首页 > 最新文献

Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology最新文献

英文 中文
Repositioning of Alogliptin to Mitigate Secondary Injury Induced by Repetitive TBI: Potential Role of its Antioxidant and Anti- Inflammatory Effects. 阿格列汀重新定位以减轻重复性脑损伤引起的继发性损伤:其抗氧化和抗炎作用的潜在作用。
Hossam A Raslan, Haidy E Michel, Esther T Menze, Amira A El-Gazar

Repetitive traumatic brain injury (RTBI) refers to brain injuries resulting from an external mechanical force causing cumulative and frequently severe neurological consequences. This study aimed to explore the neuroprotective effect of alogliptin (ALO) on RTBI-provoked endoplasmic reticulum (ER) stress and investigate the potential underlying mechanisms. For RTBI induction, rats were exposed to a sharp-edged weight at the right interior frontal area of the right cortex, one drop per day for five successive days. ALO (20 mg/kg/day, p.o.) was administered for one week. Results depicted that ALO recovered motor abnormalities and enhanced motor coordination in the open field test, decreased immobility and increased climbing time in the forced swimming test, and corrected histological aberrations. Moreover, ALO counteracted RTBI-triggered ER stress via suppression of activating transcription factor 6 (ATF6), glucose-regulated protein 78 (GRP78), aggregation of β-amyloid and Tau proteins, as well as elevation of the cortical content of brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin receptor kinase B (TrKB). ALO also exhibited an antioxidant and anti-inflammatory potential in addition to its effect on the gene expression of miRNAs (miRNA-322 and miRNA-125b). In conclusion, ALO exhibited a neuroprotective effect by mitigating ER stress induced in an RTBI rat model.

重复性创伤性脑损伤(RTBI)是指由外部机械力引起的脑损伤,通常会引起严重的神经系统后果。本研究旨在探讨阿格列汀(ALO)对rtbi引起的内质网(ER)应激的神经保护作用,并探讨其潜在机制。对于RTBI诱导,大鼠在右侧皮质的右侧内额叶区暴露于锋利的边缘重量,每天一滴,连续五天。alo20 mg/kg/天,口服1周。结果显示,在野外测试中,ALO恢复了运动异常,增强了运动协调,在强迫游泳测试中减少了不动和增加了攀爬时间,并纠正了组织学畸变。此外,ALO通过抑制激活转录因子6 (ATF6)、葡萄糖调节蛋白78 (GRP78)、β-淀粉样蛋白和Tau蛋白的聚集,以及脑源性神经营养因子(BDNF)及其受体原肌球蛋白受体激酶B (TrKB)皮质含量的升高,抵消了rtbi触发的内质网应激。除了影响miRNAs (miRNA-322和miRNA-125b)的基因表达外,ALO还表现出抗氧化和抗炎潜能。综上所述,在RTBI大鼠模型中,ALO通过减轻内质网应激表现出神经保护作用。
{"title":"Repositioning of Alogliptin to Mitigate Secondary Injury Induced by Repetitive TBI: Potential Role of its Antioxidant and Anti- Inflammatory Effects.","authors":"Hossam A Raslan, Haidy E Michel, Esther T Menze, Amira A El-Gazar","doi":"10.1007/s11481-025-10271-w","DOIUrl":"10.1007/s11481-025-10271-w","url":null,"abstract":"<p><p>Repetitive traumatic brain injury (RTBI) refers to brain injuries resulting from an external mechanical force causing cumulative and frequently severe neurological consequences. This study aimed to explore the neuroprotective effect of alogliptin (ALO) on RTBI-provoked endoplasmic reticulum (ER) stress and investigate the potential underlying mechanisms. For RTBI induction, rats were exposed to a sharp-edged weight at the right interior frontal area of the right cortex, one drop per day for five successive days. ALO (20 mg/kg/day, p.o.) was administered for one week. Results depicted that ALO recovered motor abnormalities and enhanced motor coordination in the open field test, decreased immobility and increased climbing time in the forced swimming test, and corrected histological aberrations. Moreover, ALO counteracted RTBI-triggered ER stress via suppression of activating transcription factor 6 (ATF6), glucose-regulated protein 78 (GRP78), aggregation of β-amyloid and Tau proteins, as well as elevation of the cortical content of brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin receptor kinase B (TrKB). ALO also exhibited an antioxidant and anti-inflammatory potential in addition to its effect on the gene expression of miRNAs (miRNA-322 and miRNA-125b). In conclusion, ALO exhibited a neuroprotective effect by mitigating ER stress induced in an RTBI rat model.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"21 1","pages":"6"},"PeriodicalIF":3.5,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12799724/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145960749","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Dexmedetomidine Mitigates Sevoflurane-Induced Neurodevelopmental Effects in Paediatric Anaesthesia: A Meta-Analysis and Preclinical Study. 右美托咪定减轻小儿麻醉中七氟醚诱导的神经发育影响:荟萃分析和临床前研究。
Hsuan-Chih Lao, Chia-Wei Huang, Ssu-Han Wang, Yen-Lin Su, Chien-Hui Chang, Cheng-Yen Liao, Jen-Chieh Wu, Ying-Chun Lin, Jin-Wu Tsai

Sevoflurane (Sevo) anaesthesia in children is linked to an increased incidence of postoperative emergence agitation (EA) and potential neurotoxicity in developing brains. However, the specific risks of subanaesthetic foetal or neonatal exposure to Sevo remain unclear. This study evaluates the safety and efficacy of combining dexmedetomidine (Dex) with Sevo to manage EA in paediatric anaesthesia. A systematic review and meta-analysis of randomized controlled clinical trials involving children under 8 years old revealed that Dex significantly reduces EA incidence when administered via intravenous, perineural, and intranasal routes. Using in utero electroporation, we found that pregnant mice exposed to 2.5% Sevo at embryonic days 14.5 and 15.5 exhibited transient neuronal migration deficits, with 25% of neurons delayed in deeper cortical layers. However, these neurons migrated to the cortex by postnatal day 8. Neonatal mice exposed to 2.5% Sevo experienced a 10% reduction in dendritic spine density in adolescence, associated with impaired somatosensory function, as assessed by the Von Frey test. Remarkably, Dex pretreatment ameliorated these pathological and functional changes. Thus, foetal or neonatal Sevo exposure can delay neuronal migration and reduce dendritic spine density. Dex co-administration effectively mitigates these adverse outcomes, supporting its potential use in paediatric anaesthesia to protect developing brains.

