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
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":"https://doi.org/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":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145795630","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
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
The Small Molecule Compound Eupalinolide B Alleviates Neuropathic Pain by Regulating the USP7/Keap1/Nrf2 Pathway. 小分子化合物upalinolide B通过调节USP7/Keap1/Nrf2通路减轻神经性疼痛。
Xuesong Yang, Fan Jiang, Juan Li, Yanqiong Wu, Hongbing Xiang

Neuropathic pain is a chronic pain condition characterized by complex pathogenesis and poor prognosis. EB (Eupalinolide B), a highly bioactive sesquiterpene lactone derived from Eupatorium lindleyanum DC, has been demonstrated to possess multiple pharmacological activities, including antihistamine, antibacterial, and antioxidant effects. USP7 (ubiquitin-specific protease 7) is a crucial deubiquitinating enzyme in eukaryotes, while the Keap1, Nrf2, and HO-1 signaling pathways play pivotal roles in the development of neuropathic pain. Our study established a spared nerve injury model in mice and employed multiple molecular biology experiments to investigate the regulatory role of EB in the USP7/Keap1/Nrf2 pathway and its mechanisms in neuropathic pain. Results showed significantly elevated USP7 and Keap1 protein expression in the spinal cord of SNI mice, while Nrf2 and HO-1 levels were markedly reduced. EB treatment downregulated USP7 expression, promoted Keap1 ubiquitination and degradation, thereby elevating Nrf2/HO-1 protein levels. This inhibited microglial proliferation and M1 polarization, reduced the production of proinflammatory factors (TNF-α, IL-1β, IL-6), and significantly ameliorated mechanical and thermal hyperalgesia in SNI mice. Long-term intraperitoneal injection of EB did not cause any significant side effects in the heart, liver, or kidneys of SNI mice. In summary, EB exerts anti-inflammatory and analgesic effects by modulating the USP7/Keap1/Nrf2 signaling pathway, offering a potential novel therapeutic strategy for neuropathic pain.

神经性疼痛是一种发病复杂、预后差的慢性疼痛。EB (Eupalinolide B)是一种高生物活性的倍半萜内酯,从泽兰中提取,已被证明具有多种药理活性,包括抗组胺、抗菌和抗氧化作用。USP7(泛素特异性蛋白酶7)是真核生物中重要的去泛素化酶,而Keap1、Nrf2和HO-1信号通路在神经性疼痛的发生发展中起关键作用。本研究建立小鼠神经损伤模型,通过多种分子生物学实验,探讨EB对USP7/Keap1/Nrf2通路的调控作用及其在神经性疼痛中的机制。结果显示SNI小鼠脊髓USP7和Keap1蛋白表达显著升高,Nrf2和HO-1蛋白表达显著降低。EB处理下调USP7表达,促进Keap1泛素化和降解,从而提高Nrf2/HO-1蛋白水平。这抑制了小胶质细胞增殖和M1极化,减少了促炎因子(TNF-α, IL-1β, IL-6)的产生,并显著改善了SNI小鼠的机械和热痛觉过敏。长期腹腔注射EB对SNI小鼠的心脏、肝脏、肾脏均无明显副作用。综上所述,EB通过调节USP7/Keap1/Nrf2信号通路发挥抗炎和镇痛作用,为神经性疼痛的治疗提供了一种潜在的新策略。
{"title":"The Small Molecule Compound Eupalinolide B Alleviates Neuropathic Pain by Regulating the USP7/Keap1/Nrf2 Pathway.","authors":"Xuesong Yang, Fan Jiang, Juan Li, Yanqiong Wu, Hongbing Xiang","doi":"10.1007/s11481-025-10266-7","DOIUrl":"https://doi.org/10.1007/s11481-025-10266-7","url":null,"abstract":"<p><p>Neuropathic pain is a chronic pain condition characterized by complex pathogenesis and poor prognosis. EB (Eupalinolide B), a highly bioactive sesquiterpene lactone derived from Eupatorium lindleyanum DC, has been demonstrated to possess multiple pharmacological activities, including antihistamine, antibacterial, and antioxidant effects. USP7 (ubiquitin-specific protease 7) is a crucial deubiquitinating enzyme in eukaryotes, while the Keap1, Nrf2, and HO-1 signaling pathways play pivotal roles in the development of neuropathic pain. Our study established a