首页 > 最新文献

Experimental Neurology最新文献

英文 中文
Targeting astrocytic TDAG8 with delayed CO2 postconditioning improves functional outcomes after controlled cortical impact injury in mice 以星形胶质细胞 TDAG8 为靶点的延迟二氧化碳后处理技术可改善小鼠受控皮层撞击损伤后的功能预后。
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-07-22 DOI: 10.1016/j.expneurol.2024.114892

T-cell death-associated gene 8 (TDAG8), a G-protein-coupled receptor sensing physiological or weak acids, regulates inflammatory responses. However, its role in traumatic brain injury (TBI) remains unknown. Our recent study showed that delayed CO2 postconditioning (DCPC) has neuroreparative effects after TBI. We hypothesized that activating astrocytic TDAG8 is a key mechanism for DCPC. WT and TDAG8−/− mice received DCPC daily by transiently inhaling 10% CO2 after controlled cortical impact (CCI). HBAAV2/9-GFAP-m-TDAG8-3xflag-EGFP was used to overexpress TDAG8 in astrocytes. The beam walking test, mNSS, immunofluorescence and Golgi-Cox staining were used to evaluate motor function, glial activation and dendritic plasticity. DCPC significantly improved motor function; increased total dendritic length, neuronal complexity and spine density; inhibited overactivation of astrocytes and microglia; and promoted the expression of astrocytic brain-derived neurotrophic factor in WT but not TDAG8−/− mice. Overexpressing TDAG8 in astrocytes surrounding the lesion in TDAG8−/− mice restored the beneficial effects of DCPC. Although the effects of DCPC on Days 14–28 were much weaker than those of DCPC on Days 3–28 in WT mice, these effects were further enhanced by overexpressing astrocytic TDAG8. Astrocytic TDAG8 is a key target of DCPC for TBI rehabilitation. Its overexpression is a strategy that broadens the therapeutic window and enhances the effects of DCPC.

T细胞死亡相关基因8(TDAG8)是一种感应生理酸或弱酸的G蛋白偶联受体,可调节炎症反应。然而,它在创伤性脑损伤(TBI)中的作用仍然未知。我们最近的研究表明,延迟二氧化碳后处理(DCPC)具有创伤性脑损伤后神经恢复的作用。我们假设激活星形胶质细胞 TDAG8 是 DCPC 的关键机制。WT 小鼠和 TDAG8-/- 小鼠每天在受控皮质冲击(CCI)后瞬时吸入 10% CO2,从而接受 DCPC。HBAAV2/9-GFAP-m-TDAG8-3xflag-EGFP用于在星形胶质细胞中过表达TDAG8。横梁行走试验、mNSS、免疫荧光和 Golgi-Cox 染色用于评估运动功能、神经胶质活化和树突可塑性。DCPC 能明显改善 WT 小鼠的运动功能;增加树突总长度、神经元复杂性和脊柱密度;抑制星形胶质细胞和小胶质细胞的过度活化;促进星形胶质细胞脑源性神经营养因子的表达,但不能促进 TDAG8-/- 小鼠的表达。在 TDAG8-/- 小鼠病变周围的星形胶质细胞中过表达 TDAG8 可恢复 DCPC 的有益作用。虽然第 14-28 天 DCPC 对 WT 小鼠的作用比第 3-28 天 DCPC 的作用弱得多,但过表达星形胶质细胞 TDAG8 可进一步增强这些作用。星形胶质细胞 TDAG8 是 DCPC 用于创伤性脑损伤康复的关键靶点。过表达 TDAG8 是扩大治疗窗口和增强 DCPC 效果的一种策略。
{"title":"Targeting astrocytic TDAG8 with delayed CO2 postconditioning improves functional outcomes after controlled cortical impact injury in mice","authors":"","doi":"10.1016/j.expneurol.2024.114892","DOIUrl":"10.1016/j.expneurol.2024.114892","url":null,"abstract":"<div><p>T-cell death-associated gene 8 (TDAG8), a G-protein-coupled receptor sensing physiological or weak acids, regulates inflammatory responses. However, its role in traumatic brain injury (TBI) remains unknown. Our recent study showed that delayed CO<sub>2</sub> postconditioning (DCPC) has neuroreparative effects after TBI. We hypothesized that activating astrocytic TDAG8 is a key mechanism for DCPC. WT and TDAG8<sup>−/−</sup> mice received DCPC daily by transiently inhaling 10% CO<sub>2</sub> after controlled cortical impact (CCI). HBAAV2/9-GFAP-m-TDAG8-3xflag-EGFP was used to overexpress TDAG8 in astrocytes. The beam walking test, mNSS, immunofluorescence and Golgi-Cox staining were used to evaluate motor function, glial activation and dendritic plasticity. DCPC significantly improved motor function; increased total dendritic length, neuronal complexity and spine density; inhibited overactivation of astrocytes and microglia; and promoted the expression of astrocytic brain-derived neurotrophic factor in WT but not TDAG8<sup>−/−</sup> mice. Overexpressing TDAG8 in astrocytes surrounding the lesion in TDAG8<sup>−/−</sup> mice restored the beneficial effects of DCPC. Although the effects of DCPC on Days 14–28 were much weaker than those of DCPC on Days 3–28 in WT mice, these effects were further enhanced by overexpressing astrocytic TDAG8. Astrocytic TDAG8 is a key target of DCPC for TBI rehabilitation. Its overexpression is a strategy that broadens the therapeutic window and enhances the effects of DCPC.</p></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141758092","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
Neuro-molecular perspectives on long COVID-19 impacted cerebrovascular diseases - a role for dipeptidyl peptidase IV 长 COVID-19 影响脑血管疾病的神经分子观点--二肽基肽酶 IV 的作用。
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-07-20 DOI: 10.1016/j.expneurol.2024.114890

The coronavirus disease 2019 (COVID-19) has caused immense devastation globally with many outcomes that are now extending to its long-term sequel called long COVID. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects not only lungs, but also the brain and heart in association with endothelial cell dysfunction, coagulation abnormalities, and thrombosis leading to cardio-cerebrovascular health issues. Fatigue, cognitive decline, and brain fog are common neurological symptoms in persisting long COVID. Neurodegenerative processes and SARS-CoV-2 infection manifest overlapping molecular mechanisms, such as cytokine dysregulation, inflammation, protein aggregation, mitochondrial dysfunction, and oxidative stress. Identifying the key molecules in these processes is of importance for prevention and treatment of this disease. In particular, Dipeptidyl peptidase IV (DPPIV), a multifunctional peptidase has recently drawn attention as a potential co-receptor for SARS-CoV-2 infection and cellular entry. DPPIV is a known co-receptor for some other COVID viruses including MERS-Co-V. DPPIV regulates the immune responses, obesity, glucose metabolism, diabetes, and hypertension that are associated with cerebrovascular manifestations including stroke. DPPIV likely worsens persisting COVID-19 by disrupting inflammatory signaling pathways and the neurovascular system. This review highlights the neurological, cellular and molecular processes concerning long COVID, and DPPIV as a potential key factor contributing to cerebrovascular dysfunctions following SARS-CoV-2 infection.

