首页 > 最新文献

Experimental Neurology最新文献

英文 中文
Intestinal dysbiosis causes spatial memory impairment in alcohol-exposed male mice by inducing neuroinflammation 肠道菌群失调通过诱发神经炎症导致酒精暴露雄性小鼠空间记忆受损
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-10-28 DOI: 10.1016/j.expneurol.2024.115028
Alcohol abuse damages the brain and triggers cognitive impairment. Intestinal dysbiosis has recently been shown to be involved in psychiatric disorders, which suggests the possibility of intestine-to-brain interactions in the development of alcohol abuse. In this study, chronic intermittent alcohol exposure (CIAE) model was established in C57BL/6 male mice and the spatial memory were detected by Barnes maze (n = 16/group). The fecal microbiota and its metabolites were detected by 16S rDNA sequencing and non-target liquid chromatograph mass spectrometer (LC-MS) (n = 8/group). Effects of alcohol on intestinal barrier and blood-brain barrier (BBB) permeability were detected by Evens blue leakage assay (n = 4/group), and the activation state of microglia and TLR4 expression were conducted by immunofluorescence co-localization (n = 4/group). The morphological changes of microglia were analyzed with Image J Analyze Skeleton software, and the protein levels of TLR4 and inflammatory factors were detected by Western Blot (n = 8/group). Results indicated that alcohol alters the components of fecal microbiota and metabolites, and damages the intestinal barrier and BBB, leading to spatial memory impairment in mice. By giving mice specific prebiotics (n = 16/group), we pointed out that increased endotoxin coming from Gram negative bacteria such as lipopolysaccharides (LPS) cross the BBB to activate microglia and inflammatory pathways in the prefrontal cortical (PFC) and hippocampus (HIP), releasing inflammatory factors and resulting in neuroinflammation. Thus, the fecal microbiota seems to be a potential target in the management of alcoholic brain disease.
酗酒会损害大脑并引发认知障碍。最近有研究表明,肠道菌群失调与精神疾病有关,这表明在酒精滥用的发展过程中可能存在肠道与大脑之间的相互作用。本研究以C57BL/6雄性小鼠为研究对象,建立了慢性间歇性酒精暴露(CIAE)模型,并通过巴恩斯迷宫检测了小鼠的空间记忆能力(n = 16/组)。通过 16S rDNA 测序和非目标液相色谱质谱仪(LC-MS)检测粪便微生物群及其代谢物(n = 8/组)。用伊文思蓝渗漏试验检测酒精对肠道屏障和血脑屏障(BBB)通透性的影响(4人/组),用免疫荧光共定位法检测小胶质细胞的活化状态和TLR4的表达(4人/组)。用 Image J Analyze Skeleton 软件分析小胶质细胞的形态学变化,用 Western Blot 检测 TLR4 和炎症因子的蛋白水平(n = 8/组)。结果表明,酒精会改变粪便微生物群的成分和代谢产物,破坏肠道屏障和 BBB,导致小鼠空间记忆受损。通过给小鼠服用特定的益生元(n = 16/组),我们指出,来自革兰氏阴性细菌的内毒素增加,如脂多糖(LPS)穿过BBB,激活前额叶皮质(PFC)和海马(HIP)的小胶质细胞和炎症通路,释放炎症因子,导致神经炎症。因此,粪便微生物群似乎是治疗酒精性脑病的一个潜在靶点。
{"title":"Intestinal dysbiosis causes spatial memory impairment in alcohol-exposed male mice by inducing neuroinflammation","authors":"","doi":"10.1016/j.expneurol.2024.115028","DOIUrl":"10.1016/j.expneurol.2024.115028","url":null,"abstract":"<div><div>Alcohol abuse damages the brain and triggers cognitive impairment. Intestinal dysbiosis has recently been shown to be involved in psychiatric disorders, which suggests the possibility of intestine-to-brain interactions in the development of alcohol abuse. In this study, chronic intermittent alcohol exposure (CIAE) model was established in C57BL/6 male mice and the spatial memory were detected by Barnes maze (<em>n</em> = 16/group). The fecal microbiota and its metabolites were detected by 16S rDNA sequencing and non-target liquid chromatograph mass spectrometer (LC-MS) (<em>n</em> = 8/group). Effects of alcohol on intestinal barrier and blood-brain barrier (BBB) permeability were detected by Evens blue leakage assay (<em>n</em> = 4/group), and the activation state of microglia and TLR4 expression were conducted by immunofluorescence co-localization (<em>n</em> = 4/group). The morphological changes of microglia were analyzed with Image J Analyze Skeleton software, and the protein levels of TLR4 and inflammatory factors were detected by Western Blot (<em>n</em> = 8/group). Results indicated that alcohol alters the components of fecal microbiota and metabolites, and damages the intestinal barrier and BBB, leading to spatial memory impairment in mice. By giving mice specific prebiotics (<em>n</em> = 16/group), we pointed out that increased endotoxin coming from Gram negative bacteria such as lipopolysaccharides (LPS) cross the BBB to activate microglia and inflammatory pathways in the prefrontal cortical (PFC) and hippocampus (HIP), releasing inflammatory factors and resulting in neuroinflammation. Thus, the fecal microbiota seems to be a potential target in the management of alcoholic brain disease.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142555013","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Gypenoside XLIX alleviates sepsis-associated encephalopathy by targeting PPAR-α Gypenoside XLIX 通过靶向 PPAR-α 减轻败血症相关脑病
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-10-28 DOI: 10.1016/j.expneurol.2024.115027
Sepsis-related systemic inflammation is a deadly condition with high rates of morbidity and mortality. There is evidence that sepsis affects the brain, and the most frequent organ dysfunction linked to sepsis is sepsis-associated encephalopathy. Sepsis-related brain damage can drastically reduce a patient's chances of survival. However, a specific treatment for sepsis-associated encephalopathy is not currently available. Consequently, to treat the brain damage caused by sepsis, investigating novel therapeutic strategies is imperative. After establishing the CLP-induced mouse SAE model, we treated the mice with Gyp-XLIX and evaluated apoptosis, neuroinflammation, brain damage, and oxidative stress in the brain tissue of each group of mice. Furthermore, the protective effects of Gyp-XLIX on LPS-treated BV-2 cells were assessed. We discovered that Gyp-XLIX treatment increased the survival rate of CLP-treated mice, alleviated SAE-related cerebral nerve abnormalities, and decreased blood–brain barrier breakdown, all of which could better preserve brain tissue in vivo. Furthermore, we identified associated proteins and found that Gyp-XLIX may reduce oxidative stress, cell apoptosis, and inflammation in the brain tissues of SAE mice. This observation was further validated in vitro. We established that Gyp-XLIX alleviates SAE by targeting PPAR-α. These findings may be important for the clinical applicability of Gyp-XLIX in SAE treatment. We found that Gyp-XLIX can alleviate brain injury in SAE by targeting PPAR-α and is a potential protective agent for SAE.
