首页 > 最新文献

Brain Research Bulletin最新文献

英文 中文
Bleomycin triggers chronic mechanical nociception by activating TRPV1 and glial reaction-mediated neuroinflammation via TSLP/TSLPR/pSTAT5 signals
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-09-12 DOI: 10.1016/j.brainresbull.2024.111081

Chronic pain is a universal public health problem with nearly one third of global human involved, which causes significant distressing personal burden. After painful stimulus, neurobiological changes occur not only in peripheral nervous system but also in central nervous system where somatosensory cortex is important for nociception. Being an ion channel, transient receptor potential vanilloid 1 (TRPV1) act as an inflammatory detector in the brain. Thymic stromal lymphopoietin (TSLP) is a potent neuroinflammation mediator after nerve injury. Bleomycin is applied to treat dermatologic diseases, and its administration elicits local painful sensation. However, whether bleomycin administration can cause chronic pain remains unknown. In the present study, we aimed to investigate how mice develop chronic pain after receiving repeated bleomycin administration. In addition, the relevant neurobiological brain changes after noxious stimuli were clarified. C57BL/6 mice aged five- to six-weeks were randomly classified into two group, PBS (normal) group and bleomycin group which bleomycin was intradermally administered to back five times a week over a three-week period. Calibrated forceps testing was used to measure mouse pain threshold. Western blots were used to assess neuroinflammatory response; immunofluorescence assay was used to measure the status of neuron apoptosis, glial reaction, and neuro-glial communication. Bleomycin administration induced mechanical nociception and activated both TRPV1 and TSLP/TSLPR/pSTAT5 signals in mouse somatosensory cortex. Through these pathways, bleomycin not only activates glial reaction but also causes neuronal apoptosis. TRPV1 and TSLP/TSLPR/pSTAT5 signaling had co-labeled each other by immunofluorescence assay. Taken together, our study provides a new chronic pain model by repeated intradermal bleomycin injection by activating TRPV1 and glial reaction-mediated neuroinflammation via TSLP/TSLPR/pSTAT5 signals.

{"title":"Bleomycin triggers chronic mechanical nociception by activating TRPV1 and glial reaction-mediated neuroinflammation via TSLP/TSLPR/pSTAT5 signals","authors":"","doi":"10.1016/j.brainresbull.2024.111081","DOIUrl":"10.1016/j.brainresbull.2024.111081","url":null,"abstract":"<div><p>Chronic pain is a universal public health problem with nearly one third of global human involved, which causes significant distressing personal burden. After painful stimulus, neurobiological changes occur not only in peripheral nervous system but also in central nervous system where somatosensory cortex is important for nociception. Being an ion channel, transient receptor potential vanilloid 1 (TRPV1) act as an inflammatory detector in the brain. Thymic stromal lymphopoietin (TSLP) is a potent neuroinflammation mediator after nerve injury. Bleomycin is applied to treat dermatologic diseases, and its administration elicits local painful sensation. However, whether bleomycin administration can cause chronic pain remains unknown. In the present study, we aimed to investigate how mice develop chronic pain after receiving repeated bleomycin administration. In addition, the relevant neurobiological brain changes after noxious stimuli were clarified. C57BL/6 mice aged five- to six-weeks were randomly classified into two group, PBS (normal) group and bleomycin group which bleomycin was intradermally administered to back five times a week over a three-week period. Calibrated forceps testing was used to measure mouse pain threshold. Western blots were used to assess neuroinflammatory response; immunofluorescence assay was used to measure the status of neuron apoptosis, glial reaction, and neuro-glial communication. Bleomycin administration induced mechanical nociception and activated both TRPV1 and TSLP/TSLPR/pSTAT5 signals in mouse somatosensory cortex. Through these pathways, bleomycin not only activates glial reaction but also causes neuronal apoptosis. TRPV1 and TSLP/TSLPR/pSTAT5 signaling had co-labeled each other by immunofluorescence assay. Taken together, our study provides a new chronic pain model by repeated intradermal bleomycin injection by activating TRPV1 and glial reaction-mediated neuroinflammation via TSLP/TSLPR/pSTAT5 signals.</p></div>","PeriodicalId":9302,"journal":{"name":"Brain Research Bulletin","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0361923024002156/pdfft?md5=b09749f9a94f8305bbcf88ca702a7b95&pid=1-s2.0-S0361923024002156-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142243777","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Quercetin alleviates microglial-induced inflammation after traumatic brain injury via the PGC-1α/Nrf2 pathway dependent on HDAC3 inhibition
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-09-12 DOI: 10.1016/j.brainresbull.2024.111080

Inflammation and neuronal apoptosis play a key role in traumatic brain injury (TBI). Quercetin (Que) has been shown to exhibit a neuroprotective effect after TBI, but the underlying molecular mechanism remains unclear. In this study, We established a weight-drop mouse model to illustrate the effects of Que on microglial-induced inflammation in TBI. Mice were divided into four groups: the Sham group, TBI group, TBI+vehicle group, and TBI+Que group. The TBI+Que group was treated with Que 30 min after TBI. Brain water content, neurological score, and neuronal apoptosis were measured. Western blotting, TUNEL staining, Nissl staining, quantitative polymerase chain reaction, and immunofluorescence staining were performed to assess the activation of the PGC-1α/Nrf2 pathway and nuclear translocation of HDAC3 with Que treatment. The results showed that Que administration alleviated TBI-induced neurobehavioral deficits, encephaledema, and neuron apoptosis. Que also restrained TBI-induced microglial activity and the subsequent expression of the inflammatory factor in the contusion cortex. Moreover, Que treatment activated the PGC-1α/Nrf2 pathway, attributable to the inhibition of HDAC3 translocation to the nucleus. Overall, these results reveal the role of Que in protecting against TBI-induced neuroinflammation and promoting neurological functional recovery, which is achieved through the negative regulation of HDAC3.