2019 年冠状病毒病(COVID-19)在全球范围内造成了巨大的破坏,导致了许多后果,现在又延伸出了被称为 "长期冠状病毒病 "的长期后遗症。严重急性呼吸系统综合征冠状病毒 2(SARS-CoV-2)不仅感染肺部,还感染大脑和心脏,并伴有内皮细胞功能障碍、凝血异常和血栓形成,导致心脑血管健康问题。疲劳、认知能力下降和脑雾是长期 COVID 患者常见的神经系统症状。神经退行性过程和 SARS-CoV-2 感染表现出重叠的分子机制,如细胞因子失调、炎症、蛋白质聚集、线粒体功能障碍和氧化应激。确定这些过程中的关键分子对于预防和治疗这种疾病非常重要。特别是二肽基肽酶 IV(DPPIV),它是一种多功能肽酶,最近作为 SARS-CoV-2 感染和进入细胞的潜在共受体引起了人们的关注。DPPIV 是包括 MERS-Co-V 在内的一些其他 COVID 病毒的已知共受体。DPPIV 调节免疫反应、肥胖、糖代谢、糖尿病和高血压,而这些都与包括中风在内的脑血管表现有关。DPPIV 很可能会通过破坏炎症信号通路和神经血管系统而加重 COVID-19 的持续存在。本综述强调了与长COVID有关的神经、细胞和分子过程,以及DPPIV作为导致SARS-CoV-2感染后脑血管功能障碍的潜在关键因素。
{"title":"Neuro-molecular perspectives on long COVID-19 impacted cerebrovascular diseases - a role for dipeptidyl peptidase IV","authors":"","doi":"10.1016/j.expneurol.2024.114890","DOIUrl":"10.1016/j.expneurol.2024.114890","url":null,"abstract":"<div><p>The coronavirus disease 2019 (COVID-19) has caused immense devastation globally with many outcomes that are now extending to its long-term sequel called long COVID. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects not only lungs, but also the brain and heart in association with endothelial cell dysfunction, coagulation abnormalities, and thrombosis leading to cardio-cerebrovascular health issues. Fatigue, cognitive decline, and brain fog are common neurological symptoms in persisting long COVID. Neurodegenerative processes and SARS-CoV-2 infection manifest overlapping molecular mechanisms, such as cytokine dysregulation, inflammation, protein aggregation, mitochondrial dysfunction, and oxidative stress. Identifying the key molecules in these processes is of importance for prevention and treatment of this disease. In particular, Dipeptidyl peptidase IV (DPPIV), a multifunctional peptidase has recently drawn attention as a potential co-receptor for SARS-CoV-2 infection and cellular entry. DPPIV is a known co-receptor for some other COVID viruses including MERS-Co-V. DPPIV regulates the immune responses, obesity, glucose metabolism, diabetes, and hypertension that are associated with cerebrovascular manifestations including stroke. DPPIV likely worsens persisting COVID-19 by disrupting inflammatory signaling pathways and the neurovascular system. This review highlights the neurological, cellular and molecular processes concerning long COVID, and DPPIV as a potential key factor contributing to cerebrovascular dysfunctions following SARS-CoV-2 infection.</p></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-07-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141747803","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
A facilitatory role of astrocytes in axonal regeneration after acute and chronic spinal cord injury 星形胶质细胞在急性和慢性脊髓损伤后轴突再生中的促进作用
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-07-15 DOI: 10.1016/j.expneurol.2024.114889

Neuroscience dogma avers that astrocytic “scars” inhibit axonal regeneration after spinal cord injury (SCI). A recent report suggested however that astrocytes form “borders” around lesions that are permissive rather than inhibitory to axonal growth. We now provide further evidence supporting a facilitatory role of astrocytes in axonal regeneration after SCI. First, even 6months after SCI, injured axons are retained within regions of densely reactive astrocytes, in direct contact with astrocyte processes without being repelled. Second, 6 month-delayed implants of neural stem cells extend axons into reactive astrocyte borders surrounding lesions, densely contacting astrocyte surfaces. Third, bioengineered hydrogels implanted into sites of SCI re-orient reactive astrocytic processes to align along the rostral-to-caudal spinal cord axis resulting in successful regeneration into the lesion/scaffold in close association with astrocytic processes. Fourth, corticospinal axons regenerate into neural progenitor cells implanted six months after injury in close association with host astrocytic processes. Thus, astrocytes do not appear to inhibit axonal regeneration, and the close association of newly growing axons with astrocytic processes suggests a facilitatory role in axonal regeneration.

