors":"Xiaopeng Li,&nbsp;Yan Liu,&nbsp;Hong Hu,&nbsp;Shenghong Li","doi":"10.1007/s11064-025-04623-4","DOIUrl":"10.1007/s11064-025-04623-4","url":null,"abstract":"<div><p>Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and the leading cause of dementia, marked by cognitive decline and memory loss. Its multifactorial etiology involves genetic, environmental, and cellular factors, with key pathological features including amyloid-beta (Aβ) plaques and tau tangles. Recent studies have highlighted the roles of liquid-liquid phase separation (LLPS) and autophagy in AD onset and progression. LLPS, an emerging biophysical phenomenon, facilitates protein aggregation and may contribute to early disease stages. Dysregulated autophagy results in the accumulation of toxic proteins, such as Aβ and tau, exacerbating neurodegeneration. This review explores the interplay between LLPS and autophagy in AD, a relationship often overlooked in the literature. It examines their biological mechanisms, synergistic effects on AD pathology, and potential therapeutic strategies. Additionally, we discuss the therapeutic potential of both natural and non-natural compounds in modulating LLPS and autophagy. While compounds like curcumin show promise, a comprehensive framework for their targeted use remains under development. This review provides theoretical support for the advancement of more precise AD therapies.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"51 1","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145666657","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
Molecular Mechanisms Underlying Chronic High-Dose Ketamine-Induced Apoptosis in the Hippocampus: A Narrative Review 慢性大剂量氯胺酮诱导海马细胞凋亡的分子机制:一项叙述性综述。
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-01 DOI: 10.1007/s11064-025-04619-0
Ali Ahmad Azadbakht, Habibeh Mashayekhi-sardoo, Yousef Baghcheghi

Chronic high-dose ketamine, widely recognized for its rapid antidepressant effects, poses significant risks to brain health, particularly in the hippocampus, a region critical for learning, memory, and emotional regulation. This narrative review aims to elucidate the molecular mechanisms underlying ketamine-induced apoptosis in hippocampal neurons, providing a comprehensive synthesis of current research findings. We examine how chronic exposure to high doses of ketamine disrupts glutamatergic signaling through NMDA receptor antagonism, leading to an imbalance in excitatory neurotransmission that triggers apoptotic pathways. Additionally, we explore the roles of neuroinflammation and oxidative stress in exacerbating neuronal vulnerability, highlighting the interplay between these mechanisms. The review discusses how chronic ketamine use activates glial cells, resulting in the release of pro-inflammatory cytokines and increased oxidative damage, further promoting neuronal cell death. Furthermore, we consider the implications of altered neurotrophic factor signaling and mitochondrial dysfunction in the context of ketamine’s neurotoxic effects. By integrating these molecular pathways, we provide insights into the critical factors contributing to ketamine-induced apoptosis. Finally, we highlight the need for further research to clarify the dose-response relationship, individual variability in treatment outcomes, and potential neuroprotective strategies. Ultimately, this review emphasizes the importance of balancing the therapeutic benefits of ketamine with its associated risks, advocating for a nuanced understanding of its long-term effects on brain health to inform clinical practices and optimize patient care.

Graphical Abstract

This graphical abstract summarizes the key molecular pathways underlying chronic high-dose ketamine-induced apoptosis in hippocampal neurons, as explored in this narrative review. The illustration depicts how ketamine's primary mechanism as an NMDA receptor antagonist initiates a cascade of detrimental events: (1) disruption of glutamatergic signaling homeostasis; (2) activation of microglia and astrocytes, leading to neuroinflammation through pro-inflammatory cytokine release; (3) induction of oxidative stress via reactive oxygen species (ROS) generation; (4) impairment of mitochondrial function, promoting cytochrome c release; and (5) dysregulation of neurotrophic factor signaling (e.g., BDNF). These interconnected pathways ultimately converge to activate caspase-dependent apoptotic cascades, resulting in hippocampal neuronal death. This integrative process highlights the critical balance required between ketamine's rapid antidepressant potential and its neurotoxic risks, emphasizing the need for further research into neuroprotective strategies.