炎症和神经细胞凋亡在创伤性脑损伤(TBI)中起着关键作用。槲皮素(Que)已被证明在创伤性脑损伤后具有神经保护作用,但其潜在的分子机制仍不清楚。在本研究中,我们建立了一个体重下降的小鼠模型,以说明阙对创伤性脑损伤中小胶质细胞诱导的炎症的影响。小鼠被分为四组:Sham 组、TBI 组、TBI+车辆组和 TBI+Que 组。TBI+Que 组在 TBI 后 30 分钟用 Que 治疗。测量脑含水量、神经系统评分和神经细胞凋亡。通过 Western 印迹、TUNEL 染色、Nissl 染色、定量聚合酶链反应和免疫荧光染色来评估阙治疗对 PGC-1α/Nrf2 通路的激活和 HDAC3 的核转位。结果表明,服用阙可减轻创伤性脑损伤引起的神经行为障碍、脑水肿和神经元凋亡。阙还抑制了创伤性脑损伤诱发的小胶质细胞活性以及随后在挫伤皮层中炎症因子的表达。此外,阙治疗激活了 PGC-1α/Nrf2 通路,这归因于抑制了 HDAC3 转位至细胞核。总之,这些结果揭示了阙在防止创伤性脑损伤引起的神经炎症和促进神经功能恢复中的作用,而这是通过对 HDAC3 的负调控实现的。
{"title":"Quercetin alleviates microglial-induced inflammation after traumatic brain injury via the PGC-1α/Nrf2 pathway dependent on HDAC3 inhibition","authors":"","doi":"10.1016/j.brainresbull.2024.111080","DOIUrl":"10.1016/j.brainresbull.2024.111080","url":null,"abstract":"<div><p>Inflammation and neuronal apoptosis play a key role in traumatic brain injury (TBI). Quercetin (Que) has been shown to exhibit a neuroprotective effect after TBI, but the underlying molecular mechanism remains unclear. In this study, We established a weight-drop mouse model to illustrate the effects of Que on microglial-induced inflammation in TBI. Mice were divided into four groups: the Sham group, TBI group, TBI+vehicle group, and TBI+Que group. The TBI+Que group was treated with Que 30 min after TBI. Brain water content, neurological score, and neuronal apoptosis were measured. Western blotting, TUNEL staining, Nissl staining, quantitative polymerase chain reaction, and immunofluorescence staining were performed to assess the activation of the PGC-1α/Nrf2 pathway and nuclear translocation of HDAC3 with Que treatment. The results showed that Que administration alleviated TBI-induced neurobehavioral deficits, encephaledema, and neuron apoptosis. Que also restrained TBI-induced microglial activity and the subsequent expression of the inflammatory factor in the contusion cortex. Moreover, Que treatment activated the PGC-1α/Nrf2 pathway, attributable to the inhibition of HDAC3 translocation to the nucleus. Overall, these results reveal the role of Que in protecting against TBI-induced neuroinflammation and promoting neurological functional recovery, which is achieved through the negative regulation of HDAC3.</p></div>","PeriodicalId":9302,"journal":{"name":"Brain Research Bulletin","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0361923024002144/pdfft?md5=9f5ac149037d4f07fa11df7a94fbb158&pid=1-s2.0-S0361923024002144-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142243773","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Projections from the ventral tegmental area to zona incerta regulate fear generalization in a mouse model of PTSD 在创伤后应激障碍小鼠模型中,从腹侧被盖区向内侧区的投射调节恐惧泛化
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-09-11 DOI: 10.1016/j.brainresbull.2024.111079

Generalized fear is a maladaptive behavior in which non-threatening stimuli elicit a fearful response. The ventral tegmental area (VTA) has been demonstrated to play important roles in fear response and fear memory generalization, but the precious neural circuit mechanism is still unclear. Here, we demonstrated that VTA-zona incerta (ZI) glutamatergic projection is involved in regulating high-intensity threatening training induced generalization and anxiety. Combining calcium signal recording and chemogentics, our work reveals that VTA glutamatergic neurons respond to closed arm entering in the model of PTSD. Inhibition of VTA glutamatergic neurons or the glutamatergic projection to ZI could both relieve fear generalization and anxiety. Together, our study proves the VTA - ZI glutamatergic circuit is involved in mediating fear generalization and anxiety, and provides a potential target for treating post-traumatic stress disorder.

泛化恐惧是一种适应不良行为,即非威胁性刺激引起恐惧反应。腹侧被盖区(VTA)已被证实在恐惧反应和恐惧记忆泛化中发挥重要作用,但其重要的神经回路机制仍不清楚。在这里,我们证明了VTA-zona incerta(ZI)谷氨酸能投射参与调节高强度威胁训练诱导的泛化和焦虑。结合钙信号记录和化学效应,我们的研究揭示了在创伤后应激障碍模型中,VTA 谷氨酸能神经元对手臂闭合进入有反应。抑制VTA谷氨酸能神经元或谷氨酸能投射到ZI都能缓解恐惧泛化和焦虑。总之,我们的研究证明了VTA-ZI谷氨酸能回路参与介导恐惧泛化和焦虑,并为治疗创伤后应激障碍提供了一个潜在的靶点。
{"title":"Projections from the ventral tegmental area to zona incerta regulate fear generalization in a mouse model of PTSD","authors":"","doi":"10.1016/j.brainresbull.2024.111079","DOIUrl":"10.1016/j.brainresbull.2024.111079","url":null,"abstract":"<div><p>Generalized fear is a maladaptive behavior in which non-threatening stimuli elicit a fearful response. The ventral tegmental area (VTA) has been demonstrated to play important roles in fear response and fear memory generalization, but the precious neural circuit mechanism is still unclear. Here, we demonstrated that VTA-zona incerta (ZI) glutamatergic projection is involved in regulating high-intensity threatening training induced generalization and anxiety. Combining calcium signal recording and chemogentics, our work reveals that VTA glutamatergic neurons respond to closed arm entering in the model of PTSD. Inhibition of VTA glutamatergic neurons or the glutamatergic projection to ZI could both relieve fear generalization and anxiety. Together, our study proves the VTA - ZI glutamatergic circuit is involved in mediating fear generalization and anxiety, and provides a potential target for treating post-traumatic stress disorder.</p></div>","PeriodicalId":9302,"journal":{"name":"Brain Research Bulletin","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0361923024002132/pdfft?md5=1a3a27457bbf0fbde1b93b06854a7cca&pid=1-s2.0-S0361923024002132-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142232530","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GDNF facilitates cognitive function recovery following neonatal surgical-induced learning and memory impairment via activation of the RET pathway and modulation of downstream effectors PKMζ and Kalirin in rats
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-09-11 DOI: 10.1016/j.brainresbull.2024.111078

Objective

The aim of this study is to elucidate the underlying mechanism through which glial cell line-derived neurotrophic factor (GDNF) improves cognitive deficits in adults resulting from neonatal surgical interventions.