神经科学教条认为,星形胶质细胞的 "疤痕 "会抑制脊髓损伤(SCI)后的轴突再生。然而,最近的一份报告指出,星形胶质细胞在病变周围形成的 "边界 "对轴突生长是允许性的,而不是抑制性的。现在,我们提供了进一步的证据,支持星形胶质细胞在脊髓损伤后的轴突再生中发挥促进作用。首先,即使在 SCI 6 个月后,受伤的轴突仍能保留在密集反应的星形胶质细胞区域内,与星形胶质细胞过程直接接触而不被排斥。其次,延迟6个月植入的神经干细胞可将轴突延伸至病变周围的反应性星形胶质细胞边界,密集接触星形胶质细胞表面。第三,将生物工程水凝胶植入脊髓损伤部位,可重新定向反应性星形胶质细胞过程,使其沿着脊髓喙向尾轴线排列,从而成功再生到与星形胶质细胞过程密切相关的病变/支架部位。第四,皮质脊髓轴突再生到损伤后六个月植入的神经祖细胞中,与宿主星形胶质细胞过程紧密结合。因此,星形胶质细胞似乎并不抑制轴突再生,而新生长的轴突与星形胶质细胞过程的密切联系表明,星形胶质细胞在轴突再生中起着促进作用。
{"title":"A facilitatory role of astrocytes in axonal regeneration after acute and chronic spinal cord injury","authors":"","doi":"10.1016/j.expneurol.2024.114889","DOIUrl":"10.1016/j.expneurol.2024.114889","url":null,"abstract":"<div><p>Neuroscience dogma avers that astrocytic “scars” inhibit axonal regeneration after spinal cord injury (SCI). A recent report suggested however that astrocytes form “borders” around lesions that are permissive rather than inhibitory to axonal growth. We now provide further evidence supporting a facilitatory role of astrocytes in axonal regeneration after SCI. First, even 6months after SCI, injured axons are retained within regions of densely reactive astrocytes, in direct contact with astrocyte processes without being repelled. Second, 6 month-delayed implants of neural stem cells extend axons <em>into</em> reactive astrocyte borders surrounding lesions, densely contacting astrocyte surfaces. Third, bioengineered hydrogels implanted into sites of SCI <em>re-orient</em> reactive astrocytic processes to align along the rostral-to-caudal spinal cord axis resulting in successful regeneration into the lesion/scaffold in close association with astrocytic processes. Fourth, corticospinal axons regenerate into neural progenitor cells implanted six months after injury in close association with host astrocytic processes. Thus, astrocytes do not appear to inhibit axonal regeneration, and the close association of newly growing axons with astrocytic processes suggests a facilitatory role in axonal regeneration.</p></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0014488624002152/pdfft?md5=9a71f7ccde011f41f3d42497225d51e7&pid=1-s2.0-S0014488624002152-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141633109","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
The role of exosomes derived from stem cells in nerve regeneration: A contribution to neurological repair 干细胞外泌体在神经再生中的作用:对神经修复的贡献
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-07-14 DOI: 10.1016/j.expneurol.2024.114882

Stem cell-derived exosomes have gained attention in regenerative medicine for their role in encouraging nerve regeneration and potential use in treating neurological diseases. These nanosized extracellular vesicles act as carriers of bioactive molecules, facilitating intercellular communication and enhancing the regenerative process in neural tissues. This comprehensive study explores the methods by which exosomes produced from various stem cells contribute to nerve healing, with a particular emphasis on their role in angiogenesis, inflammation, and cellular signaling pathways. By examining cutting-edge developments and exploring the potential of exosomes in delivering disease-specific miRNAs and proteins, we highlight their versatility in tailoring personalized therapeutic strategies. The findings presented here highlight the potential of stem cell-produced exosomes for use in neurological diseases therapy, establishing the door for future research into exosome-based neurotherapies.

干细胞衍生的外泌体在促进神经再生和治疗神经系统疾病的潜在用途方面发挥着重要作用,因而在再生医学领域备受关注。这些纳米级细胞外囊泡是生物活性分子的载体,可促进细胞间的交流并增强神经组织的再生过程。本综合研究探讨了各种干细胞产生的外泌体促进神经愈合的方法,特别强调了它们在血管生成、炎症和细胞信号通路中的作用。通过研究前沿发展和探索外泌体在传递疾病特异性miRNA和蛋白质方面的潜力,我们强调了外泌体在定制个性化治疗策略方面的多功能性。本文的研究结果凸显了干细胞产生的外泌体用于神经系统疾病治疗的潜力,为未来基于外泌体的神经疗法研究打开了大门。
{"title":"The role of exosomes derived from stem cells in nerve regeneration: A contribution to neurological repair","authors":"","doi":"10.1016/j.expneurol.2024.114882","DOIUrl":"10.1016/j.expneurol.2024.114882","url":null,"abstract":"<div><p>Stem cell-derived exosomes have gained attention in regenerative medicine for their role in encouraging nerve regeneration and potential use in treating neurological diseases. These nanosized extracellular vesicles act as carriers of bioactive molecules, facilitating intercellular communication and enhancing the regenerative process in neural tissues. This comprehensive study explores the methods by which exosomes produced from various stem cells contribute to nerve healing, with a particular emphasis on their role in angiogenesis, inflammation, and cellular signaling pathways. By examining cutting-edge developments and exploring the potential of exosomes in delivering disease-specific miRNAs and proteins, we highlight their versatility in tailoring personalized therapeutic strategies. The findings presented here highlight the potential of stem cell-produced exosomes for use in neurological diseases therapy, establishing the door for future research into exosome-based neurotherapies.</p></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-07-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141603551","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
⍺-Synuclein levels in Parkinson's disease – Cell types and forms that contribute to pathogenesis 帕金森病中的突触核蛋白水平--导致发病的细胞类型和形式。
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-07-14 DOI: 10.1016/j.expneurol.2024.114887

Parkinson's disease (PD) has two main pathological hallmarks, the loss of nigral dopamine neurons and the proteinaceous aggregations of ⍺-synuclein (⍺Syn) in neuronal Lewy pathology. These two co-existing features suggest a causative association between ⍺Syn aggregation and the underpinning mechanism of neuronal degeneration in PD. Both increased levels and post-translational modifications of ⍺Syn can contribute to the formation of pathological aggregations of ⍺Syn in neurons. Recent studies have shown that the protein is also expressed by multiple types of non-neuronal cells in the brain and peripheral tissues, suggesting additional roles of the protein and potential diversity in non-neuronal pathogenic triggers. It is important to determine (1) the threshold levels triggering ⍺Syn to convert from a biological to a pathologic form in different brain cells in PD; (2) the dominant form of pathologic ⍺Syn and the associated post-translational modification of the protein in each cell type involved in PD; and (3) the cell type associated biological processes impacted by pathologic ⍺Syn in PD. This review integrates these aspects and speculates on potential pathological mechanisms and their impact on neuronal and non-neuronal ⍺Syn in the brains of patients with PD.