败血症相关的全身炎症是一种致命疾病,发病率和死亡率都很高。有证据表明,败血症会影响大脑,而与败血症有关的最常见器官功能障碍就是败血症相关性脑病。与败血症相关的脑损伤会大大降低患者的生存几率。然而,目前还没有治疗败血症相关性脑病的特效药。因此,要治疗败血症引起的脑损伤,研究新的治疗策略势在必行。在建立了 CLP 诱导的小鼠 SAE 模型后,我们用 Gyp-XLIX 对小鼠进行了治疗,并对各组小鼠脑组织中的细胞凋亡、神经炎症、脑损伤和氧化应激进行了评估。此外,还评估了 Gyp-XLIX 对经 LPS 处理的 BV-2 细胞的保护作用。我们发现,Gyp-XLIX 治疗提高了中毒性胰腺炎小鼠的存活率,缓解了与 SAE 相关的脑神经异常,并减少了血脑屏障的破坏,所有这些都能更好地保护体内脑组织。此外,我们还鉴定了相关蛋白,发现Gyp-XLIX可减少SAE小鼠脑组织中的氧化应激、细胞凋亡和炎症反应。这一观察结果在体外得到了进一步验证。我们证实,Gyp-XLIX 可通过靶向 PPAR-α 减轻 SAE。这些发现可能对 Gyp-XLIX 治疗 SAE 的临床应用具有重要意义。我们发现,Gyp-XLIX 可通过靶向 PPAR-α 减轻 SAE 的脑损伤,是一种潜在的 SAE 保护剂。
{"title":"Gypenoside XLIX alleviates sepsis-associated encephalopathy by targeting PPAR-α","authors":"","doi":"10.1016/j.expneurol.2024.115027","DOIUrl":"10.1016/j.expneurol.2024.115027","url":null,"abstract":"<div><div>Sepsis-related systemic inflammation is a deadly condition with high rates of morbidity and mortality. There is evidence that sepsis affects the brain, and the most frequent organ dysfunction linked to sepsis is sepsis-associated encephalopathy. Sepsis-related brain damage can drastically reduce a patient's chances of survival. However, a specific treatment for sepsis-associated encephalopathy is not currently available. Consequently, to treat the brain damage caused by sepsis, investigating novel therapeutic strategies is imperative. After establishing the CLP-induced mouse SAE model, we treated the mice with Gyp-XLIX and evaluated apoptosis, neuroinflammation, brain damage, and oxidative stress in the brain tissue of each group of mice. Furthermore, the protective effects of Gyp-XLIX on LPS-treated BV-2 cells were assessed. We discovered that Gyp-XLIX treatment increased the survival rate of CLP-treated mice, alleviated SAE-related cerebral nerve abnormalities, and decreased blood–brain barrier breakdown, all of which could better preserve brain tissue in vivo. Furthermore, we identified associated proteins and found that Gyp-XLIX may reduce oxidative stress, cell apoptosis, and inflammation in the brain tissues of SAE mice. This observation was further validated in vitro. We established that Gyp-XLIX alleviates SAE by targeting PPAR-α. These findings may be important for the clinical applicability of Gyp-XLIX in SAE treatment. We found that Gyp-XLIX can alleviate brain injury in SAE by targeting PPAR-α and is a potential protective agent for SAE.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142555014","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Zebrafish as a model organism to study sporadic Alzheimer's disease: Behavioural, biochemical and histological validation 斑马鱼作为研究散发性阿尔茨海默病的模式生物:行为、生化和组织学验证。
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-10-26 DOI: 10.1016/j.expneurol.2024.115034
Alzheimer's disease (AD) is a global burden to the healthcare system with no viable treatment options till date. Rodents and primates have been extensively used as models for understanding AD pathogenesis and identifying therapeutic targets. However, the focus is now shifting towards developing alternate models. Zebrafish is emerging as a preferred model for neurodegenerative conditions because of its simple nervous system, highly conserved genome and short duration required to model disease condition. The present study is aimed to develop streptozotocin (STZ)-induced model of sporadic AD (sAD) in zebrafish. STZ was administered to adult zebrafish (4–6 mo) at different doses (1 to 50 mg/kg body weight, intracerebroventricularly). Kaplan-Meier survival analysis revealed time and dose dependent mortality in the zebrafish administered with STZ. Based on survival analysis, 1 to 10 mg/kg body weight of STZ was selected for behavioural, molecular and histological studies. STZ administered fish had anxiety-like and stress behaviour in novel tank and light/dark preference tests. STZ-induced cognitive and memory deficits assessed using novel object recognition and spatial alternation tests. Further, expression of markers of amyloidogenic pathway (appa and bace1) were increased in terms of mRNA and protein levels in a time and dose dependent manner following STZ administration. However, expression of non-amyloidogenic pathway mediator (adam10) was reduced at both mRNA and protein level. Histological assessment using hematoxylin and eosin, and Nissl stain revealed loss of neurons in STZ administered fish. The ratio of phosphor-tauser396/total-tau was increased in STZ administered fish. Based on these findings, 5 mg/kg body weight of STZ was found to be most appropriate dose to exhibit sAD phenotype. Mass spectrometric analysis confirmed the presence of amyloid beta oligomers in brains of STZ administered fish. Transmission electron microscopy also showed the presence of higher order insoluble amyloid fibrils with twists. Immunohistochemical analysis revealed amyloid beta deposits in brain of STZ administered fish. Golgi-cox staining indicated decreased number of dendrites, whereas microglia had increased density, span ratio, soma area and lacunarity. The results of the present study demonstrate presence of AD hallmarks and phenotype in zebrafish 7 days post STZ administration (5 mg/kg). The study validates the potential of STZ-induced sAD in zebrafish as a reliable model for studying pathophysiology and rapid screening of therapeutic molecules against sAD.