Methods

Newborn Sprague–Dawley rats, regardless of gender, were randomly allocated into seven groups on postnatal day 7 as follows (n=15): (1) Control group (not subjected to anesthesia, surgery, or any pharmaceutical interventions); (2) GDNF group (received intracerebroventricular injection of GDNF); (3) Surgery group (underwent right carotid artery exposure under anesthesia with 3 % sevoflurane); (4) Surgery plus GDNF group; (5) Surgery plus GDNF and type II JAK inhibitor NVP-BBT594 (BBT594) group (administered intraperitoneal injection of BBT594); (6) BBT group; and (7) Surgery plus BBT group. Starting from postnatal day 33, all rats underwent Barnes maze and fear conditioning tests, followed by decapitation under sevoflurane anesthesia for subsequent analyses. The left hemibrains underwent Golgi staining, while the right hemibrains were used for hippocampal protein extraction to assess Protein kinase Mζ (PKMζ) and Kalirin expression through western blotting.

Results

GDNF demonstrated a mitigating effect on spatial learning and memory impairment, as well as context-related fear memory impairment, reductions in dendritic total lengths, and spinal density within the hippocampus induced by surgical intervention. Notably, all of these ameliorative effects of GDNF were reversed upon administration of the RET inhibitor BBT594. Additionally, GDNF alleviated the downregulation of protein expression of PKMζ and Kalirin in the hippocampus of rats subjected to surgery, subsequently reversed by BBT594.

Conclusion

The effective impact of GDNF on learning and memory impairment caused by surgical intervention appears to be mediated through the RET pathway. Moreover, GDNF may exert its influence by upregulating the expression of PKMζ and Kalirin, consequently enhancing the development of dendrites and dendritic spines.

{"title":"GDNF facilitates cognitive function recovery following neonatal surgical-induced learning and memory impairment via activation of the RET pathway and modulation of downstream effectors PKMζ and Kalirin in rats","authors":"","doi":"10.1016/j.brainresbull.2024.111078","DOIUrl":"10.1016/j.brainresbull.2024.111078","url":null,"abstract":"<div><h3>Objective</h3><p>The aim of this study is to elucidate the underlying mechanism through which glial cell line-derived neurotrophic factor (GDNF) improves cognitive deficits in adults resulting from neonatal surgical interventions.</p></div><div><h3>Methods</h3><p>Newborn Sprague–Dawley rats, regardless of gender, were randomly allocated into seven groups on postnatal day 7 as follows (<em>n</em>=15): (1) Control group (not subjected to anesthesia, surgery, or any pharmaceutical interventions); (2) GDNF group (received intracerebroventricular injection of GDNF); (3) Surgery group (underwent right carotid artery exposure under anesthesia with 3 % sevoflurane); (4) Surgery plus GDNF group; (5) Surgery plus GDNF and type II JAK inhibitor NVP-BBT594 (BBT594) group (administered intraperitoneal injection of BBT594); (6) BBT group; and (7) Surgery plus BBT group. Starting from postnatal day 33, all rats underwent Barnes maze and fear conditioning tests, followed by decapitation under sevoflurane anesthesia for subsequent analyses. The left hemibrains underwent Golgi staining, while the right hemibrains were used for hippocampal protein extraction to assess Protein kinase Mζ (PKMζ) and Kalirin expression through western blotting.</p></div><div><h3>Results</h3><p>GDNF demonstrated a mitigating effect on spatial learning and memory impairment, as well as context-related fear memory impairment, reductions in dendritic total lengths, and spinal density within the hippocampus induced by surgical intervention. Notably, all of these ameliorative effects of GDNF were reversed upon administration of the RET inhibitor BBT594. Additionally, GDNF alleviated the downregulation of protein expression of PKMζ and Kalirin in the hippocampus of rats subjected to surgery, subsequently reversed by BBT594.</p></div><div><h3>Conclusion</h3><p>The effective impact of GDNF on learning and memory impairment caused by surgical intervention appears to be mediated through the RET pathway. Moreover, GDNF may exert its influence by upregulating the expression of PKMζ and Kalirin, consequently enhancing the development of dendrites and dendritic spines.</p></div>","PeriodicalId":9302,"journal":{"name":"Brain Research Bulletin","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0361923024002120/pdfft?md5=3beff9a5217a66fb993188060c8fcb4c&pid=1-s2.0-S0361923024002120-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142243776","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Sodium aescinate alleviates neuropathic pain through suppressing OGT-mediated O-GlcNAc modification of TLR3 to inactivate MAPK signaling pathway
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-09-11 DOI: 10.1016/j.brainresbull.2024.111077

Neuropathic pain results from damage to nerves or the brain, and is characterized by symptoms such as allodynia, spontaneous pain, and hyperalgesia. The causes of this type of pain are intricate, which can make it difficult to treat. Sodium aescinate (SA), a natural extract from horse chestnut tree seeds, has been shown to act as a neuroprotector by inhibiting microglia activation. This study aims to explore the therapeutic potential of SA for neuropathic pain and the molecular mechanisms regulated by SA treatment. Through in vivo animal models and experiments, we found that SA treatment significantly reduced mechanical allodynia and heat hyperalgesia in neuropathic pain models. Additionally, SA inhibited O-GlcNAc-transferase (OGT)-induced O-GlcNAcylation (O-GlcNAc) modification in neuropathic pain mice. OGT overexpression could impede the therapeutic effects of SA on neuropathic pain. Further investigation revealed that Toll-like receptor 3 (TLR3), stabilized by OGT-induced O-GlcNAc modification, could activate the Mitogen activated protein kinase (MAPK) signaling pathway. Further in vivo experiments demonstrated that TLR3-mediated p38 mitogen-activated protein kinase (p38MAPK) activation is involved in SA-mediated relief of neuropathic pain. In conclusion, this study uncovers a novel molecular pathway deactivated by SA treatment in neuropathic pain.