慢性大剂量氯胺酮因其快速抗抑郁作用而被广泛认可,对大脑健康,特别是对学习、记忆和情绪调节至关重要的海马区构成重大风险。本文旨在阐述氯胺酮诱导海马神经元凋亡的分子机制,并对目前的研究成果进行综合评述。我们研究了长期暴露于高剂量氯胺酮如何通过NMDA受体拮抗破坏谷氨酸能信号,导致兴奋性神经传递失衡,从而引发凋亡通路。此外,我们探讨了神经炎症和氧化应激在加剧神经元易感性中的作用,强调了这些机制之间的相互作用。这篇综述讨论了长期使用氯胺酮如何激活神经胶质细胞,导致促炎细胞因子的释放和氧化损伤的增加,进一步促进神经元细胞死亡。此外,我们考虑在氯胺酮的神经毒性作用的背景下改变的神经营养因子信号和线粒体功能障碍的影响。通过整合这些分子途径,我们提供了对氯胺酮诱导细胞凋亡的关键因素的见解。最后,我们强调需要进一步研究以阐明剂量-反应关系、治疗结果的个体差异和潜在的神经保护策略。最后,本综述强调了平衡氯胺酮的治疗益处及其相关风险的重要性,提倡对其对大脑健康的长期影响进行细致的了解,以告知临床实践和优化患者护理。这篇综述总结了慢性大剂量氯胺酮诱导海马神经元凋亡的关键分子通路。该图描述了氯胺酮作为NMDA受体拮抗剂的主要机制如何引发一系列有害事件:(1)破坏谷氨酸能信号稳态;(2)激活小胶质细胞和星形胶质细胞,通过释放促炎细胞因子导致神经炎症;(3)通过活性氧(ROS)的生成诱导氧化应激;(4)线粒体功能受损,促进细胞色素c释放;(5)神经营养因子信号(如BDNF)的失调。这些相互关联的通路最终汇聚激活caspase依赖性的凋亡级联反应,导致海马神经元死亡。这一综合过程强调了氯胺酮的快速抗抑郁潜力与其神经毒性风险之间的关键平衡,强调了进一步研究神经保护策略的必要性。
{"title":"Molecular Mechanisms Underlying Chronic High-Dose Ketamine-Induced Apoptosis in the Hippocampus: A Narrative Review","authors":"Ali Ahmad Azadbakht,&nbsp;Habibeh Mashayekhi-sardoo,&nbsp;Yousef Baghcheghi","doi":"10.1007/s11064-025-04619-0","DOIUrl":"10.1007/s11064-025-04619-0","url":null,"abstract":"<div><p>Chronic high-dose ketamine, widely recognized for its rapid antidepressant effects, poses significant risks to brain health, particularly in the hippocampus, a region critical for learning, memory, and emotional regulation. This narrative review aims to elucidate the molecular mechanisms underlying ketamine-induced apoptosis in hippocampal neurons, providing a comprehensive synthesis of current research findings. We examine how chronic exposure to high doses of ketamine disrupts glutamatergic signaling through NMDA receptor antagonism, leading to an imbalance in excitatory neurotransmission that triggers apoptotic pathways. Additionally, we explore the roles of neuroinflammation and oxidative stress in exacerbating neuronal vulnerability, highlighting the interplay between these mechanisms. The review discusses how chronic ketamine use activates glial cells, resulting in the release of pro-inflammatory cytokines and increased oxidative damage, further promoting neuronal cell death. Furthermore, we consider the implications of altered neurotrophic factor signaling and mitochondrial dysfunction in the context of ketamine’s neurotoxic effects. By integrating these molecular pathways, we provide insights into the critical factors contributing to ketamine-induced apoptosis. Finally, we highlight the need for further research to clarify the dose-response relationship, individual variability in treatment outcomes, and potential neuroprotective strategies. Ultimately, this review emphasizes the importance of balancing the therapeutic benefits of ketamine with its associated risks, advocating for a nuanced understanding of its long-term effects on brain health to inform clinical practices and optimize patient care.