儿童七氟醚(Sevo)麻醉与术后出现躁动(EA)发生率增加和发育中的大脑潜在神经毒性有关。然而,亚麻醉胎儿或新生儿暴露于Sevo的具体风险仍不清楚。本研究评估右美托咪定(Dex)与Sevo联合治疗儿科麻醉EA的安全性和有效性。一项针对8岁以下儿童的随机对照临床试验的系统回顾和荟萃分析显示,通过静脉、神经周和鼻内给药,右美托咪定可显著降低EA的发生率。利用子宫电穿孔,我们发现在胚胎期14.5和15.5暴露于2.5% Sevo的怀孕小鼠表现出短暂的神经元迁移缺陷,其中25%的神经元在更深的皮质层延迟。然而,这些神经元在出生后第8天迁移到皮层。Von Frey测试显示,暴露于2.5% Sevo的新生小鼠在青春期的树突棘密度降低了10%,与体感觉功能受损有关。值得注意的是,右美托咪唑预处理改善了这些病理和功能变化。因此,胎儿或新生儿暴露于Sevo可延迟神经元迁移和减少树突棘密度。右美托咪定联合给药可有效减轻这些不良后果,支持其在儿科麻醉中保护发育中的大脑的潜在应用。
{"title":"Dexmedetomidine Mitigates Sevoflurane-Induced Neurodevelopmental Effects in Paediatric Anaesthesia: A Meta-Analysis and Preclinical Study.","authors":"Hsuan-Chih Lao, Chia-Wei Huang, Ssu-Han Wang, Yen-Lin Su, Chien-Hui Chang, Cheng-Yen Liao, Jen-Chieh Wu, Ying-Chun Lin, Jin-Wu Tsai","doi":"10.1007/s11481-025-10273-8","DOIUrl":"10.1007/s11481-025-10273-8","url":null,"abstract":"<p><p>Sevoflurane (Sevo) anaesthesia in children is linked to an increased incidence of postoperative emergence agitation (EA) and potential neurotoxicity in developing brains. However, the specific risks of subanaesthetic foetal or neonatal exposure to Sevo remain unclear. This study evaluates the safety and efficacy of combining dexmedetomidine (Dex) with Sevo to manage EA in paediatric anaesthesia. A systematic review and meta-analysis of randomized controlled clinical trials involving children under 8 years old revealed that Dex significantly reduces EA incidence when administered via intravenous, perineural, and intranasal routes. Using in utero electroporation, we found that pregnant mice exposed to 2.5% Sevo at embryonic days 14.5 and 15.5 exhibited transient neuronal migration deficits, with 25% of neurons delayed in deeper cortical layers. However, these neurons migrated to the cortex by postnatal day 8. Neonatal mice exposed to 2.5% Sevo experienced a 10% reduction in dendritic spine density in adolescence, associated with impaired somatosensory function, as assessed by the Von Frey test. Remarkably, Dex pretreatment ameliorated these pathological and functional changes. Thus, foetal or neonatal Sevo exposure can delay neuronal migration and reduce dendritic spine density. Dex co-administration effectively mitigates these adverse outcomes, supporting its potential use in paediatric anaesthesia to protect developing brains.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"21 1","pages":"5"},"PeriodicalIF":3.5,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12779744/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145914028","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Gut Microbiome-Sphingolipid Metabolism-Brain Axis Interactions: Neuroprotective Effects of Amitriptyline as Functional Inhibitor of Acid Sphingomyelinase in a Mouse Model of Tauopathy. 肠道微生物群-鞘脂代谢-脑轴相互作用:阿米替林作为酸性鞘磷脂酶功能抑制剂在牛皮病小鼠模型中的神经保护作用。
Mennatallah Ibrahim, Asmaa M Khalil, Heba Attia, Saleh Alseekh, Ahmed F Mohamed, Mohammed F El-Yamany

Tauopathies are neurodegenerative diseases characterized by accumulation of hyperphosphorylated tau protein (P-tau). The gut microbiota (GM) is symbiotic with the host and altered in neurodegenerative diseases. Amitriptyline (AMI) is a functional inhibitor of acid sphingomyelinase (ASM) which is abnormally highly expressed in brains of Alzheimer patients. Little data is known about the role of colonic ASM in management of tauopathy. Therefore, the aim of this study was to investigate the role of AMI on reversing gut dysbiosis, ceramide levels, colonic inflammation and intestinal barrier disruption in tauopathy through the bidirectional gut-brain axis. P301S transgenic mice were administered AMI for 35 days. Colonic ASM, ceramides, inflammation and membrane integrity were assessed besides fecal microbiome analysis and serum lipopolysaccharides to assess intestinal membrane disruption. Levels of hippocampal P-tau, protein phosphatase 2 A and neurogenesis were assessed along with cognitive behavior. AMI treatment significantly reduced colonic ASM, ceramide levels, increased abundance of Harryflintia, Dubosiella, and Parasutterella and decreased abundance of Lactobacillus, Lachnoclostridium, Oscillibacter, Oscillospiracea UCG-003, Colidextribacter, Roseburia, Butyricicoccus, and Sphingomondales. In contrast, P301S mice displayed an altered GM profile with enriched Firmicutes and Clostridia, and low proportions of Bacteroidota- a phylum associated with intestinal barrier protection-, and Ruminococcaceae. Also, AMI treatment decreased inflammation and restored colonic membrane integrity with subsequent decrease in serum lipopolysaccharides, P-tau in hippocampus and improvement in cognitive behaviour and neurogenesis. The current results indicate that AMI has neuroprotective effects against tauopathy through modulation of ASM activity, associated ceramide levels, GM composition, colonic inflammation and membrane integrity through bidirectional gut-brain axis.

