首页 > 最新文献

Molecular and Cellular Neuroscience最新文献

英文 中文
Potential key pathophysiological participant and treatment target in autism spectrum disorder: Microglia 自闭症谱系障碍的潜在关键病理生理参与者和治疗目标:小胶质细胞
IF 2.6 3区 医学 Q3 NEUROSCIENCES Pub Date : 2024-11-22 DOI: 10.1016/j.mcn.2024.103980
Zehua Tan , Ruixin Xia , Xin Zhao , Zile Yang, Haiying Liu, Wenting Wang
Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders characterized by social and communication deficits, as well as restricted or repetitive behaviors or interests. Although the etiology of ASD remains unclear, there is abundant evidence suggesting that microglial dysfunction is likely to be a significant factor in the pathophysiology of ASD. Microglia, the primary innate immune cells in the central nervous system (CNS), play a crucial role in brain development and homeostasis. Recently, numerous studies have shown that microglia in ASD models display various abnormalities including morphology, function, cellular interactions, genetic and epigenetic factors, as well as the expression of receptors, transcription factors, and cytokines. They impact normal neural development through various mechanisms contributing to ASD, such as neuroinflammation, and alterations in synaptic formation and pruning. The focus of this review is on recent studies regarding microglial abnormalities in ASD and their effects on the onset and progression of ASD at both cellular and molecular levels. It can provide insight into the specific contribution of microglia to ASD pathogenesis and help in designing potential therapeutic and preventative strategies targeting microglia.
自闭症谱系障碍(ASD)是一组神经发育障碍,其特点是社交和沟通障碍,以及行为或兴趣受限或重复。虽然自闭症谱系障碍的病因尚不清楚,但有大量证据表明,小胶质细胞功能障碍可能是自闭症谱系障碍病理生理学中的一个重要因素。小胶质细胞是中枢神经系统(CNS)的主要先天性免疫细胞,在大脑发育和平衡中发挥着至关重要的作用。最近,大量研究表明,ASD 模型中的小胶质细胞表现出各种异常,包括形态、功能、细胞相互作用、遗传和表观遗传因素,以及受体、转录因子和细胞因子的表达。它们通过导致 ASD 的各种机制影响正常的神经发育,如神经炎症、突触形成和修剪的改变。本综述的重点是有关 ASD 中小胶质细胞异常及其在细胞和分子水平上对 ASD 发病和进展的影响的最新研究。它有助于深入了解小胶质细胞对 ASD 发病机制的具体作用,并有助于设计针对小胶质细胞的潜在治疗和预防策略。
{"title":"Potential key pathophysiological participant and treatment target in autism spectrum disorder: Microglia","authors":"Zehua Tan ,&nbsp;Ruixin Xia ,&nbsp;Xin Zhao ,&nbsp;Zile Yang,&nbsp;Haiying Liu,&nbsp;Wenting Wang","doi":"10.1016/j.mcn.2024.103980","DOIUrl":"10.1016/j.mcn.2024.103980","url":null,"abstract":"<div><div>Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders characterized by social and communication deficits, as well as restricted or repetitive behaviors or interests. Although the etiology of ASD remains unclear, there is abundant evidence suggesting that microglial dysfunction is likely to be a significant factor in the pathophysiology of ASD. Microglia, the primary innate immune cells in the central nervous system (CNS), play a crucial role in brain development and homeostasis. Recently, numerous studies have shown that microglia in ASD models display various abnormalities including morphology, function, cellular interactions, genetic and epigenetic factors, as well as the expression of receptors, transcription factors, and cytokines. They impact normal neural development through various mechanisms contributing to ASD, such as neuroinflammation, and alterations in synaptic formation and pruning. The focus of this review is on recent studies regarding microglial abnormalities in ASD and their effects on the onset and progression of ASD at both cellular and molecular levels. It can provide insight into the specific contribution of microglia to ASD pathogenesis and help in designing potential therapeutic and preventative strategies targeting microglia.</div></div>","PeriodicalId":18739,"journal":{"name":"Molecular and Cellular Neuroscience","volume":"131 ","pages":"Article 103980"},"PeriodicalIF":2.6,"publicationDate":"2024-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142695557","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
Sphingosine-1-phosphate receptor 3 promotes neuronal apoptosis via the TNF-α/caspase-3 signaling pathway after acute intracerebral hemorrhage 急性脑内出血后,鞘磷脂-1-磷酸受体 3 通过 TNF-α/caspase-3 信号通路促进神经细胞凋亡
IF 2.6 3区 医学 Q3 NEUROSCIENCES Pub Date : 2024-11-22 DOI: 10.1016/j.mcn.2024.103979
Dengpan Song , Mengyuan Li , Longxiao Zhang , Kaiyuan Zhang , Yuan An , Mengzhao Feng , Fang Wang , Chi-Tai Yeh , Jian Wang , Fuyou Guo