spared nerve injury model in mice and employed multiple molecular biology experiments to investigate the regulatory role of EB in the USP7/Keap1/Nrf2 pathway and its mechanisms in neuropathic pain. Results showed significantly elevated USP7 and Keap1 protein expression in the spinal cord of SNI mice, while Nrf2 and HO-1 levels were markedly reduced. EB treatment downregulated USP7 expression, promoted Keap1 ubiquitination and degradation, thereby elevating Nrf2/HO-1 protein levels. This inhibited microglial proliferation and M1 polarization, reduced the production of proinflammatory factors (TNF-α, IL-1β, IL-6), and significantly ameliorated mechanical and thermal hyperalgesia in SNI mice. Long-term intraperitoneal injection of EB did not cause any significant side effects in the heart, liver, or kidneys of SNI mice. In summary, EB exerts anti-inflammatory and analgesic effects by modulating the USP7/Keap1/Nrf2 signaling pathway, offering a potential novel therapeutic strategy for neuropathic pain.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"106"},"PeriodicalIF":3.5,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145607665","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
The Ubiquitin-Proteasome System in Brain Disorders: Pathogenic Pathways, Post-Translational Tweaks, and Therapeutic Frontiers. 脑疾病中的泛素-蛋白酶体系统:致病途径、翻译后调整和治疗前沿。
Rohan Gupta, M Yasmin Begum, Reetesh Kumar, Jyoti Gupta, Rupak Nagraik, Siva Parsad Panda, Mosleh Mohammad Abomughaid, Sorabh Lakhanpal, Avinash D, Riyaz Ali M Osmani, Niraj Kumar Jha

Ubiquitination is a key enzymatic process where ubiquitin molecules covalently attach to substrate proteins, regulating their degradation, trafficking, and signaling. This process ensures cellular homeostasis by controlling protein quality and abundance, and it plays a vital role in immunity, DNA repair, and the cell cycle. Further, ubiquitination involves a sophisticated network of enzymes, domains, and receptors, providing pathway flexibility. However, dysregulation of ubiquitination due to aberrant enzyme function is implicated in various disorders, including cancer, diabetes, stroke, and neurodegenerative diseases (NDDs). Additionally, the ubiquitin-proteasome system (UPS) not only mediates protein degradation but also influences inflammation and subcellular localization. This review explores the pivotal role of ubiquitination and deubiquitination enzymes in the onset and progression of NDDs. It highlights their involvement in protein aggregation, mitochondrial impairment, neuroinflammation, and altered synaptic function. Special focus is placed on mutations in E3 ligases (e.g., E3 ubiquitin ligase encoded by PARK2 (Parkin), C-terminus of Hsp70-interacting protein (CHIP)) and deubiquitinases (e.g., USP14, ubiquitin C-terminal hydrolases (UCHL1)), which disrupt proteostasis and lead to the accumulation of neurotoxic proteins, such as Aβ, tau, α-synuclein, and mHtt. Moreover, post-translational modifications (PTMs), including phosphorylation, acetylation, and oxidative stress, further modulate UPS activity and disease progression. Lastly, the review also evaluates emerging therapeutic strategies aimed at restoring proteostasis, including proteasome-targeting small molecules (e.g., bortezomib, IU1-47), natural compounds (e.g., curcumin, resveratrol), RNA-based therapies (e.g., miR-101, circHIPK3), and dietary approaches (e.g., Mediterranean and ketogenic diets), offering a foundation for future neurodegenerative disease treatment.