tau病是神经退行性疾病,其特征是过度磷酸化tau蛋白(P-tau)的积累。肠道菌群(GM)与宿主共生,并在神经退行性疾病中发生改变。阿米替林(Amitriptyline, AMI)是阿尔茨海默病患者大脑中异常高表达的酸性鞘磷脂酶(ASM)的功能性抑制剂。关于结肠ASM在牛头病治疗中的作用知之甚少。因此,本研究的目的是探讨AMI通过双向肠-脑轴逆转牛头病肠道生态失调、神经酰胺水平、结肠炎症和肠屏障破坏的作用。P301S转基因小鼠给予AMI治疗35天。除了粪便微生物组分析和血清脂多糖评估外,还评估结肠ASM、神经酰胺、炎症和膜完整性。海马P-tau、蛋白磷酸酶2a和神经发生水平与认知行为一起被评估。AMI治疗显著降低了结肠ASM、神经酰胺水平,增加了Harryflintia、Dubosiella和Parasutterella的丰度,降低了Lactobacillus、Lachnoclostridium、Oscillibacter、Oscillospiracea UCG-003、Colidextribacter、Roseburia、Butyricicoccus和Sphingomondales的丰度。相比之下,P301S小鼠的转基因基因谱发生了改变,其中厚壁菌门和梭状芽胞杆菌丰富,拟杆菌门(一种与肠道屏障保护有关的门)和瘤胃球菌科的比例较低。此外,AMI治疗减少炎症,恢复结肠膜完整性,随后降低血清脂多糖,海马P-tau,改善认知行为和神经发生。目前的研究结果表明,AMI通过双向肠-脑轴调节ASM活性、相关神经酰胺水平、GM成分、结肠炎症和膜完整性,对牛头病具有神经保护作用。
{"title":"Gut Microbiome-Sphingolipid Metabolism-Brain Axis Interactions: Neuroprotective Effects of Amitriptyline as Functional Inhibitor of Acid Sphingomyelinase in a Mouse Model of Tauopathy.","authors":"Mennatallah Ibrahim, Asmaa M Khalil, Heba Attia, Saleh Alseekh, Ahmed F Mohamed, Mohammed F El-Yamany","doi":"10.1007/s11481-025-10270-x","DOIUrl":"10.1007/s11481-025-10270-x","url":null,"abstract":"<p><p>Tauopathies are neurodegenerative diseases characterized by accumulation of hyperphosphorylated tau protein (P-tau). The gut microbiota (GM) is symbiotic with the host and altered in neurodegenerative diseases. Amitriptyline (AMI) is a functional inhibitor of acid sphingomyelinase (ASM) which is abnormally highly expressed in brains of Alzheimer patients. Little data is known about the role of colonic ASM in management of tauopathy. Therefore, the aim of this study was to investigate the role of AMI on reversing gut dysbiosis, ceramide levels, colonic inflammation and intestinal barrier disruption in tauopathy through the bidirectional gut-brain axis. P301S transgenic mice were administered AMI for 35 days. Colonic ASM, ceramides, inflammation and membrane integrity were assessed besides fecal microbiome analysis and serum lipopolysaccharides to assess intestinal membrane disruption. Levels of hippocampal P-tau, protein phosphatase 2 A and neurogenesis were assessed along with cognitive behavior. AMI treatment significantly reduced colonic ASM, ceramide levels, increased abundance of Harryflintia, Dubosiella, and Parasutterella and decreased abundance of Lactobacillus, Lachnoclostridium, Oscillibacter, Oscillospiracea UCG-003, Colidextribacter, Roseburia, Butyricicoccus, and Sphingomondales. In contrast, P301S mice displayed an altered GM profile with enriched Firmicutes and Clostridia, and low proportions of Bacteroidota- a phylum associated with intestinal barrier protection-, and Ruminococcaceae. Also, AMI treatment decreased inflammation and restored colonic membrane integrity with subsequent decrease in serum lipopolysaccharides, P-tau in hippocampus and improvement in cognitive behaviour and neurogenesis. The current results indicate that AMI has neuroprotective effects against tauopathy through modulation of ASM activity, associated ceramide levels, GM composition, colonic inflammation and membrane integrity through bidirectional gut-brain axis.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"21 1","pages":"3"},"PeriodicalIF":3.5,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12764700/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145897151","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Prenatal Interferon-Alpha Exposure Induces Autism-Like Neurobehavioral and Neurochemical Alterations in Male Offspring. 产前干扰素- α暴露诱导雄性后代自闭症样神经行为和神经化学改变。
Güliz Otkıran, Mümin Alper Erdoğan, Yiğit Uyanıkgil, Oytun Erbaş

Maternal immune activation (MIA) during pregnancy has been implicated as a key environmental risk factor in autism spectrum disorder (ASD). Interferon-alpha (IFN-α), a type I interferon, may disrupt fetal neurodevelopment, yet its mechanistic impact remains insufficiently understood. This study explores the effects of maternal IFN-α exposure on neurobehavioral and neurobiological outcomes in a Wistar rat model. Pregnant rats received IFN-α on gestational day 10, and offspring were evaluated through behavioral assays, neurochemical analyses, and histopathological assessments. IFN-α exposure resulted in significant reductions in GABA, 5-HIAA, and GAD-67 levels, particularly in male offspring, indicating neurotransmitter dysregulation. Histologically, neuronal loss was observed in the hippocampal CA1 and CA3 regions and cerebellar Purkinje cells. Astrocyte activation, reflected by increased GFAP immunoreactivity, was prominent, suggesting a neuroinflammatory response. Additionally, reduced brain-derived neurotrophic factor (BDNF) and elevated tumor necrosis factor-alpha (TNF-α) levels support the presence of inflammation-induced synaptic dysfunction and impaired neuroplasticity. Behaviorally, male offspring exhibited reduced sociability and impaired social novelty recognition. Both sexes demonstrated deficits in motor coordination and exploratory activity. These findings align with core ASD phenotypes and underscore a heightened male vulnerability. Overall, the study provides compelling evidence that prenatal IFN-α exposure leads to persistent neuroimmune, neurochemical, and structural alterations resembling ASD. The results highlight the need for further research into immune-mediated neurodevelopmental disruptions and sex-specific vulnerabilities, offering potential pathways for preventive and therapeutic interventions targeting MIA-related risk mechanisms.

妊娠期母体免疫激活(MIA)已被认为是自闭症谱系障碍(ASD)的关键环境风险因素。干扰素-α (IFN-α)是一种I型干扰素,可能会破坏胎儿神经发育,但其机制影响仍未充分了解。本研究在Wistar大鼠模型中探讨母体IFN-α暴露对神经行为和神经生物学结果的影响。怀孕大鼠在妊娠第10天接受IFN-α治疗,并通过行为分析、神经化学分析和组织病理学评估后代。IFN-α暴露导致GABA, 5-HIAA和GAD-67水平显著降低,特别是在雄性后代中,表明神经递质失调。组织学上,海马CA1、CA3区及小脑浦肯野细胞均出现神经元丢失。星形胶质细胞激活,反映在GFAP免疫反应性增加,是突出的,提示神经炎症反应。此外,脑源性神经营养因子(BDNF)的减少和肿瘤坏死因子-α (TNF-α)水平的升高支持炎症诱导的突触功能障碍和神经可塑性受损的存在。在行为上,雄性后代表现出社交能力下降和社会新颖性认知受损。男女均表现出运动协调和探索活动的缺陷。这些发现与核心的ASD表型一致,并强调了男性的易感性。总的来说,该研究提供了令人信服的证据,表明产前暴露于IFN-α会导致持续的神经免疫、神经化学和结构改变,类似于ASD。这些结果强调了进一步研究免疫介导的神经发育中断和性别特异性脆弱性的必要性,为针对mia相关风险机制的预防和治疗干预提供了潜在的途径。
{"title":"Prenatal Interferon-Alpha Exposure Induces Autism-Like Neurobehavioral and Neurochemical Alterations in Male Offspring.","authors":"Güliz Otkıran, Mümin Alper Erdoğan, Yiğit Uyanıkgil, Oytun Erbaş","doi":"10.1007/s11481-025-10275-6","DOIUrl":"10.1007/s11481-025-10275-6","url":null,"abstract":"<p><p>Maternal immune activation (MIA) during pregnancy has been implicated as a key environmental risk factor in autism spectrum disorder (ASD). Interferon-alpha (IFN-α), a type I interferon, may disrupt fetal neurodevelopment, yet its mechanistic impact remains insufficiently understood. This study explores the effects of maternal IFN-α exposure on neurobehavioral and neurobiological outcomes in a Wistar rat model. Pregnant rats received IFN-α on gestational day 10, and offspring were evaluated through behavioral assays, neurochemical analyses, and histopathological assessments. IFN-α exposure resulted in significant reductions in GABA, 5-HIAA, and GAD-67 levels, particularly in male offspring, indicating neurotransmitter dysregulation. Histologically, neuronal loss was observed in the hippocampal CA1 and CA3 regions and cerebellar Purkinje cells. Astrocyte activation, reflected by increased GFAP immunoreactivity, was prominent, suggesting a neuroinflammatory response. Additionally, reduced brain-derived neurotrophic factor (BDNF) and elevated tumor necrosis factor-alpha (TNF-α) levels support the presence of inflammation-induced synaptic dysfunction and impaired neuroplasticity. Behaviorally, male offspring exhibited reduced sociability and impaired social novelty recognition. Both sexes demonstrated deficits in motor coordination and exploratory activity. These findings align with core ASD phenotypes and underscore a heightened male vulnerability. Overall, the study provides compelling evidence that prenatal IFN-α exposure leads to persistent neuroimmune, neurochemical, and structural alterations resembling ASD. The results highlight the need for further research into immune-mediated neurodevelopmental disruptions and sex-specific vulnerabilities, offering potential pathways for preventive and therapeutic interventions targeting MIA-related risk mechanisms.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"21 1","pages":"4"},"PeriodicalIF":3.5,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145897156","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Modulation of Cognitive Function by TRPM3 Channels in the Dentate Gyrus of a Menopausal Rat Model: Effects of Naringenin Treatment and Ciliary Neurotrophic Factor. 绝经大鼠齿状回TRPM3通道对认知功能的调节:柚皮素治疗和纤毛神经营养因子的影响
Kamini R Shirasath, Kartik T Nakhate, Sameer N Goyal, Sanjay N Awathale