</p><h3>Graphical Abstract</h3><div><figure><div><div><picture><source><img></source></picture></div><div><p>This graphical abstract summarizes the key molecular pathways underlying chronic high-dose ketamine-induced apoptosis in hippocampal neurons, as explored in this narrative review. The illustration depicts how ketamine's primary mechanism as an NMDA receptor antagonist initiates a cascade of detrimental events: (1) disruption of glutamatergic signaling homeostasis; (2) activation of microglia and astrocytes, leading to neuroinflammation through pro-inflammatory cytokine release; (3) induction of oxidative stress via reactive oxygen species (ROS) generation; (4) impairment of mitochondrial function, promoting cytochrome c release; and (5) dysregulation of neurotrophic factor signaling (e.g., BDNF). These interconnected pathways ultimately converge to activate caspase-dependent apoptotic cascades, resulting in hippocampal neuronal death. This integrative process highlights the critical balance required between ketamine's rapid antidepressant potential and its neurotoxic risks, emphasizing the need for further research into neuroprotective strategies.</p></div></div></figure></div></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"51 1","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145647008","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
6-Shogaol Alleviates Post-Cardiopulmonary Resuscitation Brain Injury in Rats by Regulating the miRNA-26a-5p/DAPK1 6-Shogaol通过调节miRNA-26a-5p/DAPK1减轻大鼠心肺复苏后脑损伤
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-01 DOI: 10.1007/s11064-025-04610-9
Ouyang Rao, Jie Hu, Ning Zhu, Yun Li, Sha Xie, Junling Tao, Yehong Li, Ying Liu

This study aims to investigate the neuroprotective effect of 6-shogaol (6-SH) in rats after cardiopulmonary resuscitation (CPR), and to explore the molecular mechanism by which it regulates the miRNA-26a-5p/DAPK1 to inhibit excessive autophagy and calcium overload. A rat model of cerebral ischemia-reperfusion injury after cardiac arrest and CPR was established by asphyxia. Sham, CPR, and CPR + 6-SH groups were set up. DAPK1 overexpression and miRNA-26a-5p inhibition were also performed. Neurological function was evaluated using the Neurological Deficit Score (NDS). Hematoxylin-eosin staining was used to assess pathological damage in brain tissue. Immunofluorescence was used to observe the expression of autophagy markers. Quantitative real-time polymerase chain reaction (RT-qPCR) and Western blot (WB) were used to detect the expression of autophagy- and calcium overload-related genes and proteins. A dual-luciferase reporter assay was conducted to verify the targeting relationship between miRNA-26a-5p and DAPK1. Molecular docking was used to analyze the binding interaction between 6-SH and miRNA-26a-5p. The results show that 6-SH significantly reduces brain injury and improves neurological function after CPR. It decreases the expression of autophagy-related proteins (VPS34, Beclin1, LC3) and the calcium overload marker (NMDAR2B). Further mechanistic studies show that 6-SH inhibits DAPK1 expression and attenuates excessive autophagy and calcium overload. In addition, 6-SH binds to miRNA-26a-5p and upregulates its expression, which in turn suppresses DAPK1. When miRNA-26a-5p is inhibited, the effects of 6-SH on DAPK1, autophagy, and calcium overload are partially reversed. In conclusion, 6-SH attenuates brain injury after CPR by regulating the miRNA-26a-5p/DAPK1 to suppress excessive autophagy and calcium overload.