泛素化是一个关键的酶促过程,泛素分子共价附着在底物蛋白上,调节底物蛋白的降解、运输和信号传导。这个过程通过控制蛋白质的质量和丰度来确保细胞内稳态,它在免疫、DNA修复和细胞周期中起着至关重要的作用。此外,泛素化涉及一个复杂的酶、结构域和受体网络,提供了途径的灵活性。然而,由于酶功能异常引起的泛素化失调与多种疾病有关,包括癌症、糖尿病、中风和神经退行性疾病(ndd)。此外,泛素-蛋白酶体系统(UPS)不仅介导蛋白质降解,还影响炎症和亚细胞定位。本文综述了泛素化和去泛素化酶在ndd发生和发展中的关键作用。它强调了它们参与蛋白质聚集、线粒体损伤、神经炎症和突触功能改变。特别关注的是E3连接酶(例如,由PARK2编码的E3泛素连接酶(Parkin), hsp70相互作用蛋白的c端(CHIP))和去泛素酶(例如,USP14,泛素c端水解酶(UCHL1))的突变,这些突变破坏了蛋白质平衡并导致神经毒性蛋白的积累,如Aβ, tau, α-synuclein和mHtt。此外,翻译后修饰(PTMs),包括磷酸化、乙酰化和氧化应激,进一步调节UPS活性和疾病进展。最后,该综述还评估了旨在恢复蛋白质平衡的新兴治疗策略,包括蛋白酶体靶向小分子(例如,硼铁唑米,IU1-47),天然化合物(例如,姜黄素,白藜芦醇),基于rna的疗法(例如,miR-101, circHIPK3)和饮食方法(例如,地中海和生酮饮食),为未来神经退行性疾病治疗提供基础。
{"title":"The Ubiquitin-Proteasome System in Brain Disorders: Pathogenic Pathways, Post-Translational Tweaks, and Therapeutic Frontiers.","authors":"Rohan Gupta, M Yasmin Begum, Reetesh Kumar, Jyoti Gupta, Rupak Nagraik, Siva Parsad Panda, Mosleh Mohammad Abomughaid, Sorabh Lakhanpal, Avinash D, Riyaz Ali M Osmani, Niraj Kumar Jha","doi":"10.1007/s11481-025-10258-7","DOIUrl":"https://doi.org/10.1007/s11481-025-10258-7","url":null,"abstract":"<p><p>Ubiquitination is a key enzymatic process where ubiquitin molecules covalently attach to substrate proteins, regulating their degradation, trafficking, and signaling. This process ensures cellular homeostasis by controlling protein quality and abundance, and it plays a vital role in immunity, DNA repair, and the cell cycle. Further, ubiquitination involves a sophisticated network of enzymes, domains, and receptors, providing pathway flexibility. However, dysregulation of ubiquitination due to aberrant enzyme function is implicated in various disorders, including cancer, diabetes, stroke, and neurodegenerative diseases (NDDs). Additionally, the ubiquitin-proteasome system (UPS) not only mediates protein degradation but also influences inflammation and subcellular localization. This review explores the pivotal role of ubiquitination and deubiquitination enzymes in the onset and progression of NDDs. It highlights their involvement in protein aggregation, mitochondrial impairment, neuroinflammation, and altered synaptic function. Special focus is placed on mutations in E3 ligases (e.g., E3 ubiquitin ligase encoded by PARK2 (Parkin), C-terminus of Hsp70-interacting protein (CHIP)) and deubiquitinases (e.g., USP14, ubiquitin C-terminal hydrolases (UCHL1)), which disrupt proteostasis and lead to the accumulation of neurotoxic proteins, such as Aβ, tau, α-synuclein, and mHtt. Moreover, post-translational modifications (PTMs), including phosphorylation, acetylation, and oxidative stress, further modulate UPS activity and disease progression. Lastly, the review also evaluates emerging therapeutic strategies aimed at restoring proteostasis, including proteasome-targeting small molecules (e.g., bortezomib, IU1-47), natural compounds (e.g., curcumin, resveratrol), RNA-based therapies (e.g., miR-101, circHIPK3), and dietary approaches (e.g., Mediterranean and ketogenic diets), offering a foundation for future neurodegenerative disease treatment.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"105"},"PeriodicalIF":3.5,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145566513","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
Activation of µ-δ Opioid Receptor Heteromer Attenuates Chemotherapy-Induced Neuropathic Pain in Mice. µ-δ阿片受体异聚体的激活可减轻小鼠化疗诱导的神经性疼痛。
Somesh Agrawal, Vinod Tiwari

Chemotherapy-induced neuropathic pain (CINP) affects up to 80% of cancer patients treated with cytostatic drugs like paclitaxel (PTX), leading to significant chronic sensorimotor dysfunction. Current pharmacological treatments often cause CNS side effects such as sedation and addiction. Increasing evidence indicates that native µ- and δ-opioid receptors (ORs) can associate to form heteromers in discrete brain regions. However, the role of µ-δ heteromer in CINP remains unclear. Therefore, we investigated the analgesic activity of CYM51010, a µ-δ heteromer agonist in CINP and how µ-δ heteromer activation regulates neuropathic pain. Systemic CYM51010 administration significantly alleviated evoked and ongoing pain in CINP mice, without inducing drug-seeking behavior, unlike morphine, which was consistent with earlier findings observed in SNL rats. Molecular analysis revealed that CYM51010 significantly decreased the increased TRPV1 and p38α expression in the dorsal root ganglion as well as spinal tissues of CINP mice. CYM51010 also reduced the expression of NF-κB, microglial markers (ICAM-1 & IBA1), and pro-inflammatory cytokines (TNF-α, IL-1β). Findings from the current study indicate that µ-δ heteromer activation represents a promising therapeutic target for chemotherapy-induced neuropathic pain (CINP), potentially enabling effective pain relief with reduced central side effects.