Although transient receptor potential melastatin 3 (TRPM3) channels are primarily known for their role in spinal nociception, emerging evidence suggests their involvement in psychiatric conditions and central reward processing. Menopause, characterized by estrogen decline, induces neuroimmune activation and increases pro-inflammatory factors such as ciliary neurotrophic factor (CNTF). While a direct regulatory effect of CNTF on TRPM3 is not well established, both are involved in inflammation-related signaling, suggesting potential crosstalk. TRPM3 responds to neuroinflammatory and neurotrophic signals and may contribute to postmenopausal cognitive decline. However, this link remains unexplored. Naringenin, a natural flavonoid with estrogen-like properties, has been reported to inhibit TRPM3 channels and may help to alleviate postmenopausal memory impairment. This study aimed to investigate the role of elevated CNTF levels in increasing TRPM3 expression in the dentate gyrus (DG), contributing to cognitive deficits, and to assess the potential of naringenin in reversing these effects. Bilateral ovariectomy (OVX) was performed on female Sprague-Dawley rats, followed by treatment with naringenin (2.5, 5 and 10 mg/kg, intraperitoneal) for 14 days. Cognitive functions were assessed using the novel object recognition and passive avoidance tests. CNTF levels in the plasma and the DG, along with TRPM3 expression in the DG, were measured using ELISA and immunohistochemistry, respectively. Dendritic arborization in DG neurons was analyzed using Golgi-Cox staining. OVX rats showed impaired cognition, elevated CNTF and TRPM3 expression, and reduced dendritic complexity. Naringenin treatment reversed these changes, suggesting its potential to improve postmenopausal cognitive decline by modulating CNTF levels and TRPM3 activity in the DG.

虽然短暂受体电位美拉他汀3 (TRPM3)通道主要以其在脊髓伤害感受中的作用而闻名,但新出现的证据表明它们参与精神疾病和中枢奖励处理。更年期以雌激素下降为特征,诱导神经免疫激活并增加促炎因子,如纤毛神经营养因子(CNTF)。虽然CNTF对TRPM3的直接调节作用尚未完全确定,但两者都参与炎症相关的信号传导,表明可能存在串扰。TRPM3响应神经炎症和神经营养信号,并可能导致绝经后认知能力下降。然而,这种联系仍未被探索。柚皮素是一种具有雌激素样特性的天然类黄酮,据报道可抑制TRPM3通道,可能有助于缓解绝经后记忆障碍。本研究旨在探讨CNTF水平升高在增加齿状回(DG)中TRPM3表达中的作用,从而导致认知缺陷,并评估柚皮素逆转这些影响的潜力。雌性Sprague-Dawley大鼠行双侧卵巢切除术(OVX),随后给予柚皮素(2.5、5和10 mg/kg,腹腔注射)治疗14 d。认知功能评估使用新的对象识别和被动回避测试。分别用ELISA和免疫组织化学方法测定血浆和DG中的CNTF水平以及DG中的TRPM3表达。采用高尔基-考克斯染色法分析DG神经元的树突化。OVX大鼠表现出认知障碍,CNTF和TRPM3表达升高,树突复杂性降低。柚皮素治疗逆转了这些变化,表明其可能通过调节DG中的CNTF水平和TRPM3活性来改善绝经后认知能力下降。
{"title":"Modulation of Cognitive Function by TRPM3 Channels in the Dentate Gyrus of a Menopausal Rat Model: Effects of Naringenin Treatment and Ciliary Neurotrophic Factor.","authors":"Kamini R Shirasath, Kartik T Nakhate, Sameer N Goyal, Sanjay N Awathale","doi":"10.1007/s11481-025-10269-4","DOIUrl":"10.1007/s11481-025-10269-4","url":null,"abstract":"<p><p>Although transient receptor potential melastatin 3 (TRPM3) channels are primarily known for their role in spinal nociception, emerging evidence suggests their involvement in psychiatric conditions and central reward processing. Menopause, characterized by estrogen decline, induces neuroimmune activation and increases pro-inflammatory factors such as ciliary neurotrophic factor (CNTF). While a direct regulatory effect of CNTF on TRPM3 is not well established, both are involved in inflammation-related signaling, suggesting potential crosstalk. TRPM3 responds to neuroinflammatory and neurotrophic signals and may contribute to postmenopausal cognitive decline. However, this link remains unexplored. Naringenin, a natural flavonoid with estrogen-like properties, has been reported to inhibit TRPM3 channels and may help to alleviate postmenopausal memory impairment. This study aimed to investigate the role of elevated CNTF levels in increasing TRPM3 expression in the dentate gyrus (DG), contributing to cognitive deficits, and to assess the potential of naringenin in reversing these effects. Bilateral ovariectomy (OVX) was performed on female Sprague-Dawley rats, followed by treatment with naringenin (2.5, 5 and 10 mg/kg, intraperitoneal) for 14 days. Cognitive functions were assessed using the novel object recognition and passive avoidance tests. CNTF levels in the plasma and the DG, along with TRPM3 expression in the DG, were measured using ELISA and immunohistochemistry, respectively. Dendritic arborization in DG neurons was analyzed using Golgi-Cox staining. OVX rats showed impaired cognition, elevated CNTF and TRPM3 expression, and reduced dendritic complexity. Naringenin treatment reversed these changes, suggesting its potential to improve postmenopausal cognitive decline by modulating CNTF levels and TRPM3 activity in the DG.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"21 1","pages":"1"},"PeriodicalIF":3.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145893550","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Telitacicept as a New Therapeutic Avenue for Generalized Myasthenia Gravis and Thymoma-Associated Myasthenia Gravis. 泰利他赛普治疗广泛性重症肌无力及胸腺瘤相关性重症肌无力的新途径。
Ying Zhu, Benqiao Wang, Chunxiang Hu, Ruixia Zhu