阿尔茨海默病(AD)是全球医疗系统的一大负担,迄今为止还没有可行的治疗方案。啮齿类动物和灵长类动物被广泛用作了解阿尔茨海默病发病机制和确定治疗靶点的模型。然而,现在的重点正在转向开发替代模型。斑马鱼因其简单的神经系统、高度保守的基因组和较短的建模时间,正逐渐成为神经退行性疾病的首选模型。本研究旨在开发链脲佐菌素(STZ)诱导的斑马鱼散发性注意力缺失症(sAD)模型。研究人员给成年斑马鱼(4-6 个月)注射不同剂量的 STZ(1 至 50 毫克/千克体重,脑室内注射)。卡普兰-米尔存活率分析显示,使用 STZ 的斑马鱼死亡率与时间和剂量有关。根据存活率分析,选择每公斤体重 1 至 10 毫克的 STZ 进行行为、分子和组织学研究。使用 STZ 的斑马鱼在新鱼缸和光/暗偏好测试中表现出焦虑和应激样行为。使用新物体识别和空间交替测试评估了 STZ 诱导的认知和记忆缺陷。此外,STZ 施用后,淀粉样蛋白生成途径的标记物(appa 和 bace1)的 mRNA 和蛋白质水平的表达增加,且呈时间和剂量依赖性。然而,非淀粉样蛋白生成途径介质(adam10)在mRNA和蛋白质水平上的表达均有所降低。使用苏木精、伊红和 Nissl 染色法进行的组织学评估显示,STZ 给药的鱼体内神经元减少。在 STZ 施用的鱼体内,磷-tauser396/总-tau 的比率增加。基于这些发现,5 毫克/千克体重的 STZ 被认为是表现出 sAD 表型的最合适剂量。质谱分析证实了 STZ 给药鱼的大脑中存在淀粉样 beta 低聚物。透射电子显微镜也显示了高阶不溶性淀粉样蛋白纤维的存在。免疫组化分析显示,STZ 给药鱼的大脑中有淀粉样 beta 沉积。高尔基体-柯克斯染色表明树突数量减少,而小胶质细胞的密度、跨度比、体节面积和裂隙度增加。本研究结果表明,STZ 给药(5 毫克/千克)7 天后,斑马鱼出现了注意力缺失症的特征和表型。这项研究验证了 STZ 诱导的 sAD 在斑马鱼中的潜力,它是研究病理生理学和快速筛选 sAD 治疗分子的可靠模型。
{"title":"Zebrafish as a model organism to study sporadic Alzheimer's disease: Behavioural, biochemical and histological validation","authors":"","doi":"10.1016/j.expneurol.2024.115034","DOIUrl":"10.1016/j.expneurol.2024.115034","url":null,"abstract":"<div><div>Alzheimer's disease (AD) is a global burden to the healthcare system with no viable treatment options till date. Rodents and primates have been extensively used as models for understanding AD pathogenesis and identifying therapeutic targets. However, the focus is now shifting towards developing alternate models. Zebrafish is emerging as a preferred model for neurodegenerative conditions because of its simple nervous system, highly conserved genome and short duration required to model disease condition. The present study is aimed to develop streptozotocin (STZ)-induced model of sporadic AD (sAD) in zebrafish. STZ was administered to adult zebrafish (4–6 mo) at different doses (1 to 50 mg/kg body weight, intracerebroventricularly). Kaplan-Meier survival analysis revealed time and dose dependent mortality in the zebrafish administered with STZ. Based on survival analysis, 1 to 10 mg/kg body weight of STZ was selected for behavioural, molecular and histological studies. STZ administered fish had anxiety-like and stress behaviour in novel tank and light/dark preference tests. STZ-induced cognitive and memory deficits assessed using novel object recognition and spatial alternation tests. Further, expression of markers of amyloidogenic pathway (<em>appa</em> and <em>bace1</em>) were increased in terms of mRNA and protein levels in a time and dose dependent manner following STZ administration. However, expression of non-amyloidogenic pathway mediator (<em>adam10</em>) was reduced at both mRNA and protein level. Histological assessment using hematoxylin and eosin, and Nissl stain revealed loss of neurons in STZ administered fish. The ratio of phosphor-tau<sup>ser396</sup>/total-tau was increased in STZ administered fish. Based on these findings, 5 mg/kg body weight of STZ was found to be most appropriate dose to exhibit sAD phenotype. Mass spectrometric analysis confirmed the presence of amyloid beta oligomers in brains of STZ administered fish. Transmission electron microscopy also showed the presence of higher order insoluble amyloid fibrils with twists. Immunohistochemical analysis revealed amyloid beta deposits in brain of STZ administered fish. Golgi-cox staining indicated decreased number of dendrites, whereas microglia had increased density, span ratio, soma area and lacunarity. The results of the present study demonstrate presence of AD hallmarks and phenotype in zebrafish 7 days post STZ administration (5 mg/kg). The study validates the potential of STZ-induced sAD in zebrafish as a reliable model for studying pathophysiology and rapid screening of therapeutic molecules against sAD.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-10-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142566900","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GSK-3β inhibitor amplifies autophagy-lysosomal pathways by regulating TFEB in Parkinson's disease models 在帕金森病模型中,GSK-3β抑制剂通过调节TFEB扩大自噬-溶酶体途径。
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-10-26 DOI: 10.1016/j.expneurol.2024.115033
Parkinson's disease (PD), a common neurodegenerative disorder characterized by degeneration of the substantia nigra and a marked increase in Lewy bodies in the brain, primarily manifests as motor dysfunction. Glycogen synthase kinase-3 beta (GSK-3β) is known to play a critical role in various pathological processes of neurodegenerative diseases. However, the impact of GSK-3β inhibitors on PD progression and the underlying molecular mechanisms responsible for the effects have not been fully elucidated. Using in vitro and mouse models of 1-methyl-4-phenylpyridine (MPP+)-or methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD, we found that inhibition of GSK-3β activity alleviated mitochondrial damage, cell apoptosis, and neuronal cell loss by promoting the nuclear translocation of transcription factor EB (TFEB), thereby amplifying the autophagy-lysosomal pathway (ALP). Importantly, siRNA silencing of the TFEB gene impaired the GSK-3β inhibitor-mediated activation of the ALP pathway, thus negating the metabolic support required for neuronal functional improvement. Short-term treatment with the GSK-3β inhibitor significantly ameliorated motor dysfunction and improved motor coordination in model mice with MPTP-induced PD. GSK-3β inhibition increased the ALP and TFEB activities in the mice, thereby reducing α-synuclein aggregation and neuronal damage. In conclusion, our study demonstrates that inhibition of GSK-3β activity can delay the pathological processes of PD via promotion of the TFEB–ALP pathway, potentially providing a novel therapeutic target for this neurodegenerative disorder.