化疗引起的神经性疼痛(CINP)影响多达80%的接受紫杉醇(PTX)等细胞抑制药物治疗的癌症患者,导致显著的慢性感觉运动功能障碍。目前的药物治疗经常引起中枢神经系统的副作用,如镇静和成瘾。越来越多的证据表明,天然的µ-和δ-阿片受体(ORs)可以在离散的脑区形成异聚体。然而,µ-δ异构体在CINP中的作用尚不清楚。因此,我们研究了CYM51010(一种微δ异构体激动剂)在CINP中的镇痛活性,以及微δ异构体激活如何调节神经性疼痛。与吗啡不同,全身给药CYM51010显著减轻了CINP小鼠的诱发性和持续性疼痛,而不诱导寻求药物的行为,这与早期在SNL大鼠中观察到的结果一致。分子分析显示,CYM51010显著降低了CINP小鼠背根神经节和脊髓组织中TRPV1和p38α的表达。CYM51010还降低了NF-κB、小胶质细胞标志物(ICAM-1和IBA1)和促炎细胞因子(TNF-α、IL-1β)的表达。目前的研究结果表明,µ-δ异聚体激活代表了化疗诱导的神经性疼痛(CINP)的一个有希望的治疗靶点,可能有效缓解疼痛并减少中枢副作用。
{"title":"Activation of µ-δ Opioid Receptor Heteromer Attenuates Chemotherapy-Induced Neuropathic Pain in Mice.","authors":"Somesh Agrawal, Vinod Tiwari","doi":"10.1007/s11481-025-10264-9","DOIUrl":"https://doi.org/10.1007/s11481-025-10264-9","url":null,"abstract":"<p><p>Chemotherapy-induced neuropathic pain (CINP) affects up to 80% of cancer patients treated with cytostatic drugs like paclitaxel (PTX), leading to significant chronic sensorimotor dysfunction. Current pharmacological treatments often cause CNS side effects such as sedation and addiction. Increasing evidence indicates that native µ- and δ-opioid receptors (ORs) can associate to form heteromers in discrete brain regions. However, the role of µ-δ heteromer in CINP remains unclear. Therefore, we investigated the analgesic activity of CYM51010, a µ-δ heteromer agonist in CINP and how µ-δ heteromer activation regulates neuropathic pain. Systemic CYM51010 administration significantly alleviated evoked and ongoing pain in CINP mice, without inducing drug-seeking behavior, unlike morphine, which was consistent with earlier findings observed in SNL rats. Molecular analysis revealed that CYM51010 significantly decreased the increased TRPV1 and p38α expression in the dorsal root ganglion as well as spinal tissues of CINP mice. CYM51010 also reduced the expression of NF-κB, microglial markers (ICAM-1 & IBA1), and pro-inflammatory cytokines (TNF-α, IL-1β). Findings from the current study indicate that µ-δ heteromer activation represents a promising therapeutic target for chemotherapy-induced neuropathic pain (CINP), potentially enabling effective pain relief with reduced central side effects.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"103"},"PeriodicalIF":3.5,"publicationDate":"2025-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145524758","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
Neuroprotective Effects of Glucosamine in Huntington's Disease Through NLRP3 Inflammasome Inhibition. 氨基葡萄糖通过抑制NLRP3炎性体对亨廷顿病的神经保护作用。
Lichieh Julie Chu, Yi-Ting Hsu, Yu Aoh, Chih-Yuan Cheng, Hui-Hsuan Wang, Ming-Tse Wu, Hao-Hsuan Li, Xiu-Yuan Lai, Kuo-Feng Hua, Chiung-Mei Chen, Tz-Chuen Ju