Background

Intracerebral hemorrhage (ICH) has a high incidence and mortality rate among cerebrovascular diseases, and effective treatments are lacking. Sphingosine-1-phosphate receptor 3 (S1PR3) is associated with secondary immune inflammatory injury following ICH. However, its relationship with neuronal apoptosis and the specific underlying mechanism are not clear.

Methods

We observed the effect of S1PR3 on neuronal apoptosis by assessing neurobehavioral scores, performing Western blot (WB) analysis, and performing TUNEL staining in a mouse model of ICH. Moreover, WBs and flow cytometry were used to study the specific mechanism and signaling pathways in HT22 cells in vitro.

Results

The expression of S1PR3, CCL2, TNF-α, and cleaved-caspase-3 (c-caspase-3) and neuronal apoptosis were significantly increased after ICH, accompanied by neurobehavioral deterioration. These effects were significantly improved by treatment with CAY10444, a specific S1PR3 antagonist. After S1P stimulation of HT22 cells, the expression of S1PR3, CCL2, TNF-α and c-caspase-3 increased, and neuronal apoptosis increased by activating caspase-3 through the downstream PI3K/AKT apoptosis signaling pathway. After CAY10444 treatment, the expression of CCL2, TNF-α and c-caspase-3 was significantly reduced, and the PI3K/AKT apoptotic signaling pathway was regulated to reduce neuronal apoptosis.