帕金森病(Parkinson's disease,PD)是一种常见的神经退行性疾病,以黑质变性和脑内路易体明显增多为特征,主要表现为运动功能障碍。众所周知,糖原合酶激酶-3β(GSK-3β)在神经退行性疾病的各种病理过程中发挥着关键作用。然而,GSK-3β抑制剂对帕金森病进展的影响以及导致这种影响的潜在分子机制尚未完全阐明。我们利用体外和小鼠模型研究了1-甲基-4-苯基吡啶(MPP+)-或甲基-4-苯基-1,2,3,6-四氢吡啶(MPTP)-诱导的帕金森病,发现抑制GSK-3β活性可通过促进转录因子EB(TFEB)的核转位,从而扩大自噬-溶酶体途径(ALP),减轻线粒体损伤、细胞凋亡和神经细胞丢失。重要的是,siRNA 沉默 TFEB 基因会损害 GSK-3β 抑制剂介导的 ALP 通路激活,从而抵消神经元功能改善所需的代谢支持。GSK-3β抑制剂的短期治疗显著改善了运动功能障碍,并改善了MPTP诱导的帕金森病模型小鼠的运动协调性。GSK-3β抑制剂可提高小鼠的ALP和TFEB活性,从而减少α-突触核蛋白聚集和神经元损伤。总之,我们的研究表明,抑制GSK-3β的活性可通过促进TFEB-ALP通路延缓帕金森病的病理过程,从而有可能为这种神经退行性疾病提供一种新的治疗靶点。
{"title":"GSK-3β inhibitor amplifies autophagy-lysosomal pathways by regulating TFEB in Parkinson's disease models","authors":"","doi":"10.1016/j.expneurol.2024.115033","DOIUrl":"10.1016/j.expneurol.2024.115033","url":null,"abstract":"<div><div>Parkinson's disease (PD), a common neurodegenerative disorder characterized by degeneration of the substantia nigra and a marked increase in Lewy bodies in the brain, primarily manifests as motor dysfunction. Glycogen synthase kinase-3 beta (GSK-3β) is known to play a critical role in various pathological processes of neurodegenerative diseases. However, the impact of GSK-3β inhibitors on PD progression and the underlying molecular mechanisms responsible for the effects have not been fully elucidated. Using in vitro and mouse models of 1-methyl-4-phenylpyridine (MPP<sup>+</sup>)-or methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD, we found that inhibition of GSK-3β activity alleviated mitochondrial damage, cell apoptosis, and neuronal cell loss by promoting the nuclear translocation of transcription factor EB (TFEB), thereby amplifying the autophagy-lysosomal pathway (ALP). Importantly, siRNA silencing of the TFEB gene impaired the GSK-3β inhibitor-mediated activation of the ALP pathway, thus negating the metabolic support required for neuronal functional improvement. Short-term treatment with the GSK-3β inhibitor significantly ameliorated motor dysfunction and improved motor coordination in model mice with MPTP-induced PD. GSK-3β inhibition increased the ALP and TFEB activities in the mice, thereby reducing α-synuclein aggregation and neuronal damage. In conclusion, our study demonstrates that inhibition of GSK-3β activity can delay the pathological processes of PD via promotion of the TFEB–ALP pathway, potentially providing a novel therapeutic target for this neurodegenerative disorder.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-10-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142566807","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
TREM1 induces microglial ferroptosis through the PERK pathway in diabetic-associated cognitive impairment 在糖尿病相关认知障碍中,TREM1 通过 PERK 通路诱导小胶质细胞铁突变。
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-10-25 DOI: 10.1016/j.expneurol.2024.115031
Ferroptosis is involved in neurodegenerative disorders including diabetes-associated cognitive impairment (DACI). As central immune cells, microglia have strong siderophilic properties. However, the role of iron deposition in microglia and the underlying regulatory mechanism remains unclear in DACI. Here, we established high glucose (HG) model in BV2/HMC3 cells and diabetes model in C57BL/6 J mice with HFD and STZ. Transmission Electron Microscopy, Western blot, assay kits of Fe2+, GSH/GSSG, MDA and ROS were carried out in vitro. Prussian blue staining, Western blot and immunofluorescence were implemented in vivo. Y-maze and novel object recognition were performed to assess cognitive performance. LP17 was used to inhibit TREM1 (triggering receptor expressed on myeloid cells 1) specifically in vivo and vitro. We found excessively deposited iron and significant reduction in antioxidants in hippocampal microglia of mice with DACI, concomitant with increased TREM1 (a microglia-specific inflammatory amplifier). Furthermore, LP17 (TREM1 specific inhibitor) ameliorated cognitive impairment caused by HFD/STZ through relieving iron accumulation and antioxidant inactivation. In vitro, ferroptosis was induced by HG in mice microglia-BV2 and human microglia-HMC3 cells, which could be blocked by a ferroptosis inhibitor-Fer-1 and LP17. Moreover, PERK pathway of endoplasmic reticulum stress was activated by HG, and then reversed by PERK inhibitor GSK2606414 and LP17 followed by improved ferroptosis in HG-cultured BV2. In summary, our results indicated that TREM1 effectively aggravates T2DM-associated microglial iron accumulation through the PERK pathway of ERS, which contributes to antioxidant inactivation and lipid peroxidation, eventually, massively boosted ROS result in microglial ferroptosis. The mechanism elucidation in our study may shed light on targeted therapy of DACI.