帕金森病(PD)有两个主要的病理特征,即黑质多巴胺神经元的丧失和神经元路易病理学中突触核蛋白(Syn)的蛋白聚集。这两个并存的特征表明,Syn 的聚集与帕金森病神经元变性的基础机制之间存在因果关系。Syn水平的升高和翻译后修饰都可能导致神经元中Syn病理性聚集的形成。最近的研究表明,该蛋白在大脑和外周组织的多种类型的非神经元细胞中也有表达,这表明该蛋白在非神经元致病诱因中具有额外的作用和潜在的多样性。重要的是要确定:(1) 在帕金森病的不同脑细胞中,触发⍺Syn 从生物形式转化为病理形式的阈值水平;(2) 病理⍺Syn 的主要形式以及在帕金森病涉及的每种细胞类型中该蛋白的相关翻译后修饰;(3) 帕金森病中病理⍺Syn 影响的细胞类型相关生物过程。本综述综合了这些方面,并推测了潜在的病理机制及其对帕金森病患者大脑中神经元和非神经元⍺Syn的影响。
{"title":"⍺-Synuclein levels in Parkinson's disease – Cell types and forms that contribute to pathogenesis","authors":"","doi":"10.1016/j.expneurol.2024.114887","DOIUrl":"10.1016/j.expneurol.2024.114887","url":null,"abstract":"<div><p>Parkinson's disease (PD) has two main pathological hallmarks, the loss of nigral dopamine neurons and the proteinaceous aggregations of ⍺-synuclein (⍺Syn) in neuronal Lewy pathology. These two co-existing features suggest a causative association between ⍺Syn aggregation and the underpinning mechanism of neuronal degeneration in PD. Both increased levels and post-translational modifications of ⍺Syn can contribute to the formation of pathological aggregations of ⍺Syn in neurons. Recent studies have shown that the protein is also expressed by multiple types of non-neuronal cells in the brain and peripheral tissues, suggesting additional roles of the protein and potential diversity in non-neuronal pathogenic triggers. It is important to determine (1) the threshold levels triggering ⍺Syn to convert from a biological to a pathologic form in different brain cells in PD; (2) the dominant form of pathologic ⍺Syn and the associated post-translational modification of the protein in each cell type involved in PD; and (3) the cell type associated biological processes impacted by pathologic ⍺Syn in PD. This review integrates these aspects and speculates on potential pathological mechanisms and their impact on neuronal and non-neuronal ⍺Syn in the brains of patients with PD.</p></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-07-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0014488624002139/pdfft?md5=835660956cd37963c3a4564489076dcb&pid=1-s2.0-S0014488624002139-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619780","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
High-fat diet consumption negatively influences closed-head traumatic brain injury in a pediatric rodent model 高脂饮食对小儿啮齿动物模型中的闭头创伤性脑损伤有负面影响。
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-07-14 DOI: 10.1016/j.expneurol.2024.114888

Traumatic brain injury (TBI) is one of the most common causes of emergency room visits in children, and it is a leading cause of death in juveniles in the United States. Similarly, a high proportion of this population consumes diets that are high in saturated fats, and millions of children are overweight or obese. The goal of the present study was to assess the relationship between diet and TBI on cognitive and cerebrovascular outcomes in juvenile rats. In the current study, groups of juvenile male Long Evans rats were subjected to either mild TBI via the Closed-Head Injury Model of Engineered Rotational Acceleration (CHIMERA) or underwent sham procedures. The animals were provided with either a combination of high-fat diet and a mixture of high-fructose corn syrup (HFD/HFCS) or a standard chow diet (CH) for 9 days prior to injury. Prior to injury, the animals were trained on the Morris water maze for three consecutive days, and they underwent a post-injury trial on the day of the injury. Immediately after TBI, the animals' righting reflexes were tested. Four days post-injury, the animals were euthanized, and brain samples and blood plasma were collected for qRT-PCR, immunohistochemistry, and triglyceride assays. Additional subsets of animals were used to investigate cerebrovascular perfusion using Laser Speckle and perform immunohistochemistry for endothelial cell marker RECA. Following TBI, the righting reflex was significantly increased in TBI rats, irrespective of diet. The TBI worsened the rats' performance in the post-injury trial of the water maze at 3 h, p(injury) < 0.05, but not at 4 days post-injury. Reduced cerebrovascular blood flow using Laser Speckle was demonstrated in the cerebellum, p(injury) < 0.05, but not foci of the cerebral cortices or superior sagittal sinus. Immunoreactive staining for RECA in the cortex and corpus callosum was significantly reduced in HFD/HFCS TBI rats, p < 0.05. qRT-PCR showed significant increases in APOE, CREB1, FCGR2B, IL1B, and IL6, particularly in the hippocampus. The results from this study offer robust evidence that HFD/HFCS negatively influences TBI outcomes with respect to cognition and cerebrovascular perfusion of relevant brain regions in the juvenile rat.

创伤性脑损伤(TBI)是儿童看急诊最常见的原因之一,也是美国青少年死亡的主要原因。同样,这一人群中有很大一部分人的饮食中饱和脂肪含量较高,数百万儿童超重或肥胖。本研究的目的是评估饮食和创伤性脑损伤对幼鼠认知和脑血管结果的影响。在本研究中,一组幼年雄性长埃文斯大鼠通过工程旋转加速度闭头损伤模型(CHIMERA)接受了轻度创伤性脑损伤,或接受了假手术。这些动物在受伤前 9 天均食用高脂肪饮食和高果糖玉米糖浆混合物(HFD/HFCS)或标准饲料(CH)。受伤前,连续三天对动物进行莫里斯水迷宫训练,并在受伤当天进行伤后试验。创伤性脑损伤后,立即对动物的右反射进行测试。受伤四天后,动物被安乐死,收集脑样本和血浆进行 qRT-PCR、免疫组化和甘油三酯检测。此外,还利用激光斑点技术研究脑血管灌注情况,并对内皮细胞标记物 RECA 进行免疫组化。创伤性脑损伤后,无论饮食如何,创伤性脑损伤大鼠的向右反射都明显增加。创伤性脑损伤使大鼠在伤后3小时的水迷宫试验中表现恶化,p(损伤)
{"title":"High-fat diet consumption negatively influences closed-head traumatic brain injury in a pediatric rodent model","authors":"","doi":"10.1016/j.expneurol.2024.114888","DOIUrl":"10.1016/j.expneurol.2024.114888","url":null,"abstract":"<div><p>Traumatic brain injury (TBI) is one of the most common causes of emergency room visits in children, and it is a leading cause of death in juveniles in the United States. Similarly, a high proportion of this population consumes diets that are high in saturated fats, and millions of children are overweight or obese. The goal of the present study was to assess the relationship between diet and TBI on cognitive and cerebrovascular outcomes in juvenile rats. In the current study, groups of juvenile male Long Evans rats were subjected to either mild TBI via the Closed-Head Injury Model of Engineered Rotational Acceleration (CHIMERA) or underwent sham procedures. The animals were provided with either a combination of high-fat diet and a mixture of high-fructose corn syrup (HFD/HFCS) or a standard chow diet (CH) for 9 days prior to injury. Prior to injury, the animals were trained on the Morris water maze for three consecutive days, and they underwent a post-injury trial on the day of the injury. Immediately after TBI, the animals' righting reflexes were tested. Four days post-injury, the animals were euthanized, and brain samples and blood plasma were collected for qRT-PCR, immunohistochemistry, and triglyceride assays. Additional subsets of animals were used to investigate cerebrovascular perfusion using Laser Speckle and perform immunohistochemistry for endothelial cell marker RECA. Following TBI, the righting reflex was significantly increased in TBI rats, irrespective of diet. The TBI worsened the rats' performance in the post-injury trial of the water maze at 3 h, <em>p</em>(injury) &lt; 0.05, but not at 4 days post-injury. Reduced cerebrovascular blood flow using Laser Speckle was demonstrated in the cerebellum, <em>p</em>(injury) &lt; 0.05, but not foci of the cerebral cortices or superior sagittal sinus. Immunoreactive staining for RECA in the cortex and corpus callosum was significantly reduced in HFD/HFCS TBI rats, <em>p</em> &lt; 0.05. qRT-PCR showed significant increases in APOE, CREB1, FCGR2B, IL1B, and IL6, particularly in the hippocampus. The results from this study offer robust evidence that HFD/HFCS negatively influences TBI outcomes with respect to cognition and cerebrovascular perfusion of relevant brain regions in the juvenile rat.</p></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-07-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619781","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
Mechanistic insights into Lipocalin-2 in ischemic stroke and hemorrhagic brain injury: Integrating animal and clinical studies 缺血性中风和出血性脑损伤中脂质结合素-2的机制研究:整合动物和临床研究。
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-07-10 DOI: 10.1016/j.expneurol.2024.114885
Korsin Laohavisudhi , Sirawit Sriwichaiin , Tanawat Attachaipanich , Borwon Wittayachamnankul , Nipon Chattipakorn , Siriporn Chattipakorn