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin (HTT) gene. It typically manifests as a triad of progressive psychiatric, cognitive, and motor symptoms. The resulting mutant HTT (mHTT) protein disrupts cellular homeostasis and promotes neuroinflammation. The NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome is a key mediator of neuroinflammatory responses, activating caspase-1 and promoting the release of interleukin (IL)-1β and IL-18. In this study, we investigated the neuroprotective potential of glucosamine (GlcN) in HD. Our results demonstrate that GlcN effectively attenuates lipopolysaccharide (LPS)/ATP-induced NLRP3 inflammasome activation in BV2 microglia, leading to a significant reduction in IL-1β and IL-18 secretion. Mechanistically, GlcN suppresses microglial activation by inhibiting the mitogen-activated protein kinase (MAPK) signaling pathway, thereby reducing nuclear factor-κB (NF-κB) activation. In the R6/2 transgenic mouse model of HD, oral administration of GlcN significantly enhanced neuronal survival, reduced mHTT aggregation, suppressed NLRP3 inflammasome activation, and attenuated astrocytic and microglial activation. Furthermore, GlcN improved motor performance and extended the lifespan of R6/2 mice. These findings suggest that GlcN confers neuroprotection in HD by attenuating neuroinflammation through inhibition of the NLRP3 inflammasome. Our study shows that GlcN is an effective treatment candidate for HD by targeting neuroinflammatory pathways, particularly through inhibition of the NLRP3 inflammasome, thereby presenting a promising strategy to slow disease progression.

亨廷顿氏病(HD)是一种遗传性神经退行性疾病,由亨廷顿蛋白(HTT)基因CAG重复扩增引起。它通常表现为进行性精神、认知和运动症状的三联征。由此产生的突变HTT (mHTT)蛋白破坏细胞稳态并促进神经炎症。NACHT、LRR和PYD结构域蛋白3 (NLRP3)炎性小体是神经炎症反应的关键介质,可激活caspase-1并促进白细胞介素(IL)-1β和IL-18的释放。在这项研究中,我们研究了葡萄糖胺(GlcN)对HD的神经保护作用。我们的研究结果表明,GlcN有效地减弱了BV2小胶质细胞中脂多糖(LPS)/ atp诱导的NLRP3炎性体激活,导致IL-1β和IL-18分泌显著减少。机制上,GlcN通过抑制丝裂原活化蛋白激酶(MAPK)信号通路抑制小胶质细胞的活化,从而降低核因子-κB (NF-κB)的活化。在R6/2转基因HD小鼠模型中,口服GlcN可显著提高神经元存活率,降低mHTT聚集,抑制NLRP3炎性体活化,减弱星形细胞和小胶质细胞活化。此外,GlcN还能改善R6/2小鼠的运动性能,延长其寿命。这些发现表明,GlcN通过抑制NLRP3炎性小体减轻神经炎症,从而在HD患者中提供神经保护。我们的研究表明,GlcN通过靶向神经炎症途径,特别是通过抑制NLRP3炎症小体,是一种有效的HD治疗候选药物,从而提供了一种有希望的减缓疾病进展的策略。
{"title":"Neuroprotective Effects of Glucosamine in Huntington's Disease Through NLRP3 Inflammasome Inhibition.","authors":"Lichieh Julie Chu, Yi-Ting Hsu, Yu Aoh, Chih-Yuan Cheng, Hui-Hsuan Wang, Ming-Tse Wu, Hao-Hsuan Li, Xiu-Yuan Lai, Kuo-Feng Hua, Chiung-Mei Chen, Tz-Chuen Ju","doi":"10.1007/s11481-025-10265-8","DOIUrl":"https://doi.org/10.1007/s11481-025-10265-8","url":null,"abstract":"<p><p>Huntington's disease (HD) is an inherited neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin (HTT) gene. It typically manifests as a triad of progressive psychiatric, cognitive, and motor symptoms. The resulting mutant HTT (mHTT) protein disrupts cellular homeostasis and promotes neuroinflammation. The NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome is a key mediator of neuroinflammatory responses, activating caspase-1 and promoting the release of interleukin (IL)-1β and IL-18. In this study, we investigated the neuroprotective potential of glucosamine (GlcN) in HD. Our results