神经病理性疼痛由神经或大脑受损引起,以异痛症、自发性疼痛和痛觉过敏等症状为特征。这种疼痛的原因错综复杂,因此很难治疗。从七叶树种子中提取的天然提取物栎氨酸钠(SA)已被证明可抑制小胶质细胞的活化,从而起到保护神经的作用。本研究旨在探索 SA 对神经病理性疼痛的治疗潜力以及 SA 治疗调控的分子机制。通过体内动物模型和实验,我们发现 SA 治疗可显著减轻神经病理性疼痛模型中的机械异感和热痛。此外,SA还能抑制O-GlcNAc-转移酶(OGT)诱导的神经病理性疼痛小鼠的O-GlcNAc(O-GlcNAc)修饰。OGT 过度表达会阻碍 SA 对神经病理性疼痛的治疗效果。进一步研究发现,通过 OGT 诱导的 O-GlcNAc 修饰稳定的 Toll 样受体 3(TLR3)可激活丝裂原活化蛋白激酶(MAPK)信号通路。进一步的体内实验证明,TLR3 介导的 p38 丝裂原活化蛋白激酶(p38MAPK)激活参与了 SA 介导的神经病理性疼痛缓解。总之,本研究发现了一种新型分子通路,通过 SA 治疗神经病理性疼痛可使其失活。
{"title":"Sodium aescinate alleviates neuropathic pain through suppressing OGT-mediated O-GlcNAc modification of TLR3 to inactivate MAPK signaling pathway","authors":"","doi":"10.1016/j.brainresbull.2024.111077","DOIUrl":"10.1016/j.brainresbull.2024.111077","url":null,"abstract":"<div><p>Neuropathic pain results from damage to nerves or the brain, and is characterized by symptoms such as allodynia, spontaneous pain, and hyperalgesia. The causes of this type of pain are intricate, which can make it difficult to treat. Sodium aescinate (SA), a natural extract from horse chestnut tree seeds, has been shown to act as a neuroprotector by inhibiting microglia activation. This study aims to explore the therapeutic potential of SA for neuropathic pain and the molecular mechanisms regulated by SA treatment. Through in vivo animal models and experiments, we found that SA treatment significantly reduced mechanical allodynia and heat hyperalgesia in neuropathic pain models. Additionally, SA inhibited O-GlcNAc-transferase (OGT)-induced O-GlcNAcylation (O-GlcNAc) modification in neuropathic pain mice. OGT overexpression could impede the therapeutic effects of SA on neuropathic pain. Further investigation revealed that Toll-like receptor 3 (TLR3), stabilized by OGT-induced O-GlcNAc modification, could activate the Mitogen activated protein kinase (MAPK) signaling pathway. Further in vivo experiments demonstrated that TLR3-mediated p38 mitogen-activated protein kinase (p38MAPK) activation is involved in SA-mediated relief of neuropathic pain. In conclusion, this study uncovers a novel molecular pathway deactivated by SA treatment in neuropathic pain.</p></div>","PeriodicalId":9302,"journal":{"name":"Brain Research Bulletin","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0361923024002119/pdfft?md5=b39776d4ea906cc2254572a35bb8b3e6&pid=1-s2.0-S0361923024002119-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142243774","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Neuregulin-1 immunoreactivity in peripheral plasma is associated with rs6982890 polymorphism-mediated psychotic symptoms in schizophrenia 外周血浆中的神经胶质蛋白-1免疫活性与rs6982890多态性介导的精神分裂症精神病性症状有关
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-09-08 DOI: 10.1016/j.brainresbull.2024.111075

Objectives

Neuregulin 1 (NRG1) is a risk gene for schizophrenia and involved in neurodevelopment and synaptic plasticity. Polymorphisms in NRG1 may affect psychotic symptoms in schizophrenia. This study investigated the effects of the single nucleotide polymorphism (SNP) rs6982890 on peripheral plasma NRG1 immunoreactivity, clinical symptoms and cognitive functions in schizophrenia patients.

Material and methods

We recruited subjects from the Han population of northern China from 2010 to 2022. We first genotyped and analyzed 6 NRG1 SNPS in 1304 patients with schizophrenia and 871 healthy controls. Then, 91 patients with schizophrenia and 40 healthy controls were selected to detect the peripheral plasma NRG1 immunoreactivity by ELISA. Among them, 84 patients were divided into rs6982890 genotypes to analyze the correlation between NRG1 immunoreactivity and clinical symptoms.

Results

Rs6982890 allelic frequencies were statistically significant between patients and controls. Baseline peripheral plasma NRG1 immunoreactivity in patients were significantly lower than controls. NRG1 immunoreactivity in patients were significantly increased after 8 weeks of antipsychotic treatment and significantly correlated with clinical symptoms and cognitive function. Genotyping of patients with SNP rs6982890 indicated NRG1 immunoreactivity in CC genotype increased significantly after treatment, while CT genotype had no significant change. Baseline NRG1 immunoreactivity with the CT genotype were significantly higher than CC genotype.

Conclusions

NRG1 SNP rs6982890 is significantly associated with schizophrenia in the Han population of northern China, and it may affect the effect of antipsychotic drug treatment by regulating the peripheral plasma NRG1 immunoreactivity.