Brain injuries, including strokes and traumatic brain injuries (TBI), are a major global health concern, contributing significantly to both mortality and long-term disability. Recent research has identified lipocalin-2 (LCN2), a glycoprotein secreted by various brain cells, as a key factor in influencing brain injury outcomes. Evidence from animal and clinical studies firmly establishes the pivotal role of LCN2 in driving the inflammatory responses triggered by damage to brain tissue. Furthermore, increased LCN2 promotes cellular differentiation, blood-brain barrier breakdown, and decreases cell viability. Interventions with LCN2 inhibitors attenuated brain injury through a reduction in the inflammation process and enhanced cellular viability. Potential mechanisms of LCN2 involve several pathways including the Janus kinase-2 (JAK2)-signal transducers and the transcription-3 (STAT3) signaling, hypoxia-inducible factor 1-alpha (HIF-1α)-LCN2-vascular endothelial growth factor alpha (VEGFα), and the PKR-like ER kinase (PERK) pathways. LCN2 itself interacts with diverse inflammatory cytokines in TBI and intracranial hemorrhage (ICH), resulting in disruption of the blood-brain barrier, increased programmed cell death, and an imbalance in iron homeostasis. Clinical studies have also shown that increased LCN2 level can act as a prognostic biomarker of outcomes following brain injuries. Therefore, this review aims to comprehensively evaluate the role and underlying mechanisms of LCN2 in brain injuries, including stroke and TBI, and explore potential therapeutic interventions targeting LCN2 in these conditions.

脑损伤,包括脑卒中和创伤性脑损伤(TBI),是全球关注的主要健康问题,是导致死亡和长期残疾的重要因素。最新研究发现,脂钙蛋白-2(LCN2)是由各种脑细胞分泌的一种糖蛋白,是影响脑损伤结果的关键因素。来自动物和临床研究的证据牢固确立了 LCN2 在推动脑组织损伤引发的炎症反应中的关键作用。此外,LCN2 的增加会促进细胞分化、破坏血脑屏障并降低细胞活力。使用 LCN2 抑制剂进行干预可通过减少炎症过程和提高细胞活力来减轻脑损伤。LCN2 的潜在机制涉及多个途径,包括 Janus 激酶-2(JAK2)-信号转导和转录-3(STAT3)信号、缺氧诱导因子 1-α(HIF-1α)-LCN2-血管内皮生长因子α(VEGFα)和 PKR 样 ER 激酶(PERK)途径。在创伤性脑损伤和颅内出血(ICH)中,LCN2 本身与多种炎症细胞因子相互作用,导致血脑屏障破坏、程序性细胞死亡增加以及铁平衡失调。临床研究还表明,LCN2 水平的升高可作为脑损伤后预后的生物标志物。因此,本综述旨在全面评估 LCN2 在脑损伤(包括中风和创伤性脑损伤)中的作用和潜在机制,并探讨针对 LCN2 的潜在治疗干预措施。
{"title":"Mechanistic insights into Lipocalin-2 in ischemic stroke and hemorrhagic brain injury: Integrating animal and clinical studies","authors":"Korsin Laohavisudhi ,&nbsp;Sirawit Sriwichaiin ,&nbsp;Tanawat Attachaipanich ,&nbsp;Borwon Wittayachamnankul ,&nbsp;Nipon Chattipakorn ,&nbsp;Siriporn Chattipakorn","doi":"10.1016/j.expneurol.2024.114885","DOIUrl":"10.1016/j.expneurol.2024.114885","url":null,"abstract":"<div><p>Brain injuries, including strokes and traumatic brain injuries (TBI), are a major global health concern, contributing significantly to both mortality and long-term disability. Recent research has identified lipocalin-2 (LCN2), a glycoprotein secreted by various brain cells, as a key factor in influencing brain injury outcomes. Evidence from animal and clinical studies firmly establishes the pivotal role of LCN2 in driving the inflammatory responses triggered by damage to brain tissue. Furthermore, increased LCN2 promotes cellular differentiation, blood-brain barrier breakdown, and decreases cell viability. Interventions with LCN2 inhibitors attenuated brain injury through a reduction in the inflammation process and enhanced cellular viability. Potential mechanisms of LCN2 involve several pathways including the Janus kinase-2 (JAK2)-signal transducers and the transcription-3 (STAT3) signaling, hypoxia-inducible factor 1-alpha (HIF-1α)-LCN2-vascular endothelial growth factor alpha (VEGFα), and the PKR-like ER kinase (PERK) pathways. LCN2 itself interacts with diverse inflammatory cytokines in TBI and intracranial hemorrhage (ICH), resulting in disruption of the blood-brain barrier, increased programmed cell death, and an imbalance in iron homeostasis. Clinical studies have also shown that increased LCN2 level can act as a prognostic biomarker of outcomes following brain injuries. Therefore, this review aims to comprehensively evaluate the role and underlying mechanisms of LCN2 in brain injuries, including stroke and TBI, and explore potential therapeutic interventions targeting LCN2 in these conditions.</p></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141598910","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
Anti-oxidative stress and cognitive improvement of a semi-synthetic isoorientin-based GSK-3β inhibitor in rat pheochromocytoma cell PC12 and scopolamine-induced AD model mice via AKT/GSK-3β/Nrf2 pathway 半合成异连翘素GSK-3β抑制剂通过AKT/GSK-3β/Nrf2通路在大鼠嗜铬细胞瘤细胞PC12和东莨菪碱诱导的AD模型小鼠中抗氧化和改善认知能力
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-07-10 DOI: 10.1016/j.expneurol.2024.114881

Background

Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive cognitive deficits. Although the pathogenesis of AD is unclear, oxidative stress has been implicated to play a dominant role in its development. The flavonoid isoorientin (ISO) and its synthetic derivatives TFGF-18 selectively inhibit glycogen synthase kinase-3β (GSK-3β), a potential target of AD treatment.