demonstrate that GlcN effectively attenuates lipopolysaccharide (LPS)/ATP-induced NLRP3 inflammasome activation in BV2 microglia, leading to a significant reduction in IL-1β and IL-18 secretion. Mechanistically, GlcN suppresses microglial activation by inhibiting the mitogen-activated protein kinase (MAPK) signaling pathway, thereby reducing nuclear factor-κB (NF-κB) activation. In the R6/2 transgenic mouse model of HD, oral administration of GlcN significantly enhanced neuronal survival, reduced mHTT aggregation, suppressed NLRP3 inflammasome activation, and attenuated astrocytic and microglial activation. Furthermore, GlcN improved motor performance and extended the lifespan of R6/2 mice. These findings suggest that GlcN confers neuroprotection in HD by attenuating neuroinflammation through inhibition of the NLRP3 inflammasome. Our study shows that GlcN is an effective treatment candidate for HD by targeting neuroinflammatory pathways, particularly through inhibition of the NLRP3 inflammasome, thereby presenting a promising strategy to slow disease progression.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"104"},"PeriodicalIF":3.5,"publicationDate":"2025-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145524778","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
Intravenous 64zn-Aspartate Mitigates Neuroinflammation and Motor Dysfunction in an Lps-Induced Parkinson's Disease Rat Model. 静脉注射64zn-天冬氨酸减轻脂多糖诱导的帕金森病大鼠模型的神经炎症和运动功能障碍
Max Temnik, Mariia Rudyk, Alexandr Balakin, Sergey Gurin, Taisa Dovbynchuk, Roman Byshovets, Nataliia Dzubenko, Ganna Tolstanova, Larysa Skivka

Emerging evidence links zinc dyshomeostasis to the pathogenesis of Parkinson's disease (PD), highlighting the need to explore zinc-based interventions. Zinc has five stable isotopes, with 64Zn and 66Zn being the most abundant. Notably, healthy brain tissue is enriched in the lighter isotope 64Zn, while heavier isotopes are hypothesized to accumulate with age. This study examined the therapeutic potential of intravenously administered isotopically enriched 64Zn aspartate (64Zn-asp) in a rat model of PD induced by a single stereotactic intranigral injection of lipopolysaccharide (LPS, 10 μg), which simulates acute neuroinflammation followed by progressive neurodegeneration. Treatment effects were evaluated using behavioral assessments, immunological profiling, biochemical and molecular analyses, and histopathology. Rats treated with 64Zn-asp showed a pronounced anti-inflammatory shift in microglial/macrophage metabolic profiles and reduced reactive astrogliosis. These changes were accompanied by improved motor performance and decreased anxiety-like behavior. Immunohistochemistry confirmed preservation of dopaminergic neurons. Overall, these findings suggest that 64Zn-asp attenuates neuroinflammation and supports neuronal survival, indicating its potential as a candidate for disease-modifying strategies in PD.