目的神经胶质蛋白 1(NRG1)是精神分裂症的风险基因,参与神经发育和突触可塑性。NRG1 的多态性可能会影响精神分裂症患者的精神症状。本研究探讨了单核苷酸多态性(SNP)rs6982890对精神分裂症患者外周血浆NRG1免疫活性、临床症状和认知功能的影响。我们首先对1304名精神分裂症患者和871名健康对照者的6个NRG1 SNPS进行了基因分型和分析。然后,我们选取了91名精神分裂症患者和40名健康对照者,用ELISA方法检测外周血浆NRG1免疫反应。其中,84名患者被分为rs6982890基因型,以分析NRG1免疫反应与临床症状之间的相关性。患者的基线外周血浆 NRG1 免疫活性明显低于对照组。抗精神病药物治疗8周后,患者的NRG1免疫活性明显升高,并与临床症状和认知功能明显相关。对患者进行的 SNP rs6982890 基因分型显示,CC 基因型患者的 NRG1 免疫反应性在治疗后明显增加,而 CT 基因型则无明显变化。结论在中国北方汉族人群中,NRG1 SNP rs6982890与精神分裂症显著相关,它可能通过调节外周血浆NRG1免疫反应影响抗精神病药物的治疗效果。
{"title":"Neuregulin-1 immunoreactivity in peripheral plasma is associated with rs6982890 polymorphism-mediated psychotic symptoms in schizophrenia","authors":"","doi":"10.1016/j.brainresbull.2024.111075","DOIUrl":"10.1016/j.brainresbull.2024.111075","url":null,"abstract":"<div><h3>Objectives</h3><p>Neuregulin 1 (<em>NRG1</em>) is a risk gene for schizophrenia and involved in neurodevelopment and synaptic plasticity. Polymorphisms in <em>NRG1</em> may affect psychotic symptoms in schizophrenia. This study investigated the effects of the single nucleotide polymorphism (SNP) rs6982890 on peripheral plasma NRG1 immunoreactivity, clinical symptoms and cognitive functions in schizophrenia patients.</p></div><div><h3>Material and methods</h3><p>We recruited subjects from the Han population of northern China from 2010 to 2022. We first genotyped and analyzed 6 NRG1 SNPS in 1304 patients with schizophrenia and 871 healthy controls. Then, 91 patients with schizophrenia and 40 healthy controls were selected to detect the peripheral plasma NRG1 immunoreactivity by ELISA. Among them, 84 patients were divided into rs6982890 genotypes to analyze the correlation between NRG1 immunoreactivity and clinical symptoms.</p></div><div><h3>Results</h3><p>Rs6982890 allelic frequencies were statistically significant between patients and controls. Baseline peripheral plasma NRG1 immunoreactivity in patients were significantly lower than controls. NRG1 immunoreactivity in patients were significantly increased after 8 weeks of antipsychotic treatment and significantly correlated with clinical symptoms and cognitive function. Genotyping of patients with SNP rs6982890 indicated NRG1 immunoreactivity in CC genotype increased significantly after treatment, while CT genotype had no significant change. Baseline NRG1 immunoreactivity with the CT genotype were significantly higher than CC genotype.</p></div><div><h3>Conclusions</h3><p><em>NRG1</em> SNP rs6982890 is significantly associated with schizophrenia in the Han population of northern China, and it may affect the effect of antipsychotic drug treatment by regulating the peripheral plasma NRG1 immunoreactivity.</p></div>","PeriodicalId":9302,"journal":{"name":"Brain Research Bulletin","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-09-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0361923024002090/pdfft?md5=f95059a27ca47425033f386d9c067055&pid=1-s2.0-S0361923024002090-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142172938","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Prediction models of the aphasia severity after stroke by lesion load of cortical language areas and white matter tracts: An atlas-based study 根据大脑皮层语言区和白质层的病变负荷预测脑卒中后失语症严重程度的模型:基于图谱的研究
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-09-07 DOI: 10.1016/j.brainresbull.2024.111074

Objective

To construct relatively objective, atlas-based multivariate models for predicting early aphasia severity after stroke, using structural magnetic resonance imaging.

Methods

We analyzed the clinical and imaging data of 46 patients with post-stroke aphasia. The aphasia severity was identified with a Western Aphasia Battery Aphasia Quotient. The assessments of stroke lesions were indicated by the lesion load of both the cortical language areas (Areas-LL) and four white matter tracts (i.e., the superior longitudinal fasciculus, SLF-LL; the inferior frontal occipital fasciculi, IFOF-LL; the inferior longitudinal, ILF-LL; and the uncinate fasciculi, UF-LL) extracted from human brain atlas. Correlation analyses and multiple linear regression analyses were conducted to evaluate the correlations between demographic, stroke- and lesion-related variables and aphasia severity. The predictive models were then established according to the identified significant variables. Finally, the receiver operating characteristic (ROC) curve was utilized to assess the accuracy of the predictive models.

Results

The variables including Areas-LL, the SLF-LL, and the IFOF-LL were significantly negatively associated with aphasia severity (p < 0.05). In multiple linear regression analyses, these variables accounted for 59.4 % of the variance (p < 0.05). The ROC curve analyses yielded the validated area under the curve (AUC) 0.84 both for Areas-LL and SLF-LL and 0.76 for IFOF-LL, indicating good predictive performance (p < 0.01). Adding the combination of SLF-LL and IFOF-LL to this model increased the explained variance to 62.6 % and the AUC to 0.92.

Conclusions

The application of atlas-based multimodal lesion assessment may help predict the aphasia severity after stroke, which needs to be further validated and generalized for the prediction of more outcome measures in populations with various brain injuries.

目的利用结构性磁共振成像构建相对客观的、基于图谱的多变量模型,用于预测中风后早期失语症的严重程度:我们分析了 46 名卒中后失语患者的临床和影像学数据。方法:我们对 46 名中风后失语患者的临床和影像学数据进行了分析,失语严重程度通过西方失语测验(Western Aphasia Battery Aphasia Quotient)来确定。对脑卒中病变的评估是通过从人脑图谱中提取的大脑皮质语言区(Area-LL)和四条白质束(即上纵筋束,SLF-LL;下额枕筋束,IFOF-LL;下纵筋束,ILF-LL;钩状筋束,UF-LL)的病变负荷来进行的。通过相关性分析和多元线性回归分析来评估人口统计学、卒中和病变相关变量与失语症严重程度之间的相关性。然后根据确定的重要变量建立预测模型。最后,利用接收者操作特征曲线(ROC)评估预测模型的准确性:结果:Areas-LL、SLF-LL 和 IFOF-LL 等变量与失语症严重程度呈显著负相关(P < 0.05)。在多元线性回归分析中,这些变量占方差的 59.4%(p < 0.05)。通过 ROC 曲线分析,Areas-LL 和 SLF-LL 的有效曲线下面积 (AUC) 均为 0.84,IFOF-LL 为 0.76,表明预测效果良好(p < 0.01)。在该模型中加入 SLF-LL 和 IFOF-LL 组合后,解释方差增加到 62.6%,AUC 增加到 0.92:基于地图集的多模态病变评估可帮助预测脑卒中后失语症的严重程度,该方法需要进一步验证和推广,以预测各种脑损伤人群的更多结果。
{"title":"Prediction models of the aphasia severity after stroke by lesion load of cortical language areas and white matter tracts: An atlas-based study","authors":"","doi":"10.1016/j.brainresbull.2024.111074","DOIUrl":"10.1016/j.brainresbull.2024.111074","url":null,"abstract":"<div><h3>Objective</h3><p>To construct relatively objective, atlas-based multivariate models for predicting early aphasia severity after stroke, using structural magnetic resonance imaging.</p></div><div><h3>Methods</h3><p>We analyzed the clinical and imaging data of 46 patients with post-stroke aphasia. The aphasia severity was identified with a Western Aphasia Battery Aphasia Quotient. The assessments of stroke lesions were indicated by the lesion load of both the cortical language areas (Areas-LL) and four white matter tracts (i.e., the superior longitudinal fasciculus, SLF-LL; the inferior frontal occipital fasciculi, IFOF-LL; the inferior longitudinal, ILF-LL; and the uncinate fasciculi, UF-LL) extracted from human brain atlas. Correlation analyses and multiple linear regression analyses were conducted to evaluate the correlations between demographic, stroke- and lesion-related variables and aphasia severity. The predictive models were then established according to the identified significant variables. Finally, the receiver operating characteristic (ROC) curve was utilized to assess the accuracy of the predictive models.</p></div><div><h3>Results</h3><p>The variables including Areas-LL, the SLF-LL, and the IFOF-LL were significantly negatively associated with aphasia severity (p &lt; 0.05). In multiple linear regression analyses, these variables accounted for 59.4 % of the variance (p &lt; 0.05). The ROC curve analyses yielded the validated area under the curve (AUC) 0.84 both for Areas-LL and SLF-LL and 0.76 for IFOF-LL, indicating good predictive performance (p &lt; 0.01). Adding the combination of SLF-LL and IFOF-LL to this model increased the explained variance to 62.6 % and the AUC to 0.92.</p></div><div><h3>Conclusions</h3><p>The application of atlas-based multimodal lesion assessment may help predict the aphasia severity after stroke, which needs to be further validated and generalized for the prediction of more outcome measures in populations with various brain injuries.</p></div>","PeriodicalId":9302,"journal":{"name":"Brain Research Bulletin","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-09-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0361923024002089/pdfft?md5=b5a2f92693bae2f36046063e025c08f6&pid=1-s2.0-S0361923024002089-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142153162","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Role of Glutathione Peroxidase 4 in Neuronal Ferroptosis and Its Therapeutic Potential in Ischemic and Hemorrhagic Stroke. 谷胱甘肽过氧化物酶 4 在神经元铁氧化中的作用及其在缺血性和出血性中风中的治疗潜力
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-09-05 DOI: 10.1016/j.brainresbull.2024.111065
Chao Wei