本研究旨在探讨6-shogaol (6-SH)对大鼠心肺复苏后的神经保护作用,并探讨其调控miRNA-26a-5p/DAPK1抑制过度自噬和钙超载的分子机制。采用窒息法建立大鼠心脏骤停及心肺复苏术后脑缺血再灌注损伤模型。设假手术组、CPR组和CPR + 6-SH组。还进行了DAPK1过表达和miRNA-26a-5p抑制。使用神经功能缺损评分(NDS)评估神经功能。采用苏木精-伊红染色评价脑组织病理损伤。采用免疫荧光法观察自噬标志物的表达。采用实时定量聚合酶链反应(RT-qPCR)和Western blot (WB)检测自噬和钙超载相关基因和蛋白的表达。双荧光素酶报告基因实验验证了miRNA-26a-5p和DAPK1之间的靶向关系。分子对接分析6-SH与miRNA-26a-5p的结合相互作用。结果表明,6-SH可显著减轻心肺复苏术后脑损伤,改善神经功能。它降低了自噬相关蛋白(VPS34、Beclin1、LC3)和钙超载标志物(NMDAR2B)的表达。进一步的机制研究表明,6-SH抑制DAPK1表达,减轻过度自噬和钙超载。此外,6-SH结合miRNA-26a-5p并上调其表达,进而抑制DAPK1。当miRNA-26a-5p被抑制时,6-SH对DAPK1、自噬和钙超载的影响部分逆转。综上所述,6-SH通过调节miRNA-26a-5p/DAPK1抑制过度自噬和钙超载来减轻心肺复苏术后脑损伤。
{"title":"6-Shogaol Alleviates Post-Cardiopulmonary Resuscitation Brain Injury in Rats by Regulating the miRNA-26a-5p/DAPK1","authors":"Ouyang Rao,&nbsp;Jie Hu,&nbsp;Ning Zhu,&nbsp;Yun Li,&nbsp;Sha Xie,&nbsp;Junling Tao,&nbsp;Yehong Li,&nbsp;Ying Liu","doi":"10.1007/s11064-025-04610-9","DOIUrl":"10.1007/s11064-025-04610-9","url":null,"abstract":"<div><p>This study aims to investigate the neuroprotective effect of 6-shogaol (6-SH) in rats after cardiopulmonary resuscitation (CPR), and to explore the molecular mechanism by which it regulates the miRNA-26a-5p/DAPK1 to inhibit excessive autophagy and calcium overload. A rat model of cerebral ischemia-reperfusion injury after cardiac arrest and CPR was established by asphyxia. Sham, CPR, and CPR + 6-SH groups were set up. DAPK1 overexpression and miRNA-26a-5p inhibition were also performed. Neurological function was evaluated using the Neurological Deficit Score (NDS). Hematoxylin-eosin staining was used to assess pathological damage in brain tissue. Immunofluorescence was used to observe the expression of autophagy markers. Quantitative real-time polymerase chain reaction (RT-qPCR) and Western blot (WB) were used to detect the expression of autophagy- and calcium overload-related genes and proteins. A dual-luciferase reporter assay was conducted to verify the targeting relationship between miRNA-26a-5p and DAPK1. Molecular docking was used to analyze the binding interaction between 6-SH and miRNA-26a-5p. The results show that 6-SH significantly reduces brain injury and improves neurological function after CPR. It decreases the expression of autophagy-related proteins (VPS34, Beclin1, LC3) and the calcium overload marker (NMDAR2B). Further mechanistic studies show that 6-SH inhibits DAPK1 expression and attenuates excessive autophagy and calcium overload. In addition, 6-SH binds to miRNA-26a-5p and upregulates its expression, which in turn suppresses DAPK1. When miRNA-26a-5p is inhibited, the effects of 6-SH on DAPK1, autophagy, and calcium overload are partially reversed. In conclusion, 6-SH attenuates brain injury after CPR by regulating the miRNA-26a-5p/DAPK1 to suppress excessive autophagy and calcium overload.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"51 1","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145646979","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
Ciwujianoside C Attenuates Cerebral Ischemia-Reperfusion Injury by Suppressing Ferroptosis via NNAT-Mediated Inhibition of NF-κB Signaling 刺五加皂苷C通过nnat介导的NF-κB信号抑制抑制铁凋亡,减轻脑缺血再灌注损伤