新出现的证据将锌平衡失调与帕金森病(PD)的发病机制联系起来,强调了探索锌基干预措施的必要性。锌有五种稳定的同位素,其中64Zn和66Zn最为丰富。值得注意的是,健康的脑组织富含较轻的同位素64Zn,而较重的同位素则随着年龄的增长而积累。本研究研究了静脉注射同位素富集的64Zn天冬氨酸(64Zn-asp)对单次立体定向内注射脂多糖(LPS, 10 μg)诱导的PD大鼠模型的治疗潜力,该模型模拟急性神经炎症后进行性神经退行性变。通过行为评估、免疫学分析、生化和分子分析以及组织病理学来评估治疗效果。用64Zn-asp处理的大鼠显示出明显的小胶质细胞/巨噬细胞代谢谱的抗炎变化和反应性星形胶质细胞增生的减少。这些变化伴随着运动表现的改善和焦虑样行为的减少。免疫组织化学证实了多巴胺能神经元的保存。总的来说,这些发现表明64Zn-asp可以减轻神经炎症并支持神经元存活,这表明它有可能成为PD疾病改善策略的候选药物。
{"title":"Intravenous <sup>64</sup>zn-Aspartate Mitigates Neuroinflammation and Motor Dysfunction in an Lps-Induced Parkinson's Disease Rat Model.","authors":"Max Temnik, Mariia Rudyk, Alexandr Balakin, Sergey Gurin, Taisa Dovbynchuk, Roman Byshovets, Nataliia Dzubenko, Ganna Tolstanova, Larysa Skivka","doi":"10.1007/s11481-025-10257-8","DOIUrl":"10.1007/s11481-025-10257-8","url":null,"abstract":"<p><p>Emerging evidence links zinc dyshomeostasis to the pathogenesis of Parkinson's disease (PD), highlighting the need to explore zinc-based interventions. Zinc has five stable isotopes, with <sup>64</sup>Zn and <sup>66</sup>Zn being the most abundant. Notably, healthy brain tissue is enriched in the lighter isotope <sup>64</sup>Zn, while heavier isotopes are hypothesized to accumulate with age. This study examined the therapeutic potential of intravenously administered isotopically enriched <sup>64</sup>Zn aspartate (<sup>64</sup>Zn-asp) in a rat model of PD induced by a single stereotactic intranigral injection of lipopolysaccharide (LPS, 10 μg), which simulates acute neuroinflammation followed by progressive neurodegeneration. Treatment effects were evaluated using behavioral assessments, immunological profiling, biochemical and molecular analyses, and histopathology. Rats treated with <sup>64</sup>Zn-asp showed a pronounced anti-inflammatory shift in microglial/macrophage metabolic profiles and reduced reactive astrogliosis. These changes were accompanied by improved motor performance and decreased anxiety-like behavior. Immunohistochemistry confirmed preservation of dopaminergic neurons. Overall, these findings suggest that <sup>64</sup>Zn-asp attenuates neuroinflammation and supports neuronal survival, indicating its potential as a candidate for disease-modifying strategies in PD.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"102"},"PeriodicalIF":3.5,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12612024/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145496289","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
Gallic Acid Alleviates Cerebral Ischemia-reperfusion Injury in Mice by Mediating Microglial Polarization Through the NLRP3/mTOR Axis. 没食子酸通过NLRP3/mTOR轴介导小胶质细胞极化减轻小鼠脑缺血再灌注损伤
Weilong Hua, Hongye Xu, Rundong Chen, Hongjian Zhang, Yongxin Zhang, Xiaoxi Zhang, Yongwei Zhang, Jianmin Liu, Lei Zhang, Pengfei Yang

Cerebral ischemia-reperfusion (I/R) injury is a critical condition leading to severe neurological deficits. Inflammation, driven by microglial polarization, plays a significant role in the progression of I/R injury. Gallic acid (GA), a natural polyphenol, has been recognized for its anti-inflammatory and neuroprotective properties. Male mice subjected to middle cerebral artery occlusion (MCAO) were treated with GA. Neurological deficits, infarct size, and brain edema were assessed to evaluate the neuroprotective effects of GA. In vitro, oxygen-glucose deprivation/reoxygenation (OGD/R) models were used to simulate I/R injury in microglial cells. The polarization of microglia was analyzed by flow cytometry, qPCR, and Western blot, focusing on M1 and M2 markers. Autophagy and inflammasome activation were investigated using Western blot, immunofluorescence, and flow cytometry, with the effects of GA modulated by autophagy and inflammasome inhibitors. GA treatment significantly improved neurological outcomes in MCAO mice by reducing infarct size, brain edema, and promoting the M2 polarization of microglia while inhibiting M1 polarization. GA enhanced autophagy and suppressed NLRP3 inflammasome activation via the mTOR pathway, reducing pro-inflammatory cytokine expression. Inhibition of autophagy reversed the protective effects of GA, leading to increased M1 polarization and exacerbated neuroinflammation. Additionally, activation of the NLRP3 inflammasome counteracted GA's effects, emphasizing the role of this pathway in microglial modulation. GA exerts neuroprotective effects in cerebral I/R injury by modulating microglial polarization through the NLRP3/mTOR axis. Its ability to promote autophagy and suppress inflammasome activation positions GA as a potential therapeutic agent for reducing neuroinflammation and improving outcomes in I/R injury.