Ferroptosis is a type of cell death that depends on iron and is driven by lipid peroxidation, playing a crucial role in neuronal death during stroke. A central element in this process is the inactivation of glutathione peroxidase 4 (GPx4), an antioxidant enzyme that helps maintain redox balance by reducing lipid hydroperoxides. This review examines the critical function of GPx4 in controlling neuronal ferroptosis following ischemic and hemorrhagic stroke. We explore the mechanisms through which GPx4 becomes inactivated in various stroke subtypes. In ischemic strokes, excess glutamate depletes glutathione (GSH) and products of hemoglobin breakdown overwhelm GPx4. Studies using genetic models with GPx4 deficiency underscore its vital role in maintaining neuronal survival and function. We also consider new therapeutic approaches to enhance GPx4 activity, including novel small molecule activators, adjustments in GSH metabolism, and selenium supplementation. Additionally, we outline the potential benefits of combining these GPx4-focused strategies with other anti-ferroptotic methods like iron chelation and lipoxygenase inhibition for enhanced neuroprotection. Furthermore, we highlight the significance of understanding the timing of GPx4 inactivation during stroke progression to design effective therapeutic interventions.

铁过氧化是一种依赖于铁的细胞死亡类型,由脂质过氧化驱动,在中风期间的神经元死亡中起着至关重要的作用。这一过程的核心要素是谷胱甘肽过氧化物酶 4(GPx4)的失活,GPx4 是一种抗氧化酶,通过还原脂质氢过氧化物来帮助维持氧化还原平衡。本综述探讨了 GPx4 在缺血性和出血性中风后控制神经元铁氧化的关键功能。我们探讨了各种中风亚型中 GPx4 失活的机制。在缺血性中风中,过量的谷氨酸会消耗谷胱甘肽(GSH),血红蛋白分解产物会使 GPx4 失活。利用缺乏 GPx4 的遗传模型进行的研究强调了 GPx4 在维持神经元存活和功能方面的重要作用。我们还考虑了增强 GPx4 活性的新治疗方法,包括新型小分子激活剂、调整 GSH 代谢和补硒。此外,我们还概述了将这些以 GPx4 为重点的策略与铁螯合剂和脂氧合酶抑制剂等其他抗铁细胞生成方法相结合以增强神经保护的潜在益处。此外,我们还强调了了解中风进展过程中 GPx4 失活的时间对设计有效的治疗干预措施的重要意义。
{"title":"The Role of Glutathione Peroxidase 4 in Neuronal Ferroptosis and Its Therapeutic Potential in Ischemic and Hemorrhagic Stroke.","authors":"Chao Wei","doi":"10.1016/j.brainresbull.2024.111065","DOIUrl":"https://doi.org/10.1016/j.brainresbull.2024.111065","url":null,"abstract":"<p><p>Ferroptosis is a type of cell death that depends on iron and is driven by lipid peroxidation, playing a crucial role in neuronal death during stroke. A central element in this process is the inactivation of glutathione peroxidase 4 (GPx4), an antioxidant enzyme that helps maintain redox balance by reducing lipid hydroperoxides. This review examines the critical function of GPx4 in controlling neuronal ferroptosis following ischemic and hemorrhagic stroke. We explore the mechanisms through which GPx4 becomes inactivated in various stroke subtypes. In ischemic strokes, excess glutamate depletes glutathione (GSH) and products of hemoglobin breakdown overwhelm GPx4. Studies using genetic models with GPx4 deficiency underscore its vital role in maintaining neuronal survival and function. We also consider new therapeutic approaches to enhance GPx4 activity, including novel small molecule activators, adjustments in GSH metabolism, and selenium supplementation. Additionally, we outline the potential benefits of combining these GPx4-focused strategies with other anti-ferroptotic methods like iron chelation and lipoxygenase inhibition for enhanced neuroprotection. Furthermore, we highlight the significance of understanding the timing of GPx4 inactivation during stroke progression to design effective therapeutic interventions.</p>","PeriodicalId":9302,"journal":{"name":"Brain Research Bulletin","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142145255","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Keap1-independent GSK-3β/Nrf2 signaling mediates electroacupuncture inhibition of oxidative stress to induce cerebral ischemia-reperfusion tolerance Keap1依赖性GSK-3β/Nrf2信号介导电针抑制氧化应激以诱导脑缺血再灌注耐受性
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-09-05 DOI: 10.1016/j.brainresbull.2024.111071

Purpose

Cerebral ischemia-reperfusion (CIR) injury is a devastating consequence of stroke characterized by oxidative stress-induced neuronal damage. Electroacupuncture (EA) has emerged as a potential therapeutic intervention for ischemic stroke, but its underlying mechanisms remain incompletely understood. This study aimed to elucidate whether EA exerts anti-oxidative stress effects against CIR injury by modulating the GSK-3β/Nrf2 pathway.