Generalized myasthenia gravis (gMG) is an antibody mediated autoimmune neuromuscular junction disorder characterized by muscle weakness and fatigue as well as acetylcholine receptor antibody (AChR-Ab) as the main presence. A proportion of patients fail to achieve minimal symptom expression (MSE), furthermore 10-20% of them develop into refractory under conventional immunotherapy. We conducted a retrospective study to explore the effectiveness and safety of telitacicept in gMG and thymoma-associated MG (TAMG) patients. The treatment response was assessed by the variation of QMG, MG-ADL and MG-QOL-15 scores. Time to MSE as well as usage of corticosteroid were also evaluated. In this retrospective study, we included 22 AChR-gMG patients (15 women, 7 men), including 7 refractory and 12 TAMG, who were treated with telitacicept by following-up at least 6 months. Compared to the baseline, a significant decrease in QMG, ADL and MG-QOL-15 scores was observed at every visit, especially for the QMG score with at least 3 points decline in all the patients in week 4. Twenty patients attained MSE and the time to MSE was 4 months during the observed period. At the last follow-up, the dose of prednisone of all the patients treated with telitacicept was ≤ 5 mg/d. The AChR-Ab titers and CD19+ B cells significantly decreased from baseline to week 24. Telitacicept is generally well tolerated, the most common (18%) adverse effect was mild and transient injected site swelling. Our study provides evidence to support that telitacicept is beneficial and well tolerated in the management of gMG especially in refractory MG and TAMG. Clinical outcomes showed increased efficacy of telitacicept when used earlier in the disease course, which leads to a sparing of prednisone.

全身性重症肌无力(gMG)是一种以肌肉无力和疲劳为特征,以乙酰胆碱受体抗体(AChR-Ab)为主要表现的抗体介导的自身免疫性神经肌肉连接障碍。部分患者未能达到最小症状表达(MSE),其中10-20%的患者在常规免疫治疗下发展为难治性。我们进行了一项回顾性研究,探讨泰利他塞普在gMG和胸腺瘤相关MG (TAMG)患者中的有效性和安全性。通过QMG、MG-ADL和MG-QOL-15评分的变化来评估治疗效果。同时还评估了到达MSE的时间以及皮质类固醇的使用情况。在这项回顾性研究中,我们纳入了22例AChR-gMG患者(15名女性,7名男性),其中7例难治性和12例tamm,这些患者接受了telitacicept治疗,随访至少6个月。与基线相比,每次就诊均观察到QMG、ADL和MG-QOL-15评分显著下降,特别是第4周所有患者的QMG评分至少下降3分。20例患者达到MSE,观察期间达到MSE的时间为4个月。末次随访时,所有接受泰利他塞普治疗的患者泼尼松剂量均≤5 mg/d。从基线到第24周,AChR-Ab滴度和CD19+ B细胞显著下降。Telitacicept一般耐受性良好,最常见的不良反应(18%)是轻微和短暂的注射部位肿胀。我们的研究提供了证据,支持泰利他赛普是有益的,耐受性良好的管理gMG,特别是难治性MG和TAMG。临床结果显示,在病程早期使用telitacicept的疗效增加,这导致强的松的节约。
{"title":"Telitacicept as a New Therapeutic Avenue for Generalized Myasthenia Gravis and Thymoma-Associated Myasthenia Gravis.","authors":"Ying Zhu, Benqiao Wang, Chunxiang Hu, Ruixia Zhu","doi":"10.1007/s11481-025-10272-9","DOIUrl":"https://doi.org/10.1007/s11481-025-10272-9","url":null,"abstract":"<p><p>Generalized myasthenia gravis (gMG) is an antibody mediated autoimmune neuromuscular junction disorder characterized by muscle weakness and fatigue as well as acetylcholine receptor antibody (AChR-Ab) as the main presence. A proportion of patients fail to achieve minimal symptom expression (MSE), furthermore 10-20% of them develop into refractory under conventional immunotherapy. We conducted a retrospective study to explore the effectiveness and safety of telitacicept in gMG and thymoma-associated MG (TAMG) patients. The treatment response was assessed by the variation of QMG, MG-ADL and MG-QOL-15 scores. Time to MSE as well as usage of corticosteroid were also evaluated. In this retrospective study, we included 22 AChR-gMG patients (15 women, 7 men), including 7 refractory and 12 TAMG, who were treated with telitacicept by following-up at least 6 months. Compared to the baseline, a significant decrease in QMG, ADL and MG-QOL-15 scores was observed at every visit, especially for the QMG score with at least 3 points decline in all the patients in week 4. Twenty patients attained MSE and the time to MSE was 4 months during the observed period. At the last follow-up, the dose of prednisone of all the patients treated with telitacicept was ≤ 5 mg/d. The AChR-Ab titers and CD19<sup>+</sup> B cells significantly decreased from baseline to week 24. Telitacicept is generally well tolerated, the most common (18%) adverse effect was mild and transient injected site swelling. Our study provides evidence to support that telitacicept is beneficial and well tolerated in the management of gMG especially in refractory MG and TAMG. Clinical outcomes showed increased efficacy of telitacicept when used earlier in the disease course, which leads to a sparing of prednisone.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"21 1","pages":"2"},"PeriodicalIF":3.5,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145893540","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Histamine H3 Receptor Antagonist, Thioperamide, Improves Behavioral and Neuropathological Changes Associated with Subclinical Hypersensitivity to a Cow's Milk Allergen. 组胺H3受体拮抗剂硫哌丁胺可改善与牛奶过敏原亚临床超敏反应相关的行为和神经病理改变
Danielle Germundson-Hermanson, Marilyn G Klug, Kumi Nagamoto-Combs

Mood and behavior-related comorbidities are often reported with food allergies, an atopic condition that elevates histamine (HA) levels in tissues and circulation. However, whether allergy-induced HA directly affects the central nervous system is unclear. Previously, we demonstrated that the levels of HA and its receptor subtype, H3 receptor (H3R), were elevated in the brains of mice with subclinical cow's milk allergy (CMA) generated by sensitizing C57BL/6J mice to a bovine whey allergen, β-lactoglobulin (BLG, Bos d 5). Furthermore, these BLG-sensitized CMA mice showed depression-like behavior associated with mast cell activation, neuroinflammation, and cortical demyelination, leading us to postulate that peripheral immune responses raised brain HA and dysregulated the neuronal histaminergic system. Hypothesizing that the autoregulatory function of H3R signaling is pivotal in eliciting altered behavior and neuropathologies, we investigated whether thioperamide, a brain-permeable H3R-selective antagonist, would attenuate the changes observed in CMA mice. Male and female CMA mice were fed a whey-containing diet for 2 weeks without or with thioperamide. While sensorimotor functions were not impaired in CMA mice of either sex, some aspects of affective and cognitive behaviors were significantly altered in males. Male CMA mice also showed more IgE-immunopositive, degranulated mast cells in the dura mater than females, regardless of thioperamide treatment. Importantly, thioperamide reduced CMA-associated behavioral and neuropathological changes in male mice, although it also uniquely affected female mice. Our results suggest that thioperamide ameliorates CMA-associated behavioral changes and neuropathologies via H3R inhibition in a sex-dependent manner.