Purpose

To investigate the neuroprotective effect of TFGF-18 against oxidative stress via the GSK-3β pathway in hydrogen peroxide (H2O2)-induced rat pheochromocytoma PC12 cells in vitro and scopolamine (SCOP)-induced AD mice in vivo.

Method

The oxidative stress of PC12 cells was induced by H2O2 (600 μM) and the effects of TFGF-18 (2 and 8 μM) or ISO (12.5 and 50 μM) were observed. The AD mouse model was induced by SCOP (3 mg/kg), and the effects of TFGF-18 (2 and 8 mg/kg), ISO (50 mg/kg), and donepezil (DNP) (3 mg/kg) were observed. DNP, a currently accepted drug for AD was used as a positive control. The neuronal cell damages were analyzed by flow cytometry, LDH assay, JC-1 assay and Nissl staining. The oxidative stress was evaluated by the detection of MDA, SOD, GPx and ROS. The level of ACh, and the activity of AChE, ChAT were detected by the assay kit. The expressions of Bax, Bcl-2, caspase3, cleaved-caspase3, p-AKT (Thr308), AKT, p-GSK-3β (Ser9), GSK-3β, Nrf2, and HO-1, as well as p-CREB (Ser133), CREB, and BDNF were analyzed by western blotting. Morris water maze test was performed to analyze learning and memory ability.

Results

TFGF-18 inhibited neuronal damage and the expressions of Bax, caspase3 and cleaved-caspase3, and increased the expression of Bcl-2 in vitro and in vivo. The level of MDA and ROS were decreased while the activities of SOD and GPx were increased by TFGF-18. Moreover, TFGF-18 increased the p-AKT, p-GSK-3β (Ser9), Nrf2, HO-1, p-CREB, and BDNF expression reduced by H2O2 and SCOP. Meanwhile, MK2206, an AKT inhibitor, reversed the effect of TFGF-18 on the AKT/GSK-3β pathway. In addition, the cholinergic system (ACh, ChAT, and AChE) disorders were retrained and the learning and memory impairments were prevented by TFGF-18 in SCOP-induced AD mice.

Conclusions

TFGF-18 protects against neuronal cell damage and cognitive impairment by inhibiting oxidative stress via AKT/GSK-3β/Nrf2 pathway.

背景:阿尔茨海默病(AD)是一种神经退行性疾病,以进行性认知功能障碍为特征。虽然阿尔茨海默病的发病机制尚不清楚,但氧化应激被认为在其发展过程中起着主导作用。目的:在过氧化氢(H2O2)诱导的大鼠嗜铬细胞瘤PC12细胞体外实验和东莨菪碱(SCOP)诱导的AD小鼠体内实验中,研究TFGF-18通过GSK-3β途径对氧化应激的神经保护作用:方法:用H2O2(600 μM)诱导PC12细胞氧化应激,观察TFGF-18(2和8 μM)或ISO(12.5和50 μM)的作用。用 SCOP(3 毫克/千克)诱导 AD 小鼠模型,观察 TFGF-18(2 和 8 毫克/千克)、ISO(50 毫克/千克)和多奈哌齐(DNP)(3 毫克/千克)的作用。DNP是目前公认的治疗AD的药物,被用作阳性对照。流式细胞术、LDH测定、JC-1测定和Nissl染色分析了神经细胞的损伤情况。氧化应激通过检测 MDA、SOD、GPx 和 ROS 进行评估。乙酰胆碱水平以及乙酰胆碱酯酶(AChE)、胆碱酯酶(ChAT)的活性通过检测试剂盒进行检测。Bax、Bcl-2、caspase3、裂解-caspase3、p-AKT (Thr308)、AKT、p-GSK-3β (Ser9)、GSK-3β、Nrf2和HO-1以及p-CREB (Ser133)、CREB和BDNF的表达均通过Western印迹分析。莫里斯水迷宫测试分析了学习和记忆能力:结果:TFGF-18能抑制体外和体内神经元损伤、Bax caspase3和裂解caspase3的表达,并能增加Bcl-2的表达。TFGF-18 降低了 MDA 和 ROS 的水平,提高了 SOD 和 GPx 的活性。此外,TFGF-18 还能增加 p-AKT、p-GSK-3β(Ser9)、Nrf2、HO-1、p-CREB 和 BDNF 的表达,降低 H2O2 和 SCOP 的影响。同时,AKT 抑制剂 MK2206 逆转了 TFGF-18 对 AKT/GSK-3β 通路的影响。此外,TFGF-18还能重新训练胆碱能系统(ACh、ChAT和AChE)紊乱,并防止SCOP诱导的AD小鼠出现学习和记忆障碍:结论:TFGF-18可通过AKT/GSK-3β/Nrf2途径抑制氧化应激,从而防止神经细胞损伤和认知功能障碍。
{"title":"Anti-oxidative stress and cognitive improvement of a semi-synthetic isoorientin-based GSK-3β inhibitor in rat pheochromocytoma cell PC12 and scopolamine-induced AD model mice via AKT/GSK-3β/Nrf2 pathway","authors":"","doi":"10.1016/j.expneurol.2024.114881","DOIUrl":"10.1016/j.expneurol.2024.114881","url":null,"abstract":"<div><h3>Background</h3><p>Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive cognitive deficits. Although the pathogenesis of AD is unclear, oxidative stress has been implicated to play a dominant role in its development. The flavonoid isoorientin (ISO) and its synthetic derivatives TFGF-18 selectively inhibit glycogen synthase kinase-3β (GSK-3β), a potential target of AD treatment.</p></div><div><h3>Purpose</h3><p>To investigate the neuroprotective effect of TFGF-18 against oxidative stress <em>via</em> the GSK-3β pathway in hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>)-induced rat pheochromocytoma PC12 cells <em>in vitro</em> and scopolamine (SCOP)-induced AD mice <em>in vivo</em>.</p></div><div><h3>Method</h3><p>The oxidative stress of PC12 cells was induced by H<sub>2</sub>O<sub>2</sub> (600 μM) and the effects of TFGF-18 (2 and 8 μM) or ISO (12.5 and 50 μM) were observed. The AD mouse model was induced by SCOP (3 mg/kg), and the effects of TFGF-18 (2 and 8 mg/kg), ISO (50 mg/kg), and donepezil (DNP) (3 mg/kg) were observed. DNP, a currently accepted drug for AD was used as a positive control. The neuronal cell damages were analyzed by flow cytometry, LDH assay, JC-1 assay and Nissl staining. The oxidative stress was evaluated by the detection of MDA, SOD, GPx and ROS. The level of ACh, and the activity of AChE, ChAT were detected by the assay kit. The expressions of Bax, Bcl-2, caspase3, cleaved-caspase3, p-AKT (Thr308), AKT, p-GSK-3β (Ser9), GSK-3β, Nrf2, and HO-1, as well as p-CREB (Ser133), CREB, and BDNF were analyzed by western blotting. Morris water maze test was performed to analyze learning and memory ability.</p></div><div><h3>Results</h3><p>TFGF-18 inhibited neuronal damage and the expressions of Bax, caspase3 and cleaved-caspase3, and increased the expression of Bcl-2 <em>in vitro</em> and <em>in vivo</em>. The level of MDA and ROS were decreased while the activities of SOD and GPx were increased by TFGF-18. Moreover, TFGF-18 increased the p-AKT, p-GSK-3β (Ser9), Nrf2, HO-1, p-CREB, and BDNF expression reduced by H<sub>2</sub>O<sub>2</sub> and SCOP. Meanwhile, MK2206, an AKT inhibitor, reversed the effect of TFGF-18 on the AKT/GSK-3β pathway. In addition, the cholinergic system (ACh, ChAT, and AChE) disorders were retrained and the learning and memory impairments were prevented by TFGF-18 in SCOP-induced AD mice.</p></div><div><h3>Conclusions</h3><p>TFGF-18 protects against neuronal cell damage and cognitive impairment by inhibiting oxidative stress <em>via</em> AKT/GSK-3β/Nrf2 pathway.</p></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141598908","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
Irisflorentin improves functional recovery after spinal cord injury by protecting the blood–spinal cord barrier and promoting axonal growth 伊利司命通过保护血液-脊髓屏障和促进轴突生长,改善脊髓损伤后的功能恢复。
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-07-10 DOI: 10.1016/j.expneurol.2024.114886