Methods

CIR mouse models were established using the suture-occluded method and underwent EA pretreatment. Cognitive and neurologic function, cerebral infarct volume, and neuronal damage were assessed in mice. Oxidative stress levels and the expression of components of the GSK-3β/Nrf2 pathway in the cerebral cortex were measured. The regulatory effect of GSK-3β on Nrf2 and its role in electroacupuncture to alleviate oxygen-glucose deprivation/reoxygenation (OGD/R)-induced neuronal injury were investigated by modulating GSK-3β expression in HT22 hippocampal neuronal cells and electroacupuncture serum intervention. Ultimately, Nrf2 knockout mice, GSK-3β knockout mice, and wild-type mice treated with TBHQ (an Nrf2 activator) were utilized for further validation.

Results

EA pretreatment improved cognitive impairment and neuronal damage induced by CIR injury. Mechanistically, EA inhibited oxidative stress in the cerebral cortex, manifested by reduced levels of reactive oxygen species and malondialdehyde, along with increased superoxide dismutase activity. Furthermore, EA upregulated the expression of Nrf2 and its downstream antioxidant enzymes HO-1 and NQO1, while Keap1 expression remained unaffected. In vitro, GSK-3β overexpression inhibited the protective effects of EA serum on OGD/R-induced neuronal damage. In vivo, knockout of either Nrf2 or Gsk-3β genes abolished the neuroprotective effects of EA, and TBHQ exerted effects similar to EA, confirming the significant role of GSK-3β/Nrf2 in mediating EA antioxidative effects.

Conclusion

EA exerts antioxidative stress effects against CIR injury by activating the GSK-3β/Nrf2 signaling pathway, independent of Keap1 regulation.

目的:脑缺血再灌注(CIR)损伤是中风的一种破坏性后果,其特点是氧化应激诱发神经元损伤。电针(EA)已成为缺血性中风的一种潜在治疗干预手段,但其潜在机制仍不完全清楚。本研究旨在阐明 EA 是否通过调节 GSK-3β/Nrf2 通路对 CIR 损伤发挥抗氧化应激作用:方法:采用缝合-闭塞法建立 CIR 小鼠模型,并进行 EA 预处理。对小鼠的认知和神经功能、脑梗死体积和神经元损伤进行评估。测量了氧化应激水平和大脑皮层中GSK-3β/Nrf2通路成分的表达。通过调节GSK-3β在HT22海马神经元细胞中的表达和电针血清干预,研究了GSK-3β对Nrf2的调控作用及其在电针缓解氧-葡萄糖剥夺/复氧(OGD/R)诱导的神经元损伤中的作用。最终,Nrf2基因剔除小鼠、GSK-3β基因剔除小鼠和野生型小鼠经TBHQ(一种Nrf2激活剂)治疗后的结果得到进一步验证:结果:EA预处理改善了CIR损伤引起的认知障碍和神经元损伤。从机理上讲,EA抑制了大脑皮层的氧化应激,表现为活性氧和丙二醛水平的降低以及超氧化物歧化酶活性的增加。此外,EA 还能上调 Nrf2 及其下游抗氧化酶 HO-1 和 NQO1 的表达,而 Keap1 的表达则不受影响。在体外,GSK-3β的过表达抑制了EA血清对OGD/R诱导的神经元损伤的保护作用。在体内,Nrf2或Gsk-3β基因的敲除会取消EA的神经保护作用,而TBHQ的作用与EA相似,这证实了GSK-3β/Nrf2在介导EA抗氧化作用中的重要作用:结论:EA通过激活GSK-3β/Nrf2信号通路发挥抗CIR损伤的抗氧化作用,与Keap1调控无关。
{"title":"Keap1-independent GSK-3β/Nrf2 signaling mediates electroacupuncture inhibition of oxidative stress to induce cerebral ischemia-reperfusion tolerance","authors":"","doi":"10.1016/j.brainresbull.2024.111071","DOIUrl":"10.1016/j.brainresbull.2024.111071","url":null,"abstract":"<div><h3>Purpose</h3><p>Cerebral ischemia-reperfusion (CIR) injury is a devastating consequence of stroke characterized by oxidative stress-induced neuronal damage. Electroacupuncture (EA) has emerged as a potential therapeutic intervention for ischemic stroke, but its underlying mechanisms remain incompletely understood. This study aimed to elucidate whether EA exerts anti-oxidative stress effects against CIR injury by modulating the GSK-3β/Nrf2 pathway.</p></div><div><h3>Methods</h3><p>CIR mouse models were established using the suture-occluded method and underwent EA pretreatment. Cognitive and neurologic function, cerebral infarct volume, and neuronal damage were assessed in mice. Oxidative stress levels and the expression of components of the GSK-3β/Nrf2 pathway in the cerebral cortex were measured. The regulatory effect of GSK-3β on Nrf2 and its role in electroacupuncture to alleviate oxygen-glucose deprivation/reoxygenation (OGD/R)-induced neuronal injury were investigated by modulating GSK-3β expression in HT22 hippocampal neuronal cells and electroacupuncture serum intervention. Ultimately, Nrf2 knockout mice, GSK-3β knockout mice, and wild-type mice treated with TBHQ (an Nrf2 activator) were utilized for further validation.</p></div><div><h3>Results</h3><p>EA pretreatment improved cognitive impairment and neuronal damage induced by CIR injury. Mechanistically, EA inhibited oxidative stress in the cerebral cortex, manifested by reduced levels of reactive oxygen species and malondialdehyde, along with increased superoxide dismutase activity. Furthermore, EA upregulated the expression of Nrf2 and its downstream antioxidant enzymes HO-1 and NQO1, while Keap1 expression remained unaffected. <em>In vitro</em>, GSK-3β overexpression inhibited the protective effects of EA serum on OGD/R-induced neuronal damage. <em>In vivo</em>, knockout of either Nrf2 or Gsk-3β genes abolished the neuroprotective effects of EA, and TBHQ exerted effects similar to EA, confirming the significant role of GSK-3β/Nrf2 in mediating EA antioxidative effects.</p></div><div><h3>Conclusion</h3><p>EA exerts antioxidative stress effects against CIR injury by activating the GSK-3β/Nrf2 signaling pathway, independent of Keap1 regulation.</p></div>","PeriodicalId":9302,"journal":{"name":"Brain Research Bulletin","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0361923024002053/pdfft?md5=55c1884e133ec2fa4026461e364d4b57&pid=1-s2.0-S0361923024002053-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142145253","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Delayed simvastatin treatment improves neurological recovery after cryogenic traumatic brain injury through downregulation of ELOVL1 by inhibiting mTOR signaling 延迟辛伐他汀治疗可通过抑制 mTOR 信号转导下调 ELOVL1,改善低温脑外伤后的神经功能恢复。
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-09-05 DOI: 10.1016/j.brainresbull.2024.111072