IF 3.8 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-29 DOI: 10.1007/s11064-025-04622-5
Weichuan Dai, Yingxian Chen, Wenhua Cai, Yuyan Zhu, Xieli Guo

Cerebral ischemia-reperfusion injury (CIRI) involves oxidative stress, inflammation, and regulated cell death, among which ferroptosis has emerged as a key contributor. However, therapeutic strategies targeting ferroptosis remain limited. This study investigated whether Ciwujianoside C (CC), a triterpenoid saponin from Acanthopanax senticosus, protects against CIRI by modulating ferroptosis via the NNAT/NF-κB pathway. In MCAO/R rats, CC reduced infarct size, improved neurological scores, and ameliorated oxidative stress and ferroptosis markers. In BV2 microglia and HT22 cells (a mouse hippocampal neuronal cell line) subjected to OGD/R, CC enhanced cell viability, decreased iron accumulation, and restored GPX4 and FTH1 expression while inhibiting NF-κB activation. Importantly, NNAT knockdown abolished these protective effects, demonstrating NNAT as a critical mediator. These findings reveal that CC protects against CIRI by suppressing ferroptosis through the NNAT/NF-κB axis, highlighting NNAT as a potential therapeutic target in CIRI.

脑缺血再灌注损伤(CIRI)涉及氧化应激、炎症和调节细胞死亡,其中铁凋亡是一个关键因素。然而,针对铁下垂的治疗策略仍然有限。本研究探讨刺五加三萜皂苷C (Ciwujianoside C, CC)是否通过NNAT/NF-κB通路调节铁凋亡,从而对CIRI具有保护作用。在MCAO/R大鼠中,CC减少了梗死面积,改善了神经学评分,改善了氧化应激和铁下垂标志物。在OGD/R的BV2小胶质细胞和HT22细胞(小鼠海马神经元细胞系)中,CC增强细胞活力,减少铁积累,恢复GPX4和FTH1的表达,同时抑制NF-κB的激活。重要的是,NNAT敲除消除了这些保护作用,表明NNAT是一个关键的介质。这些发现表明,CC通过NNAT/NF-κB轴抑制铁下垂来保护CIRI,突出了NNAT作为CIRI的潜在治疗靶点。
{"title":"Ciwujianoside C Attenuates Cerebral Ischemia-Reperfusion Injury by Suppressing Ferroptosis via NNAT-Mediated Inhibition of NF-κB Signaling","authors":"Weichuan Dai,&nbsp;Yingxian Chen,&nbsp;Wenhua Cai,&nbsp;Yuyan Zhu,&nbsp;Xieli Guo","doi":"10.1007/s11064-025-04622-5","DOIUrl":"10.1007/s11064-025-04622-5","url":null,"abstract":"<div><p>Cerebral ischemia-reperfusion injury (CIRI) involves oxidative stress, inflammation, and regulated cell death, among which ferroptosis has emerged as a key contributor. However, therapeutic strategies targeting ferroptosis remain limited. This study investigated whether Ciwujianoside C (CC), a triterpenoid saponin from Acanthopanax senticosus, protects against CIRI by modulating ferroptosis via the NNAT/NF-κB pathway. In MCAO/R rats, CC reduced infarct size, improved neurological scores, and ameliorated oxidative stress and ferroptosis markers. In BV2 microglia and HT22 cells (a mouse hippocampal neuronal cell line) subjected to OGD/R, CC enhanced cell viability, decreased iron accumulation, and restored GPX4 and FTH1 expression while inhibiting NF-κB activation. Importantly, NNAT knockdown abolished these protective effects, demonstrating NNAT as a critical mediator. These findings reveal that CC protects against CIRI by suppressing ferroptosis through the NNAT/NF-κB axis, highlighting NNAT as a potential therapeutic target in CIRI.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"51 1","pages":""},"PeriodicalIF":3.8,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145613103","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