情绪和行为相关的合并症通常与食物过敏一起报道,食物过敏是一种升高组织和循环中组胺(HA)水平的特应性疾病。然而,过敏诱导的血凝素是否直接影响中枢神经系统尚不清楚。先前,我们证明了HA及其受体亚型H3受体(H3R)的水平在C57BL/6J小鼠对牛乳清过敏原β-乳球蛋白(BLG, bod 5)致敏后产生的亚临床牛奶过敏(CMA)小鼠的大脑中升高。此外,这些blg致敏的CMA小鼠表现出与肥大细胞激活、神经炎症和皮质脱髓鞘相关的抑郁样行为,这使我们假设外周免疫反应提高了脑HA并调节了神经元组胺能系统。假设H3R信号的自调节功能在引发行为改变和神经病理中起关键作用,我们研究了硫哌丁胺,一种脑渗透性H3R选择性拮抗剂,是否会减弱CMA小鼠中观察到的变化。雄性和雌性CMA小鼠分别饲喂不含硫哌丁胺或含硫哌丁胺的含乳清饲料2周。虽然CMA小鼠的感觉运动功能没有受到损害,但雄性的情感和认知行为的某些方面却发生了显著改变。无论硫哌丁胺治疗,雄性CMA小鼠的硬脑膜中也显示出更多的ige免疫阳性、脱颗粒肥大细胞。重要的是,硫哌丁胺减少了雄性小鼠与cma相关的行为和神经病理变化,尽管它也只影响雌性小鼠。我们的研究结果表明,硫哌丁胺通过抑制H3R以性别依赖的方式改善cma相关的行为改变和神经病理。
{"title":"Histamine H3 Receptor Antagonist, Thioperamide, Improves Behavioral and Neuropathological Changes Associated with Subclinical Hypersensitivity to a Cow's Milk Allergen.","authors":"Danielle Germundson-Hermanson, Marilyn G Klug, Kumi Nagamoto-Combs","doi":"10.1007/s11481-025-10256-9","DOIUrl":"10.1007/s11481-025-10256-9","url":null,"abstract":"<p><p>Mood and behavior-related comorbidities are often reported with food allergies, an atopic condition that elevates histamine (HA) levels in tissues and circulation. However, whether allergy-induced HA directly affects the central nervous system is unclear. Previously, we demonstrated that the levels of HA and its receptor subtype, H3 receptor (H3R), were elevated in the brains of mice with subclinical cow's milk allergy (CMA) generated by sensitizing C57BL/6J mice to a bovine whey allergen, β-lactoglobulin (BLG, Bos d 5). Furthermore, these BLG-sensitized CMA mice showed depression-like behavior associated with mast cell activation, neuroinflammation, and cortical demyelination, leading us to postulate that peripheral immune responses raised brain HA and dysregulated the neuronal histaminergic system. Hypothesizing that the autoregulatory function of H3R signaling is pivotal in eliciting altered behavior and neuropathologies, we investigated whether thioperamide, a brain-permeable H3R-selective antagonist, would attenuate the changes observed in CMA mice. Male and female CMA mice were fed a whey-containing diet for 2 weeks without or with thioperamide. While sensorimotor functions were not impaired in CMA mice of either sex, some aspects of affective and cognitive behaviors were significantly altered in males. Male CMA mice also showed more IgE-immunopositive, degranulated mast cells in the dura mater than females, regardless of thioperamide treatment. Importantly, thioperamide reduced CMA-associated behavioral and neuropathological changes in male mice, although it also uniquely affected female mice. Our results suggest that thioperamide ameliorates CMA-associated behavioral changes and neuropathologies via H3R inhibition in a sex-dependent manner.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"110"},"PeriodicalIF":3.5,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12717242/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145795630","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ameliorative Effect of Vitamin C Against Tramadol-Induced Learning and Memory Impairment in Juvenile Rat Via Attenuation of Oxidative Stress and Dysfunctional Synaptic Plasticity. 维生素C对曲马多诱导的幼年大鼠学习记忆障碍的改善作用及其对氧化应激和突触可塑性功能障碍的抑制作用。
Lily Mohammadipoor-Ghasemabad, Khadijeh Esmaeilpour, Manzumeh Shamsi Meymandi, Farhad Iranmanesh, Sheida Amiri Khorasani, Vahid Sheibani, Farahnaz Taheri

Tramadol (TM) abuse negatively affects the central nervous system, especially brain regions like the hippocampus involved in cognition. Recent studies have demonstrated neuroprotective effects of Vitamin C (Vit C) in various neurological diseases. No study has yet examined the effects of Vit C on tramadol-induced synaptic plasticity impairment. Therefore, we aimed to investigate the neuroprotective effects of Vit C on cognitive performance and synaptic plasticity in tramadol-exposed rats. Fifty-two juvenile male rats (30 days old) were divided into four groups: TM (30 mg/kg/day, intraperitoneally in the first week, 40 mg/kg/day in the second week and 50 mg/kg/day in third and fourth weeks), Vit.C (200 mg/kg/day, orally for 4 weeks), TM + Vit.C (as in the TM and Vit C groups, Vit C administered half an hour prior to TM), and Ctrl (0.25 mL saline/day for 4 weeks). Behavioral tests (open field, Morris water maze, novel object recognition) assessed locomotor activity and memory. In vivo recordings evaluated synaptic plasticity, and hippocampal oxidative stress markers [malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), total antioxidant capacity (TAC)] were measured according to the manufacturers' protocols with ELISA. TM caused learning and memory deficits, reduced long-term potentiation (LTP) induction, and disrupted the oxidative stress balance in the hippocampus. In contrast, Vit C inhibited these changes. These findings suggest that Vit C can attenuate cognitive impairments associated with chronic TM consumption, likely through modulation of hippocampal oxidative stress and enhancement of LTP induction. Therefore, Vit C could be a promising candidate for further investigation as a potential therapeutic agent to mitigate cognitive dysfunction associated with TM use.