铁蛋白沉积与神经退行性疾病有关,包括糖尿病相关认知障碍(DACI)。作为中枢免疫细胞,小胶质细胞具有很强的嗜铁特性。然而,铁沉积在小胶质细胞中的作用以及在 DACI 中的潜在调控机制仍不清楚。在此,我们在 BV2/HMC3 细胞中建立了高糖(HG)模型,并在 C57BL/6 J 小鼠中建立了 HFD 和 STZ 糖尿病模型。在体外进行了透射电子显微镜、Western blot、Fe2+、GSH/GSSG、MDA 和 ROS 检测。在体内进行了普鲁士蓝染色、Western印迹和免疫荧光。进行了Y-迷宫和新物体识别以评估认知能力。LP17 被用于抑制体内和体外的 TREM1(髓样细胞上表达的触发受体 1)。我们发现,在DACI小鼠的海马小胶质细胞中,铁沉积过多,抗氧化剂显著减少,同时TREM1(一种小胶质细胞特异性炎症放大器)增加。此外,LP17(TREM1特异性抑制剂)通过缓解铁积累和抗氧化剂失活,改善了HFD/STZ导致的认知障碍。在体外,HG诱导小鼠小胶质细胞-BV2和人小胶质细胞-HMC3细胞发生铁变态反应,铁变态反应抑制剂-Fer-1和LP17可阻断这种反应。此外,内质网应激的PERK通路被HG激活,然后被PERK抑制剂GSK2606414和LP17逆转,随后HG培养的BV2的铁突变得到改善。综上所述,我们的研究结果表明,TREM1通过ERS的PERK通路有效地加重了T2DM相关的小胶质细胞铁蓄积,从而导致抗氧化剂失活和脂质过氧化,最终,ROS的大量增加导致小胶质细胞铁嗜酸性化。我们的研究对这一机制的阐明或许能为DACI的靶向治疗提供启示。
{"title":"TREM1 induces microglial ferroptosis through the PERK pathway in diabetic-associated cognitive impairment","authors":"","doi":"10.1016/j.expneurol.2024.115031","DOIUrl":"10.1016/j.expneurol.2024.115031","url":null,"abstract":"<div><div>Ferroptosis is involved in neurodegenerative disorders including diabetes-associated cognitive impairment (DACI). As central immune cells, microglia have strong siderophilic properties. However, the role of iron deposition in microglia and the underlying regulatory mechanism remains unclear in DACI. Here, we established high glucose (HG) model in BV2/HMC3 cells and diabetes model in C57BL/6 J mice with HFD and STZ. Transmission Electron Microscopy, Western blot, assay kits of Fe<sup>2+</sup>, GSH/GSSG, MDA and ROS were carried out <em>in vitro</em>. Prussian blue staining, Western blot and immunofluorescence were implemented <em>in vivo</em>. Y-maze and novel object recognition were performed to assess cognitive performance. LP17 was used to inhibit TREM1 (triggering receptor expressed on myeloid cells 1) specifically <em>in vivo and vitro</em>. We found excessively deposited iron and significant reduction in antioxidants in hippocampal microglia of mice with DACI, concomitant with increased TREM1 (a microglia-specific inflammatory amplifier). Furthermore, LP17 (TREM1 specific inhibitor) ameliorated cognitive impairment caused by HFD/STZ through relieving iron accumulation and antioxidant inactivation. <em>In vitro</em>, ferroptosis was induced by HG in mice microglia-BV2 and human microglia-HMC3 cells, which could be blocked by a ferroptosis inhibitor-Fer-1 and LP17. Moreover, PERK pathway of endoplasmic reticulum stress was activated by HG, and then reversed by PERK inhibitor GSK2606414 and LP17 followed by improved ferroptosis in HG-cultured BV2. In summary, our results indicated that TREM1 effectively aggravates T2DM-associated microglial iron accumulation through the PERK pathway of ERS, which contributes to antioxidant inactivation and lipid peroxidation, eventually, massively boosted ROS result in microglial ferroptosis. The mechanism elucidation in our study may shed light on targeted therapy of DACI.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-10-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142497765","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Rotating magnetic field improves cognitive and memory impairments in APP/PS1 mice by activating autophagy and inhibiting the PI3K/AKT/mTOR signaling pathway 旋转磁场通过激活自噬和抑制 PI3K/AKT/mTOR 信号通路,改善 APP/PS1 小鼠的认知和记忆障碍。
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-10-25 DOI: 10.1016/j.expneurol.2024.115029
Alzheimer's disease (AD) is a geriatric disorder that can be roughly classified into sporadic AD and hereditary AD. The latter is strongly associated with genetic factors, and its treatment poses greater challenges compared to sporadic AD. Rotating magnetic fields (RMF) is a non-invasive treatment known to have diverse biological effects, including the modulation of the central nervous system and aging. However, the impact of RMF on hereditary AD and its underlying mechanism remain unexplored. In this study, we exposed APP/PS1 mice to RMF (2 h/day, 0.2 T, 4 Hz) for a duration of 6 months. The results demonstrated that RMF treatment significantly ameliorated their cognitive and memory impairments, attenuated neuronal damage, and reduced amyloid deposition. Furthermore, RNA-sequencing analysis revealed a significant enrichment of autophagy-related genes and the PI3K/AKT-mTOR signaling pathway. Western blotting further confirmed that RMF activated autophagy and suppressed the phosphorylation of proteins associated with the PI3K/AKT/mTOR signaling pathway in APP/PS1 mice. These protective effects and the underlying mechanism were also observed in Aβ2535-exposed HT22 cells. Collectively, our findings indicate that RMF improves cognitive and memory dysfunction in APP/PS1 mice by activating autophagy and inhibiting the PI3K/AKT/mTOR signaling pathway, thus highlighting the potential of RMF as a clinical treatment for hereditary AD.