Spinal cord injury (SCI) induces the disruption of the blood–spinal cord barrier (BSCB) and the failure of axonal growth. SCI activates a complex series of responses, including cell apoptosis and endoplasmic reticulum (ER) stress. Pericytes play a critical role in maintaining BSCB integrity and facilitating tissue growth and repair. However, the roles of pericytes in SCI and the potential mechanisms underlying the improvements in functional recovery in SCI remain unclear. Recent evidence indicates that irisflorentin exerts neuroprotective effects against Parkinson's disease; however, whether it has potential protective roles in SCI or not is still unknown. In this study, we found that the administration of irisflorentin significantly inhibited pericyte apoptosis, protected BSCB integrity, promoted axonal growth, and ultimately improved locomotion recovery in a rat model of SCI. In vitro, we found that the positive effects of irisflorentin on axonal growth were likely to be mediated by regulating the crosstalk between pericytes and neurons. Furthermore, irisflorentin effectively ameliorated ER stress caused by incubation with thapsigargin (TG) in pericytes. Meanwhile, the protective effect of irisflorentin on BSCB disruption is strongly related to the reduction of pericyte apoptosis via inhibition of ER stress. Collectively, our findings demonstrate that irisflorentin is beneficial for functional recovery after SCI and that pericytes are a valid target of interest for future SCI therapies.

脊髓损伤(SCI)会导致血脊髓屏障(BSCB)破坏和轴突生长失败。脊髓损伤激活了一系列复杂的反应,包括细胞凋亡和内质网(ER)应激。周细胞在维持脐带屏障完整性、促进组织生长和修复方面发挥着关键作用。然而,周细胞在 SCI 中的作用以及 SCI 功能恢复改善的潜在机制仍不清楚。最近的证据表明,虹膜视蛋白对帕金森病有神经保护作用,但它是否对 SCI 有潜在的保护作用仍不清楚。在这项研究中,我们发现在大鼠 SCI 模型中施用虹膜视蛋白能显著抑制周细胞凋亡、保护 BSCB 的完整性、促进轴突生长并最终改善运动恢复。在体外,我们发现虹膜前列素对轴突生长的积极作用可能是通过调节周细胞和神经元之间的串扰来实现的。此外,鸢尾黄素还能有效地改善周细胞在硫代甘氨酸(TG)作用下产生的ER应激。同时,鸢尾黄素对BSCB破坏的保护作用与通过抑制ER应激减少周细胞凋亡密切相关。总之,我们的研究结果表明,虹膜脂环素有利于脊髓损伤后的功能恢复,而且周细胞是未来脊髓损伤疗法的一个有效靶点。
{"title":"Irisflorentin improves functional recovery after spinal cord injury by protecting the blood–spinal cord barrier and promoting axonal growth","authors":"","doi":"10.1016/j.expneurol.2024.114886","DOIUrl":"10.1016/j.expneurol.2024.114886","url":null,"abstract":"<div><p>Spinal cord injury (SCI) induces the disruption of the blood–spinal cord barrier (BSCB) and the failure of axonal growth. SCI activates a complex series of responses, including cell apoptosis and endoplasmic reticulum (ER) stress. Pericytes play a critical role in maintaining BSCB integrity and facilitating tissue growth and repair. However, the roles of pericytes in SCI and the potential mechanisms underlying the improvements in functional recovery in SCI remain unclear. Recent evidence indicates that irisflorentin exerts neuroprotective effects against Parkinson's disease; however, whether it has potential protective roles in SCI or not is still unknown. In this study, we found that the administration of irisflorentin significantly inhibited pericyte apoptosis, protected BSCB integrity, promoted axonal growth, and ultimately improved locomotion recovery in a rat model of SCI. In vitro, we found that the positive effects of irisflorentin on axonal growth were likely to be mediated by regulating the crosstalk between pericytes and neurons. Furthermore, irisflorentin effectively ameliorated ER stress caused by incubation with thapsigargin (TG) in pericytes. Meanwhile, the protective effect of irisflorentin on BSCB disruption is strongly related to the reduction of pericyte apoptosis via inhibition of ER stress. Collectively, our findings demonstrate that irisflorentin is beneficial for functional recovery after SCI and that pericytes are a valid target of interest for future SCI therapies.</p></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141598909","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
Cold inducible RNA binding protein-regulated mitochondria associated endoplasmic reticulum membranes-mediated Ca2+ transport play a critical role in hypothermia cerebral resuscitation 冷诱导 RNA 结合蛋白调控线粒体相关内质网膜介导的 Ca2+ 转运在低体温脑复苏中发挥关键作用。
IF 4.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2024-07-09 DOI: 10.1016/j.expneurol.2024.114883
Yu Gao , Haoxin Liu , Yaqing Zhou , Shenquan Cai , Jie Zhang , Jie Sun , Manlin Duan