曲马多(TM)滥用会对中枢神经系统产生负面影响,尤其是与认知有关的海马等大脑区域。最近的研究表明,维生素C (Vit C)在各种神经系统疾病中具有神经保护作用。尚未有研究证实Vit C对曲马多诱导的突触可塑性损伤的影响。因此,我们旨在研究Vit C对曲马多暴露大鼠认知能力和突触可塑性的神经保护作用。将52只30日龄雄性幼鼠分为四组:TM(第一周腹腔注射30 mg/kg/天,第二周注射40 mg/kg/天,第三、第四周注射50 mg/kg/天)、维生素C (200 mg/kg/天,口服4周)、TM +维生素C(与TM和Vit C组相同,Vit C在TM前半小时给药)和Ctrl (0.25 mL生理盐水/天,连续4周)。行为测试(开放场地,莫里斯水迷宫,新物体识别)评估运动活动和记忆。体内记录评估突触可塑性,并根据制造商的方案用ELISA测定海马氧化应激标志物[丙二醛(MDA)、超氧化物歧化酶(SOD)、过氧化氢酶(CAT)、总抗氧化能力(TAC)]。TM引起学习和记忆缺陷,降低长期增强(LTP)诱导,破坏海马氧化应激平衡。相反,维生素C抑制了这些变化。这些发现表明,Vit C可能通过调节海马氧化应激和增强LTP诱导来减轻与慢性TM消耗相关的认知障碍。因此,作为一种潜在的治疗药物,Vit C可能是一种有希望进一步研究的候选药物,可以减轻与TM使用相关的认知功能障碍。
{"title":"Ameliorative Effect of Vitamin C Against Tramadol-Induced Learning and Memory Impairment in Juvenile Rat Via Attenuation of Oxidative Stress and Dysfunctional Synaptic Plasticity.","authors":"Lily Mohammadipoor-Ghasemabad, Khadijeh Esmaeilpour, Manzumeh Shamsi Meymandi, Farhad Iranmanesh, Sheida Amiri Khorasani, Vahid Sheibani, Farahnaz Taheri","doi":"10.1007/s11481-025-10268-5","DOIUrl":"https://doi.org/10.1007/s11481-025-10268-5","url":null,"abstract":"<p><p>Tramadol (TM) abuse negatively affects the central nervous system, especially brain regions like the hippocampus involved in cognition. Recent studies have demonstrated neuroprotective effects of Vitamin C (Vit C) in various neurological diseases. No study has yet examined the effects of Vit C on tramadol-induced synaptic plasticity impairment. Therefore, we aimed to investigate the neuroprotective effects of Vit C on cognitive performance and synaptic plasticity in tramadol-exposed rats. Fifty-two juvenile male rats (30 days old) were divided into four groups: TM (30 mg/kg/day, intraperitoneally in the first week, 40 mg/kg/day in the second week and 50 mg/kg/day in third and fourth weeks), Vit.C (200 mg/kg/day, orally for 4 weeks), TM + Vit.C (as in the TM and Vit C groups, Vit C administered half an hour prior to TM), and Ctrl (0.25 mL saline/day for 4 weeks). Behavioral tests (open field, Morris water maze, novel object recognition) assessed locomotor activity and memory. In vivo recordings evaluated synaptic plasticity, and hippocampal oxidative stress markers [malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), total antioxidant capacity (TAC)] were measured according to the manufacturers' protocols with ELISA. TM caused learning and memory deficits, reduced long-term potentiation (LTP) induction, and disrupted the oxidative stress balance in the hippocampus. In contrast, Vit C inhibited these changes. These findings suggest that Vit C can attenuate cognitive impairments associated with chronic TM consumption, likely through modulation of hippocampal oxidative stress and enhancement of LTP induction. Therefore, Vit C could be a promising candidate for further investigation as a potential therapeutic agent to mitigate cognitive dysfunction associated with TM use.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"109"},"PeriodicalIF":3.5,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145745990","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fisetin Mitigates Ferroptosis and Promotes Remyelination in a Cuprizone Model of Multiple Sclerosis. 非瑟汀减轻铁质下垂并促进多发性硬化症铜酮模型中的再髓鞘形成。
Nahla E El-Ashmawy, Naglaa F Khedr, Nada N Helmy, Amera O Ibrahim

Multiple sclerosis (MS) is a long-lasting autoimmune condition characterized by myelin destruction and neurodegeneration. Research indicates that ferroptosis significantly influences MS pathogenesis, exacerbating neuronal tissue damage. Our study intended to explore the possible neuroprotective role of fisetin (FIS) in cuprizone (CPZ) model of MS and the associated molecular mechanisms. The 9-week experiment comprised a 5-week demyelination period in which C57BL/6 mice were provided with 0.2% w/w CPZ added to rodent chow, followed by a 4-week remyelination period in which mice were fed CPZ-free chow. FIS (80 mg/kg/day) was given by oral gavage to mice daily for 4 weeks starting in the 2nd week of demyelination. For remyelination, FIS was administered daily during the 4 weeks recovery. During demyelination, FIS significantly improved CPZ-induced behavioral and locomotor deficits, as demonstrated by tail suspension test and inverted screen grip strength test. LFB and H & E staining, MBP, GFAP and vimentin immunostaining revealed that FIS treatment significantly improved myelination, alleviated astrogliosis and neuronal injury in CPZ-fed mice throughout both phases. FIS attenuated ferroptosis and neuroinflammation during de- and remyelination as supported by reduced brain iron deposits, IL-1 β, MDA concentrations and restored GPX4. Moreover, FIS significantly downregulated NCOA4 and TfR1 gene expression and TfR1 protein level but upregulated FTH1 gene expression and ferritin protein level. Additionally, FIS upregulated Olig-1 during demyelination, but not remyelination. Fisetin has a potential neuroprotective effect in CPZ model of MS and can be studied as a promising adjuvant therapy to enhance remyelination and mitigate disability in MS patients possibly by modulating ferroptosis pathway.

多发性硬化症(MS)是一种以髓磷脂破坏和神经变性为特征的长期自身免疫性疾病。研究表明,铁下垂显著影响MS发病机制,加重神经组织损伤。本研究旨在探讨非瑟酮(FIS)在多发性硬化症铜酮(CPZ)模型中可能的神经保护作用及其分子机制。实验为期9周,第5周为脱髓鞘期,在C57BL/6小鼠的鼠粮中添加0.2% w/w的CPZ;第4周为脱髓鞘期,在此期间小鼠喂食不添加CPZ的鼠粮。从脱髓鞘第2周开始,每天灌胃FIS (80 mg/kg/天),连续4周。对于髓鞘再生,在4周恢复期间每天给予FIS。在脱髓鞘过程中,FIS显著改善cpz诱导的行为和运动缺陷,这在悬尾测试和倒立屏幕握力测试中得到了证实。LFB和h&e染色、MBP、GFAP和vimentin免疫染色显示,FIS治疗显著改善cpz喂养小鼠的髓鞘形成,减轻星形胶质细胞增生和神经元损伤。FIS通过减少脑铁沉积、IL-1 β、MDA浓度和恢复GPX4来减轻脱髓鞘和再髓鞘形成过程中的铁中毒和神经炎症。FIS显著下调NCOA4、TfR1基因表达和TfR1蛋白水平,上调FTH1基因表达和铁蛋白水平。此外,FIS在脱髓鞘过程中上调olig1,但不上调髓鞘再生。非西汀在MS CPZ模型中具有潜在的神经保护作用,可能通过调节铁凋亡通路,作为一种有前景的辅助治疗,增强MS患者的髓鞘再生和减轻残疾。
{"title":"Fisetin Mitigates Ferroptosis and Promotes Remyelination in a Cuprizone Model of Multiple Sclerosis.","authors":"Nahla E El-Ashmawy, Naglaa F Khedr, Nada N Helmy, Amera O Ibrahim","doi":"10.1007/s11481-025-10260-z","DOIUrl":"10.1007/s11481-025-10260-z","url":null,"abstract":"<p><p>Multiple sclerosis (MS) is a long-lasting autoimmune condition characterized by myelin destruction and neurodegeneration. Research indicates that ferroptosis significantly influences MS pathogenesis, exacerbating neuronal tissue damage. Our study intended to explore the possible neuroprotective role of fisetin (FIS) in cuprizone (CPZ) model of MS and the associated molecular mechanisms. The 9-week experiment comprised a 5-week demyelination period in which C57BL/6 mice were provided with 0.2% w/w CPZ added to rodent chow, followed by a 4-week remyelination period in which mice were fed CPZ-free chow. FIS (80 mg/kg/day) was given by oral gavage to mice daily for 4 weeks starting in the 2nd week of demyelination. For remyelination, FIS was administered daily during the 4 weeks recovery. During demyelination, FIS significantly improved CPZ-induced behavioral and locomotor deficits, as demonstrated by tail suspension test and inverted screen grip strength test. LFB and H & E staining, MBP, GFAP and vimentin immunostaining revealed that FIS treatment significantly improved myelination, alleviated astrogliosis and neuronal injury in CPZ-fed mice throughout both phases. FIS attenuated ferroptosis and neuroinflammation during de- and remyelination as supported by reduced brain iron deposits, IL-1 β, MDA concentrations and restored GPX4. Moreover, FIS significantly downregulated NCOA4 and TfR1 gene expression and TfR1 protein level but upregulated FTH1 gene expression and ferritin protein level. Additionally, FIS upregulated Olig-1 during demyelination, but not remyelination. Fisetin has a potential neuroprotective effect in CPZ model of MS and can be studied as a promising adjuvant therapy to enhance remyelination and mitigate disability in MS patients possibly by modulating ferroptosis pathway.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"108"},"PeriodicalIF":3.5,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12685980/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145710436","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bradykinin Type 2 Receptor Deficiency Reshapes Acute Neuroinflammation and Improves Cell Survival after Ischemic Stroke in Diabetic Mice. 缓激素2型受体缺乏重塑糖尿病小鼠缺血性卒中后的急性神经炎症并改善细胞存活
Anja Barić, Dinko Smilović, Helena Justić, Iva Šimunić, Siniša Škokić, Marina Dobrivojević Radmilović