阿尔茨海默病(AD)是一种老年性疾病,大致可分为散发性阿尔茨海默病和遗传性阿尔茨海默病。遗传性阿尔茨海默病与遗传因素密切相关,与散发性阿尔茨海默病相比,遗传性阿尔茨海默病的治疗面临更大的挑战。众所周知,旋转磁场(RMF)是一种非侵入性治疗方法,具有多种生物效应,包括调节中枢神经系统和衰老。然而,RMF 对遗传性 AD 的影响及其内在机制仍有待探索。在本研究中,我们将 APP/PS1 小鼠暴露于 RMF(2 小时/天,0.2 T,4 Hz),为期 6 个月。结果表明,RMF 治疗显著改善了小鼠的认知和记忆障碍,减轻了神经元损伤,减少了淀粉样蛋白沉积。此外,RNA序列分析显示,自噬相关基因和PI3K/AKT-mTOR信号通路显著富集。Western blotting进一步证实,在APP/PS1小鼠体内,RMF激活了自噬,并抑制了与PI3K/AKT/mTOR信号通路相关的蛋白质磷酸化。在暴露于Aβ25-35的HT22细胞中也观察到了这些保护作用及其机制。总之,我们的研究结果表明,RMF能通过激活自噬和抑制PI3K/AKT/mTOR信号通路来改善APP/PS1小鼠的认知和记忆功能障碍,从而凸显了RMF作为遗传性AD临床治疗方法的潜力。
{"title":"Rotating magnetic field improves cognitive and memory impairments in APP/PS1 mice by activating autophagy and inhibiting the PI3K/AKT/mTOR signaling pathway","authors":"","doi":"10.1016/j.expneurol.2024.115029","DOIUrl":"10.1016/j.expneurol.2024.115029","url":null,"abstract":"<div><div>Alzheimer's disease (AD) is a geriatric disorder that can be roughly classified into sporadic AD and hereditary AD. The latter is strongly associated with genetic factors, and its treatment poses greater challenges compared to sporadic AD. Rotating magnetic fields (RMF) is a non-invasive treatment known to have diverse biological effects, including the modulation of the central nervous system and aging. However, the impact of RMF on hereditary AD and its underlying mechanism remain unexplored. In this study, we exposed APP/PS1 mice to RMF (2 h/day, 0.2 T, 4 Hz) for a duration of 6 months. The results demonstrated that RMF treatment significantly ameliorated their cognitive and memory impairments, attenuated neuronal damage, and reduced amyloid deposition. Furthermore, RNA-sequencing analysis revealed a significant enrichment of autophagy-related genes and the PI3K/AKT-mTOR signaling pathway. Western blotting further confirmed that RMF activated autophagy and suppressed the phosphorylation of proteins associated with the PI3K/AKT/mTOR signaling pathway in APP/PS1 mice. These protective effects and the underlying mechanism were also observed in Aβ<sub>25</sub><sub>–</sub><sub>35</sub>-exposed HT22 cells. Collectively, our findings indicate that RMF improves cognitive and memory dysfunction in APP/PS1 mice by activating autophagy and inhibiting the PI3K/AKT/mTOR signaling pathway, thus highlighting the potential of RMF as a clinical treatment for hereditary AD.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-10-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142497763","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Combination treatment with cilostazol and isosorbide mononitrate attenuates microemboli-mediated vascular cognitive impairment and improves imaging and plasma biomarkers in diabetic rats 西洛他唑和单硝酸异山梨酯联合治疗可减轻微栓子介导的血管认知障碍,并改善糖尿病大鼠的成像和血浆生物标志物
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-10-25 DOI: 10.1016/j.expneurol.2024.115030
Diabetes is a major risk factor for all types of dementia. The underlying reasons are not fully understood, and preventive therapeutic strategies are lacking. Previously we have shown that diabetic but not control rats developed a progressive cognitive decline in a microemboli (ME) model of vascular contributions to cognitive impairment & dementia (VCID). Given the cerebrovascular dysfunction is a mutual pathological change between diabetes and VCID, we hypothesized that the cognitive impairment in this ME model can be prevented by improving the endothelial function in diabetes. Our treatment paradigm was based on the LACI-2 Trial which assessed the efficacy of isosorbide mononitrate (ISMN) and cilostazol (Cil) treatments in small vessel disease progression. Control and diabetic rats were treated with ISMN/Cil or vehicle for 4 weeks, then injected with cholesterol crystal ME and the behavioral outcomes were monitored. Brain microstructure integrity was assessed by diffusion MRI. Plasma biomarkers were assessed using angiogenesis, neurology and amyloid β 42/40 panels recommended by the MarkVCID consortium. Behavioral deficits and the loss of tissue integrity previously observed in untreated diabetic rats were not noted in the treated animals in this study. Treatment improved tissue perfusion but there were no differences in plasma biomarkers. These results suggest that restoration of endothelial function with ISMN/Cil before ME injection prevented the possible deleterious effects of ME in diabetic rats by improving the endothelial integrity and it is a practical preventive and therapeutic strategy for VCID.
糖尿病是所有类型痴呆症的主要风险因素。其根本原因尚不完全清楚,也缺乏预防性治疗策略。此前,我们已经证明,在微栓子(ME)认知障碍& 痴呆(VCID)的血管作用模型中,糖尿病大鼠而非对照组大鼠会出现进行性认知功能下降。鉴于脑血管功能障碍是糖尿病和 VCID 之间的共同病理变化,我们假设可以通过改善糖尿病大鼠的内皮功能来预防 ME 模型中的认知功能障碍。我们的治疗范式基于 LACI-2 试验,该试验评估了单硝酸异山梨酯(ISMN)和西洛他唑(Cil)治疗小血管疾病进展的疗效。对照组和糖尿病大鼠接受 ISMN/Cil 或药物治疗 4 周,然后注射胆固醇晶体 ME 并监测行为结果。通过弥散核磁共振成像评估大脑微观结构的完整性。血浆生物标志物采用 MarkVCID 联盟推荐的血管生成、神经学和淀粉样β 42/40 面板进行评估。在本研究中,接受治疗的动物没有出现之前在未经治疗的糖尿病大鼠身上观察到的行为障碍和组织完整性丧失。治疗改善了组织灌注,但血浆生物标志物并无差异。这些结果表明,在注射ME前用ISMN/Cil恢复内皮功能可通过改善内皮完整性来防止ME对糖尿病大鼠可能产生的有害影响,是一种实用的VCID预防和治疗策略。
{"title":"Combination treatment with cilostazol and isosorbide mononitrate attenuates microemboli-mediated vascular cognitive impairment and improves imaging and plasma biomarkers in diabetic rats","authors":"","doi":"10.1016/j.expneurol.2024.115030","DOIUrl":"10.1016/j.expneurol.2024.115030","url":null,"abstract":"<div><div>Diabetes is a major risk factor for all types of dementia. The underlying reasons are not fully understood, and preventive therapeutic strategies are lacking. Previously we have shown that diabetic but not control rats developed a progressive cognitive decline in a microemboli (ME) model of vascular contributions to cognitive impairment &amp; dementia (VCID). Given the cerebrovascular dysfunction is a mutual pathological change between diabetes and VCID, we hypothesized that the cognitive impairment in this ME model can be prevented by improving the endothelial function in diabetes. Our treatment paradigm was based on the LACI-2 Trial which assessed the efficacy of isosorbide mononitrate (ISMN) and cilostazol (Cil) treatments in small vessel disease progression. Control and diabetic rats were treated with ISMN/Cil or vehicle for 4 weeks, then injected with cholesterol crystal ME and the behavioral outcomes were monitored. Brain microstructure integrity was assessed by diffusion MRI. Plasma biomarkers were assessed using angiogenesis, neurology and amyloid β 42/40 panels recommended by the MarkVCID consortium. Behavioral deficits and the loss of tissue integrity previously observed in untreated diabetic rats were not noted in the treated animals in this study. Treatment improved tissue perfusion but there were no differences in plasma biomarkers. These results suggest that restoration of endothelial function with ISMN/Cil before ME injection prevented the possible deleterious effects of ME in diabetic rats by improving the endothelial integrity and it is a practical preventive and therapeutic strategy for VCID.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-10-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142554999","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Border-associated macrophages: From physiology to therapeutic targets in Alzheimer's disease 边界相关巨噬细胞:从生理学到阿尔茨海默病的治疗靶点
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-10-24 DOI: 10.1016/j.expneurol.2024.115021
Border-associated macrophages (BAMs) constitute a highly heterogeneous group of central nervous system-resident macrophages at the brain boundaries. Despite their significance, BAMs have mainly been overlooked compared to microglia, resulting in a limited understanding of their functions. However, recent advancements in single-cell immunophenotyping and transcriptomic analyses of BAMs have revealed a previously unrecognized complexity in these cells, in addition to their critical roles under non-pathological conditions and diseases like Alzheimer's disease (AD), Parkinson's disease, glioma, and ischemic stroke. In this review, we discuss the origins, self-renewal capabilities, and extensive heterogeneity of BAMs, and clarify their important physiological functions such as immune monitoring, waste removal and vascular permeability regulation. We also summarize experimental evidence linking BAMs to the progression of AD. Finally, we review therapeutic strategies targeting brain innate immune cells mainly focusing on strategies aimed at modulating BAMs to treat AD and evaluate their potential in clinical applications.