脑缺血再灌注(I/R)损伤是导致严重神经功能缺损的一种危重疾病。由小胶质细胞极化驱动的炎症在I/R损伤的进展中起重要作用。没食子酸(GA)是一种天然多酚,具有抗炎和神经保护作用。用GA治疗大脑中动脉闭塞(MCAO)的雄性小鼠。评估神经功能缺损、梗死面积和脑水肿,以评估GA的神经保护作用。体外采用氧-葡萄糖剥夺/再氧化(OGD/R)模型模拟小胶质细胞I/R损伤。采用流式细胞术、qPCR和Western blot分析小胶质细胞的极化,重点关注M1和M2标记物。采用Western blot、免疫荧光和流式细胞术研究自噬和炎性小体的激活,并通过自噬和炎性小体抑制剂调节GA的作用。GA治疗通过减少梗死面积、脑水肿、促进小胶质细胞M2极化而抑制M1极化,显著改善MCAO小鼠的神经预后。GA通过mTOR途径增强自噬,抑制NLRP3炎性体活化,降低促炎细胞因子的表达。自噬抑制逆转了GA的保护作用,导致M1极化增加,神经炎症加重。此外,NLRP3炎性小体的激活抵消了GA的作用,强调了该途径在小胶质细胞调节中的作用。GA通过NLRP3/mTOR轴调节小胶质细胞极化,在脑I/R损伤中发挥神经保护作用。其促进自噬和抑制炎性体激活的能力使GA成为减少神经炎症和改善I/R损伤预后的潜在治疗剂。
{"title":"Gallic Acid Alleviates Cerebral Ischemia-reperfusion Injury in Mice by Mediating Microglial Polarization Through the NLRP3/mTOR Axis.","authors":"Weilong Hua, Hongye Xu, Rundong Chen, Hongjian Zhang, Yongxin Zhang, Xiaoxi Zhang, Yongwei Zhang, Jianmin Liu, Lei Zhang, Pengfei Yang","doi":"10.1007/s11481-025-10251-0","DOIUrl":"10.1007/s11481-025-10251-0","url":null,"abstract":"<p><p>Cerebral ischemia-reperfusion (I/R) injury is a critical condition leading to severe neurological deficits. Inflammation, driven by microglial polarization, plays a significant role in the progression of I/R injury. Gallic acid (GA), a natural polyphenol, has been recognized for its anti-inflammatory and neuroprotective properties. Male mice subjected to middle cerebral artery occlusion (MCAO) were treated with GA. Neurological deficits, infarct size, and brain edema were assessed to evaluate the neuroprotective effects of GA. In vitro, oxygen-glucose deprivation/reoxygenation (OGD/R) models were used to simulate I/R injury in microglial cells. The polarization of microglia was analyzed by flow cytometry, qPCR, and Western blot, focusing on M1 and M2 markers. Autophagy and inflammasome activation were investigated using Western blot, immunofluorescence, and flow cytometry, with the effects of GA modulated by autophagy and inflammasome inhibitors. GA treatment significantly improved neurological outcomes in MCAO mice by reducing infarct size, brain edema, and promoting the M2 polarization of microglia while inhibiting M1 polarization. GA enhanced autophagy and suppressed NLRP3 inflammasome activation via the mTOR pathway, reducing pro-inflammatory cytokine expression. Inhibition of autophagy reversed the protective effects of GA, leading to increased M1 polarization and exacerbated neuroinflammation. Additionally, activation of the NLRP3 inflammasome counteracted GA's effects, emphasizing the role of this pathway in microglial modulation. GA exerts neuroprotective effects in cerebral I/R injury by modulating microglial polarization through the NLRP3/mTOR axis. Its ability to promote autophagy and suppress inflammasome activation positions GA as a potential therapeutic agent for reducing neuroinflammation and improving outcomes in I/R injury.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":"20 1","pages":"101"},"PeriodicalIF":3.5,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145484209","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