Cardiac arrest is a global health issue causing more deaths than many other diseases. Hypothermia therapy is commonly used to treat secondary brain injury resulting from cardiac arrest. Previous studies have shown that CIRP is induced in specific brain regions during hypothermia and inhibits mitochondrial apoptotic factors. However, the specific mechanisms by which hypothermia-induced CIRP exerts its anti-apoptotic effect are still unknown. This study aims to investigate the role of Cold-inducible RNA-binding protein (CIRP) in mitochondrial-associated endoplasmic reticulum membrane (MAM)-mediated Ca2+ transport during hypothermic brain resuscitation.We constructed a rat model of cardiac arrest and resuscitation and hippocampal neuron oxygen-glucose deprivation/reoxygenation model. We utilized shRNA transfection to interfere the expression of CIRP and observe the effect of CIRP on the structure and function of MAM.Hypothermia induced CIRP can reduce the apoptosis of hippocampal neurons, and improve the survival rate of rats. Hypothermia induced CIRP can reduce the expressions of calcium transporters IP3R and VDAC1 in MAM, reduce the concentration of calcium in mitochondria, decrease the expression of ROS, and stabilize the mitochondrial membrane potential. Immunofluorescence and immunocoprecipitation showed that CIRP could directly interact with IP3R-VDAC1 complex, thereby changing the structure of MAM, inhibiting calcium transportation and improving mitochondrial function in vivo and vitro.Both in vivo and in vitro experiments have confirmed that hypothermia induced CIRP can act on the calcium channel IP3R-VDAC1 in MAM, reduce the calcium overload in mitochondria, improve the energy metabolism of mitochondria, and thus play a role in neuron resuscitation. This study contributes to understanding hypothermia therapy and identifies potential targets for brain injury treatment.

心脏骤停是一个全球性的健康问题,造成的死亡人数超过了许多其他疾病。低温疗法通常用于治疗心脏骤停导致的继发性脑损伤。先前的研究表明,低体温时会在特定脑区诱导 CIRP,并抑制线粒体凋亡因子。然而,低体温诱导的 CIRP 发挥抗凋亡作用的具体机制尚不清楚。本研究旨在探讨冷诱导 RNA 结合蛋白(CIRP)在低体温脑复苏过程中线粒体相关内质网膜(MAM)介导的 Ca2+ 转运中的作用。利用 shRNA 转染干扰 CIRP 的表达,观察 CIRP 对 MAM 结构和功能的影响。低温诱导 CIRP 可降低 MAM 中钙转运体 IP3R 和 VDAC1 的表达,降低线粒体中钙的浓度,减少 ROS 的表达,稳定线粒体膜电位。免疫荧光和免疫沉淀显示,CIRP 可直接与 IP3R-VDAC1 复合物相互作用,从而改变 MAM 的结构,抑制钙离子运输,改善体内和体外线粒体功能。体内和体外实验均证实,低体温诱导的 CIRP 可作用于 MAM 中的钙通道 IP3R-VDAC1,减轻线粒体中的钙超载,改善线粒体的能量代谢,从而在神经元复苏中发挥作用。这项研究有助于理解低温疗法,并确定脑损伤治疗的潜在靶点。
{"title":"Cold inducible RNA binding protein-regulated mitochondria associated endoplasmic reticulum membranes-mediated Ca2+ transport play a critical role in hypothermia cerebral resuscitation","authors":"Yu Gao ,&nbsp;Haoxin Liu ,&nbsp;Yaqing Zhou ,&nbsp;Shenquan Cai ,&nbsp;Jie Zhang ,&nbsp;Jie Sun ,&nbsp;Manlin Duan","doi":"10.1016/j.expneurol.2024.114883","DOIUrl":"10.1016/j.expneurol.2024.114883","url":null,"abstract":"<div><p>Cardiac arrest is a global health issue causing more deaths than many other diseases. Hypothermia therapy is commonly used to treat secondary brain injury resulting from cardiac arrest. Previous studies have shown that CIRP is induced in specific brain regions during hypothermia and inhibits mitochondrial apoptotic factors. However, the specific mechanisms by which hypothermia-induced CIRP exerts its anti-apoptotic effect are still unknown. This study aims to investigate the role of Cold-inducible RNA-binding protein (CIRP) in mitochondrial-associated endoplasmic reticulum membrane (MAM)-mediated Ca<sup>2+</sup> transport during hypothermic brain resuscitation.We constructed a rat model of cardiac arrest and resuscitation and hippocampal neuron oxygen-glucose deprivation/reoxygenation model. We utilized shRNA transfection to interfere the expression of CIRP and observe the effect of CIRP on the structure and function of MAM.Hypothermia induced CIRP can reduce the apoptosis of hippocampal neurons, and improve the survival rate of rats. Hypothermia induced CIRP can reduce the expressions of calcium transporters IP<sub>3</sub>R and VDAC1 in MAM, reduce the concentration of calcium in mitochondria, decrease the expression of ROS, and stabilize the mitochondrial membrane potential. Immunofluorescence and immunocoprecipitation showed that CIRP could directly interact with IP<sub>3</sub>R-VDAC1 complex, thereby changing the structure of MAM, inhibiting calcium transportation and improving mitochondrial function in vivo and vitro.Both in vivo and in vitro experiments have confirmed that hypothermia induced CIRP can act on the calcium channel IP<sub>3</sub>R-VDAC1 in MAM, reduce the calcium overload in mitochondria, improve the energy metabolism of mitochondria, and thus play a role in neuron resuscitation. This study contributes to understanding hypothermia therapy and identifies potential targets for brain injury treatment.</p></div>","PeriodicalId":12246,"journal":{"name":"Experimental Neurology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141590042","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