边界相关巨噬细胞(BAMs)是大脑边界中枢神经系统驻留巨噬细胞的一个高度异质性群体。尽管边界相关巨噬细胞意义重大,但与小胶质细胞相比,它们主要被忽视,导致人们对其功能的了解有限。然而,最近在单细胞免疫分型和 BAMs 转录组学分析方面取得的进展揭示了这些细胞以前未曾认识到的复杂性,以及它们在非病理状态和疾病(如阿尔茨海默病(AD)、帕金森病、胶质瘤和缺血性中风)中的关键作用。在这篇综述中,我们讨论了 BAMs 的起源、自我更新能力和广泛的异质性,并阐明了它们的重要生理功能,如免疫监测、废物清除和血管通透性调节。我们还总结了将 BAMs 与 AD 进展联系起来的实验证据。最后,我们回顾了针对脑先天性免疫细胞的治疗策略,主要侧重于旨在调节 BAMs 以治疗 AD 的策略,并评估其在临床应用中的潜力。
{"title":"Border-associated macrophages: From physiology to therapeutic targets in Alzheimer's disease","authors":"","doi":"10.1016/j.expneurol.2024.115021","DOIUrl":"10.1016/j.expneurol.2024.115021","url":null,"abstract":"<div><div>Border-associated macrophages (BAMs) constitute a highly heterogeneous group of central nervous system-resident macrophages at the brain boundaries. Despite their significance, BAMs have mainly been overlooked compared to microglia, resulting in a limited understanding of their functions. However, recent advancements in single-cell immunophenotyping and transcriptomic analyses of BAMs have revealed a previously unrecognized complexity in these cells, in addition to their critical roles under non-pathological conditions and diseases like Alzheimer's disease (AD), Parkinson's disease, glioma, and ischemic stroke. In this review, we discuss the origins, self-renewal capabilities, and extensive heterogeneity of BAMs, and clarify their important physiological functions such as immune monitoring, waste removal and vascular permeability regulation. We also summarize experimental evidence linking BAMs to the progression of AD. Finally, we review therapeutic strategies targeting brain innate immune cells mainly focusing on strategies aimed at modulating BAMs to treat AD and evaluate their potential in clinical applications.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142497760","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Effect of vildagliptin conjugated monometallic nanoparticles and bimetallic nanocomposites on diabetes-induced cognitive deficit 维达列汀共轭单金属纳米颗粒和双金属纳米复合材料对糖尿病引起的认知缺陷的影响
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-10-24 DOI: 10.1016/j.expneurol.2024.115026
Oxidative stress is one of the major causes of different metabolic disorders, including diabetes, cardiovascular diseases, neurodegenerative diseases and cancers. Some metabolic disorders like diabetes mellitus leads to secondary complications after micro and macrovascular complications. Some of the most prevalent neurodegenerative diseases, like cognitive impairment and Alzheimer's disease, are found in chronic diabetic patients. The present study is designed to understand the mechanism of interconnection between diabetes mellitus and cognitive deficit using the alloxan model of diabetes-induced cognitive deficit in the rat model. The alloxan monohydrate produces reactive oxygen species, producing superoxide free radicals, hydrogen peroxide and hydroxyl radicals. The hydroxyl radicals ultimately cause the death of beta cells, causing diabetes. Hence, the correlation of oxidative stress and neurodegeneration in cognitive impairment is the trigger for this study. In the present study, we investigate the ameliorative effect of vildagliptin (VLD) and its conjugated nanoparticles against alloxan-associated brain damage due to oxidative stress. The gold (Au), selenium (Se) nanoparticles, and bimetallic (Se@Au) nanocomposites of VLD are synthesized and assessed for improvement in their brain availability. The in-vitro antioxidant evaluation of the VLD and nanoparticles is done using DPPH, ABTS, and FRAP assay. The memory-related neurobehavioral studies, in-vivo antioxidant studies, in-vivo biochemical studies, and histopathological examinations are evaluated in rat brains. The VLD and its nanoformulations exhibited in-vitro and in-vivo antioxidant properties significantly (p < 0.01). They reduced the activity of AChE and nitrite in the alloxan diabetic rats. The bimetallic Se@Au VLDNCs displayed a more protective effect than VLD, VLD–AuNPs, and VLD–SeNPs.