Statins are well-tolerated and widely available lipid-lowering medications with neuroprotective effects against traumatic brain injury (TBI). However, whether delayed statin therapy starting in the subacute phase promotes recovery after TBI is unknown. Elongation of the very long-chain fatty acid protein 1 (ELOVL1) is involved in astrocyte-mediated neurotoxicity, but its role in TBI and the relationship between ELOVL1 and statins are unclear. We hypothesized that delayed simvastatin treatment promotes neurological functional recovery after TBI by regulating the ELOVL1-mediated production of very long-chain fatty acids (VLCFAs). ICR male mice received daily intragastric administration of 1, 2 or 5 mg/kg simvastatin on Days 1–14, 3–14, 5–14, or 7–14 after cryogenic TBI (cTBI). The results showed that simvastatin promoted motor functional recovery in a dose-dependent manner, with a wide therapeutic window of at least 7 days postinjury. Meanwhile, simvastatin inhibited astrocyte and microglial overactivation and glial scar formation, and increased total dendritic length, neuronal complexity and spine density on day 14 after cTBI. The up-regulation of ELOVL1 expression and saturated VLCFAs concentrations in the cortex surrounding the lesion caused by cTBI was inhibited by simvastatin, which was related to the inhibition of the mTOR signaling. Overexpression of ELOVL1 in astrocytes surrounding the lesion using HBAAV2/9-GFAP-m-ELOVL1–3xFlag-EGFP partially attenuated the benefits of simvastatin. These results showed that delayed simvastatin treatment promoted functional recovery and brain tissue repair after TBI through the downregulation of ELOVL1 expression by inhibiting mTOR signaling. Astrocytic ELOVL1 may be a potential target for rehabilitation after TBI.

他汀类药物是一种耐受性良好且可广泛使用的降脂药物,对创伤性脑损伤(TBI)具有神经保护作用。然而,从亚急性阶段开始的他汀类药物延迟治疗是否能促进创伤性脑损伤后的恢复尚不清楚。超长链脂肪酸蛋白 1(ELOVL1)参与了星形胶质细胞介导的神经毒性,但它在创伤性脑损伤中的作用以及 ELOVL1 与他汀类药物之间的关系尚不清楚。我们假设,通过调节 ELOVL1 介导的超长链脂肪酸(VLCFAs)的产生,延迟辛伐他汀治疗可促进创伤性脑损伤后的神经功能恢复。在低温创伤性脑损伤(cTBI)后的第1-14天、第3-14天、第5-14天或第7-14天,每天给ICR雄性小鼠胃内注射1、2或5mg/kg辛伐他汀。结果表明,辛伐他汀以剂量依赖的方式促进了运动功能的恢复,其治疗窗口期较宽,至少为伤后7天。同时,辛伐他汀能抑制星形胶质细胞和小胶质细胞的过度激活和胶质瘢痕的形成,并能增加创伤性脑损伤后第14天的树突总长度、神经元复杂性和脊柱密度。辛伐他汀抑制了创伤性脑损伤引起的病变周围皮层中 ELOVL1 表达的上调和饱和 VLCFAs 的浓度,这与抑制 mTOR 信号转导有关。使用 HBAAV2/9-GFAP-m-ELOVL1-3xFlag-EGFP 在病变周围的星形胶质细胞中过表达 ELOVL1 部分削弱了辛伐他汀的作用。这些结果表明,延迟辛伐他汀治疗可通过抑制 mTOR 信号转导下调 ELOVL1 的表达,促进创伤性脑损伤后的功能恢复和脑组织修复。星形胶质细胞ELOVL1可能是创伤性脑损伤后康复的潜在靶点。
{"title":"Delayed simvastatin treatment improves neurological recovery after cryogenic traumatic brain injury through downregulation of ELOVL1 by inhibiting mTOR signaling","authors":"","doi":"10.1016/j.brainresbull.2024.111072","DOIUrl":"10.1016/j.brainresbull.2024.111072","url":null,"abstract":"<div><p>Statins are well-tolerated and widely available lipid-lowering medications with neuroprotective effects against traumatic brain injury (TBI). However, whether delayed statin therapy starting in the subacute phase promotes recovery after TBI is unknown. Elongation of the very long-chain fatty acid protein 1 (ELOVL1) is involved in astrocyte-mediated neurotoxicity, but its role in TBI and the relationship between ELOVL1 and statins are unclear. We hypothesized that delayed simvastatin treatment promotes neurological functional recovery after TBI by regulating the ELOVL1-mediated production of very long-chain fatty acids (VLCFAs). ICR male mice received daily intragastric administration of 1, 2 or 5 mg/kg simvastatin on Days 1–14, 3–14, 5–14, or 7–14 after cryogenic TBI (cTBI). The results showed that simvastatin promoted motor functional recovery in a dose-dependent manner, with a wide therapeutic window of at least 7 days postinjury. Meanwhile, simvastatin inhibited astrocyte and microglial overactivation and glial scar formation, and increased total dendritic length, neuronal complexity and spine density on day 14 after cTBI. The up-regulation of ELOVL1 expression and saturated VLCFAs concentrations in the cortex surrounding the lesion caused by cTBI was inhibited by simvastatin, which was related to the inhibition of the mTOR signaling. Overexpression of ELOVL1 in astrocytes surrounding the lesion using HBAAV2/9-GFAP-m-ELOVL1–3xFlag-EGFP partially attenuated the benefits of simvastatin. These results showed that delayed simvastatin treatment promoted functional recovery and brain tissue repair after TBI through the downregulation of ELOVL1 expression by inhibiting mTOR signaling. Astrocytic ELOVL1 may be a potential target for rehabilitation after TBI.</p></div>","PeriodicalId":9302,"journal":{"name":"Brain Research Bulletin","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0361923024002065/pdfft?md5=3bc2c2bc98e021affcaf45aa8ce0fc1f&pid=1-s2.0-S0361923024002065-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142145252","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Brain Research Bulletin
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