Diabetes mellitus exacerbates cerebral ischemic damage by potentiating neuroinflammation. We hypothesized that activation of the bradykinin type 2 receptor, a mediator of inflammation and vascular dynamics, might be detrimental to ischemic injury development in diabetic animals. We monitored the acute phase of cerebral ischemia in type 1 diabetic mice, diabetic bradykinin type 2 receptor knock-out mice, and their non-diabetic controls using neurological assessment, magnetic resonance imaging, and a comprehensive immuno-histochemical and morphological analysis to quantify changes in microglial, neutrophil, and neuronal populations. Our findings reveal that bradykinin type 2 receptor deficiency ameliorates neurological deficit in non-diabetic mice, despite similar ischemic lesion volumes across all investigated groups. Furthermore, in non-diabetic animals, the bradykinin type 2 receptor plays a discernible role in edema resolution, neuroprotection, and regulation of microglial response to ischemia. However, diabetes, as a stroke comorbidity, alters the involvement of the bradykinin type 2 receptor in ischemic injury development. Bradykinin type 2 receptor-deficient diabetic animals demonstrate delayed microglial cell loss and reduced microglial reactivity following ischemia compared to diabetic animals with functional bradykinin type 2 receptors. The attenuated immune response is accompanied by a marked absence of infiltrating neutrophils within the ischemic territory and improved neuronal survival. This study demonstrates that diabetes profoundly modifies the role of bradykinin type 2 receptor in cerebral ischemic injury, influencing both acute neuroinflammation and cell survival. These findings support the potential of the bradykinin type 2 receptor as a therapeutic target for stroke in diabetic population, warranting further investigation.

糖尿病通过增强神经炎症加重脑缺血损伤。我们假设缓激素2型受体(炎症和血管动力学的介质)的激活可能对糖尿病动物的缺血性损伤发展有害。我们监测了1型糖尿病小鼠、糖尿病缓激肽2型受体敲除小鼠和非糖尿病对照组的急性期脑缺血,使用神经学评估、磁共振成像、综合免疫组织化学和形态学分析来量化小胶质细胞、中性粒细胞和神经元群体的变化。我们的研究结果表明,缓激肽2型受体缺乏改善了非糖尿病小鼠的神经功能缺损,尽管所有研究组的缺血性病变体积相似。此外,在非糖尿病动物中,缓激肽2型受体在水肿消退、神经保护和小胶质细胞对缺血反应的调节中起着明显的作用。然而,糖尿病作为卒中的合并症,改变了缓激素2型受体在缺血性损伤发展中的作用。与具有功能性缓激肽2型受体的糖尿病动物相比,缺乏缓激肽2型受体的糖尿病动物表现出缺血后小胶质细胞损失延迟和小胶质反应性降低。减弱的免疫反应伴随着缺血区域内浸润性中性粒细胞的明显缺失和神经元存活的改善。本研究表明,糖尿病深刻改变缓激素2型受体在脑缺血损伤中的作用,影响急性神经炎症和细胞存活。这些发现支持缓激肽2型受体作为糖尿病人群卒中治疗靶点的潜力,值得进一步研究。
{"title":"Bradykinin Type 2 Receptor Deficiency Reshapes Acute Neuroinflammation and Improves Cell Survival after Ischemic Stroke in Diabetic Mice.","authors":"Anja Barić, Dinko Smilović, Helena Justić, Iva Šimunić, Siniša Škokić, Marina Dobrivojević Radmilović","doi":"10.1007/s11481-025-10267-6","DOIUrl":"https://doi.org/10.1007/s11481-025-10267-6","url":null,"abstract":"<p><p>Diabetes mellitus exacerbates cerebral ischemic damage by potentiating neuroinflammation. We hypothesized that activation of the bradykinin type 2 receptor, a mediator of inflammation and vascular dynamics, might be detrimental to ischemic injury development in diabetic animals. We monitored the acute phase of cerebral ischemia in type 1 diabetic mice, diabetic bradykinin type 2 receptor knock-out mice, and their non-diabetic controls using neurological assessment, magnetic resonance imaging, and a comprehensive immuno-histochemical and morphological analysis to quantify changes in microglial, neutrophil, and neuronal populations. Our findings reveal that bradykinin type 2 receptor deficiency ameliorates neurological deficit in non-diabetic mice, despite similar ischemic lesion volumes across all investigated groups. Furthermore, in non-diabetic animals, the bradykinin type 2 receptor plays a discernible role in edema resolution, neuroprotection, and regulation of microglial response to ischemia. However, diabetes, as a stroke comorbidity, alters the involvement of the bradykinin type 2 receptor in ischemic injury development. Bradykinin type 2 receptor-deficient diabetic animals demonstrate delayed microglial cell loss and reduced microglial reactivity following ischemia compared to diabetic animals with functional bradykinin type 2 receptors. The attenuated immune response is accompanied by a marked absence of infiltrating neutrophils within the ischemic territory and improved neuronal survival. This study demonstrates that diabetes profoundly modifies the role of bradykinin type 2 receptor in cerebral ischemic injury, influencing both acute neuroinflammation and cell survival. These findings support the potential of the bradykinin type 2 receptor as a therapeutic target for stroke in diabetic population, warranting further investigation.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"107"},"PeriodicalIF":3.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145650276","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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
Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology
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