氧化应激是导致糖尿病、心血管疾病、神经退行性疾病和癌症等不同代谢疾病的主要原因之一。一些代谢性疾病,如糖尿病,会导致微血管和大血管并发症后的继发性并发症。一些最常见的神经退行性疾病,如认知障碍和阿尔茨海默氏症,都是在慢性糖尿病患者中发现的。本研究旨在利用阿脲诱导的大鼠认知障碍模型,了解糖尿病与认知障碍之间的相互联系机制。一水阿脲会产生活性氧,产生超氧自由基、过氧化氢和羟自由基。羟自由基最终导致β细胞死亡,引发糖尿病。因此,认知障碍中氧化应激与神经变性的相关性是本研究的导火索。在本研究中,我们探讨了维达列汀(VLD)及其共轭纳米粒子对氧化应激导致的阿脲相关脑损伤的改善作用。研究人员合成了维达列汀的金(Au)、硒(Se)纳米颗粒和双金属(Se@Au)纳米复合材料,并评估了它们对大脑可用性的改善作用。使用 DPPH、ABTS 和 FRAP 法对 VLD 和纳米颗粒进行了体外抗氧化评估。对大鼠大脑进行了与记忆相关的神经行为研究、体内抗氧化研究、体内生化研究和组织病理学检查。结果表明,VLD 及其纳米制剂在体外和体内抗氧化性方面均有显著的表现(p
{"title":"Effect of vildagliptin conjugated monometallic nanoparticles and bimetallic nanocomposites on diabetes-induced cognitive deficit","authors":"","doi":"10.1016/j.expneurol.2024.115026","DOIUrl":"10.1016/j.expneurol.2024.115026","url":null,"abstract":"<div><div>Oxidative stress is one of the major causes of different metabolic disorders, including diabetes, cardiovascular diseases, neurodegenerative diseases and cancers. Some metabolic disorders like diabetes mellitus leads to secondary complications after micro and macrovascular complications. Some of the most prevalent neurodegenerative diseases, like cognitive impairment and Alzheimer's disease, are found in chronic diabetic patients. The present study is designed to understand the mechanism of interconnection between diabetes mellitus and cognitive deficit using the alloxan model of diabetes-induced cognitive deficit in the rat model. The alloxan monohydrate produces reactive oxygen species, producing superoxide free radicals, hydrogen peroxide and hydroxyl radicals. The hydroxyl radicals ultimately cause the death of beta cells, causing diabetes. Hence, the correlation of oxidative stress and neurodegeneration in cognitive impairment is the trigger for this study. In the present study, we investigate the ameliorative effect of vildagliptin (VLD) and its conjugated nanoparticles against alloxan-associated brain damage due to oxidative stress. The gold (Au), selenium (Se) nanoparticles, and bimetallic (Se@Au) nanocomposites of VLD are synthesized and assessed for improvement in their brain availability. The in-vitro antioxidant evaluation of the VLD and nanoparticles is done using DPPH, ABTS, and FRAP assay. The memory-related neurobehavioral studies, in-vivo antioxidant studies, in-vivo biochemical studies, and histopathological examinations are evaluated in rat brains. The VLD and its nanoformulations exhibited in-vitro and in-vivo antioxidant properties significantly (<em>p</em> &lt; 0.01). They reduced the activity of AChE and nitrite in the alloxan diabetic rats. The bimetallic Se@Au VLDNCs displayed a more protective effect than VLD, VLD–AuNPs, and VLD–SeNPs.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142497761","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting PTEN in ischemic stroke: From molecular mechanisms to therapeutic potentials 针对缺血性中风的 PTEN:从分子机制到治疗潜力。
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-10-24 DOI: 10.1016/j.expneurol.2024.115023
Ischemic stroke remains a leading cause of mortality and disability worldwide, driven by complex pathophysiological mechanisms, including excitotoxicity, oxidative stress, apoptosis, and neuroinflammation. PTEN (Phosphatase and tensin homolog deleted on chromosome 10) plays a crucial role in these processes, influencing key signaling pathways such as PI3K/Akt and mTOR. This review aims to explore PTEN's multifaceted functions in ischemic stroke, examining its interactions with non-coding RNAs, involvement in mitophagy and immune suppression, and overall impact on cellular homeostasis. We will investigate various therapeutic strategies targeting PTEN, including synthetic drugs, natural products, and exosome-based therapies enriched with specific miRNAs. Additionally, we will assess the potential of non-pharmaceutical interventions such as electroacupuncture, exercise, transcranial direct current stimulation (tDCS), and therapeutic hypothermia in modulating PTEN activity to enhance cererbroprotection and functional recovery. By elucidating these aspects, this review aims to inspire and motivate the audience in their research and clinical practice, highlighting PTEN as a promising therapeutic target and paving the way for developing effective treatments for ischemic stroke.
缺血性中风仍然是全球死亡和残疾的主要原因,其病理生理机制复杂,包括兴奋毒性、氧化应激、细胞凋亡和神经炎症。PTEN(10 号染色体上删除的磷酸酶和天丝同源物)在这些过程中发挥着关键作用,影响着 PI3K/Akt 和 mTOR 等关键信号通路。本综述旨在探讨 PTEN 在缺血性中风中的多方面功能,研究其与非编码 RNA 的相互作用、参与有丝分裂和免疫抑制以及对细胞稳态的总体影响。我们将研究针对 PTEN 的各种治疗策略,包括合成药物、天然产品和富含特定 miRNA 的外泌体疗法。此外,我们还将评估电针、运动、经颅直流电刺激(tDCS)和治疗性低温等非药物干预措施在调节 PTEN 活性以增强神经保护和功能恢复方面的潜力。通过阐明这些方面,本综述旨在启发和激励读者的研究和临床实践,突出 PTEN 作为一个有前景的治疗靶点,为开发缺血性卒中的有效治疗方法铺平道路。
{"title":"Targeting PTEN in ischemic stroke: From molecular mechanisms to therapeutic potentials","authors":"","doi":"10.1016/j.expneurol.2024.115023","DOIUrl":"10.1016/j.expneurol.2024.115023","url":null,"abstract":"<div><div>Ischemic stroke remains a leading cause of mortality and disability worldwide, driven by complex pathophysiological mechanisms, including excitotoxicity, oxidative stress, apoptosis, and neuroinflammation. PTEN (Phosphatase and tensin homolog deleted on chromosome 10) plays a crucial role in these processes, influencing key signaling pathways such as PI3K/Akt and mTOR. This review aims to explore PTEN's multifaceted functions in ischemic stroke, examining its interactions with non-coding RNAs, involvement in mitophagy and immune suppression, and overall impact on cellular homeostasis. We will investigate various therapeutic strategies targeting PTEN, including synthetic drugs, natural products, and exosome-based therapies enriched with specific miRNAs. Additionally, we will assess the potential of non-pharmaceutical interventions such as electroacupuncture, exercise, transcranial direct current stimulation (tDCS), and therapeutic hypothermia in modulating PTEN activity to enhance cererbroprotection and functional recovery. By elucidating these aspects, this review aims to inspire and motivate the audience in their research and clinical practice, highlighting PTEN as a promising therapeutic target and paving the way for developing effective treatments for ischemic stroke.</div></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142497764","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Experimental Neurology
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:481959085
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