Conclusion

An increase in S1P/S1PR3 after ICH may induce neuronal apoptosis by increasing TNF-α expression and activating the PI3K/AKT signaling pathway and the expression of caspase-3 effector proteins. CAY10444 can reduce neuronal apoptosis, improve symptoms and play a neuroprotective role by antagonizing S1PR3. S1PR3 may be a promising therapeutic target.
背景在脑血管疾病中,脑出血(ICH)的发病率和死亡率都很高,而且缺乏有效的治疗方法。两性鞘氨醇-1-磷酸受体 3(S1PR3)与 ICH 后的继发性免疫炎症损伤有关。我们在 ICH 小鼠模型中通过神经行为评分、Western 印迹(WB)分析和 TUNEL 染色观察了 S1PR3 对神经细胞凋亡的影响。结果 ICH后S1PR3、CCL2、TNF-α和裂解-caspase-3(c-caspase-3)的表达和神经元凋亡显著增加,并伴随神经行为的恶化。使用特异性 S1PR3 拮抗剂 CAY10444 治疗可明显改善这些影响。S1P刺激HT22细胞后,S1PR3、CCL2、TNF-α和c-caspase-3的表达增加,通过下游的PI3K/AKT凋亡信号通路激活caspase-3,从而增加神经元凋亡。CAY10444治疗后,CCL2、TNF-α和c-caspase-3的表达明显减少,PI3K/AKT凋亡信号通路被调控,从而减少神经元凋亡。CAY10444 可通过拮抗 S1PR3 减少神经元凋亡,改善症状,发挥神经保护作用。S1PR3 可能是一个很有前景的治疗靶点。
{"title":"Sphingosine-1-phosphate receptor 3 promotes neuronal apoptosis via the TNF-α/caspase-3 signaling pathway after acute intracerebral hemorrhage","authors":"Dengpan Song ,&nbsp;Mengyuan Li ,&nbsp;Longxiao Zhang ,&nbsp;Kaiyuan Zhang ,&nbsp;Yuan An ,&nbsp;Mengzhao Feng ,&nbsp;Fang Wang ,&nbsp;Chi-Tai Yeh ,&nbsp;Jian Wang ,&nbsp;Fuyou Guo","doi":"10.1016/j.mcn.2024.103979","DOIUrl":"10.1016/j.mcn.2024.103979","url":null,"abstract":"<div><h3>Background</h3><div>Intracerebral hemorrhage (ICH) has a high incidence and mortality rate among cerebrovascular diseases, and effective treatments are lacking. Sphingosine-1-phosphate receptor 3 (S1PR3) is associated with secondary immune inflammatory injury following ICH. However, its relationship with neuronal apoptosis and the specific underlying mechanism are not clear.</div></div><div><h3>Methods</h3><div>We observed the effect of S1PR3 on neuronal apoptosis by assessing neurobehavioral scores, performing Western blot (WB) analysis, and performing TUNEL staining in a mouse model of ICH. Moreover, WBs and flow cytometry were used to study the specific mechanism and signaling pathways in HT22 cells in vitro.</div></div><div><h3>Results</h3><div>The expression of S1PR3, CCL2, TNF-α, and cleaved-caspase-3 (c-caspase-3) and neuronal apoptosis were significantly increased after ICH, accompanied by neurobehavioral deterioration. These effects were significantly improved by treatment with CAY10444, a specific S1PR3 antagonist. After S1P stimulation of HT22 cells, the expression of S1PR3, CCL2, TNF-α and c-caspase-3 increased, and neuronal apoptosis increased by activating caspase-3 through the downstream PI3K/AKT apoptosis signaling pathway. After CAY10444 treatment, the expression of CCL2, TNF-α and c-caspase-3 was significantly reduced, and the PI3K/AKT apoptotic signaling pathway was regulated to reduce neuronal apoptosis.</div></div><div><h3>Conclusion</h3><div>An increase in S1P/S1PR3 after ICH may induce neuronal apoptosis by increasing TNF-α expression and activating the PI3K/AKT signaling pathway and the expression of caspase-3 effector proteins. CAY10444 can reduce neuronal apoptosis, improve symptoms and play a neuroprotective role by antagonizing S1PR3. S1PR3 may be a promising therapeutic target.</div></div>","PeriodicalId":18739,"journal":{"name":"Molecular and Cellular Neuroscience","volume":"131 ","pages":"Article 103979"},"PeriodicalIF":2.6,"publicationDate":"2024-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142702763","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
The mRNA expression profile of glycine receptor subunits alpha 1, alpha 2, alpha 4 and beta in female and male mice. 雌性和雄性小鼠甘氨酸受体亚基 alpha 1、alpha 2、alpha 4 和 beta 的 mRNA 表达谱。
IF 2.6 3区 医学 Q3 NEUROSCIENCES Pub Date : 2024-11-21 DOI: 10.1016/j.mcn.2024.103976
Mikaela M Ceder, Kajsa A Magnusson, Hannah M Weman, Katharina Henriksson, Linn Andréasson, Teresa Lindström, Oskar Wiggins, Malin C Lagerström

Glycine receptors are ligand-gated chloride-selective channels that control excitability in the central nervous system (CNS). Herein, we have investigated the mRNA expression of the glycine receptor alpha 1 (Glra1), alpha 2 (Glra2), alpha 4 (Glra4) and beta (Glrb) subunits, in adult female and male mice. Single-cell RNA sequencing data re-analysis of the Zeisel et al. (2018) dataset indicated widespread expression of Glra1, Glra2 and Glrb in the CNS, while only a few cells in the cortex, striatum, thalamus, midbrain and spinal cord expressed Glra4. Highest occurrence of Glra1, Glra2 and Glrb were found in the brainstem. Moreover, Glra1 and Glrb were revealed to have the highest occurrences in the spinal cord of the investigated subunits. However, both Glra2 and Glrb had a more widespread expression in the CNS compared with Glra1 and Glra4. Bulk quantitative real-time-PCR (qRT-PCR) analysis revealed Glra1 expression in the hypothalamus, thalamus, brainstem and the spinal cord, and widespread, but low, Glra2 and Glrb expression in the CNS. Moreover, Glrb could be detected in a few visceral organs. Additionally, females and males were found to express Glra1, Glra2 and Glrb differently in certain brain areas such as the brainstem. Expression levels of Glra4 were too low to be detected using qRT-PCR. Lastly, RNAscope spatially validated the expression of Glra1, Glra2 and Glrb in the areas indicated by the single-cell and bulk analyses, and further revealed that Glra4 can be detected in the cortex, amygdala, hypothalamus, thalamus, brainstem, especially the cochlear nucleus, and in the spinal cord.

甘氨酸受体是配体门控的氯离子选择性通道,可控制中枢神经系统(CNS)的兴奋性。在此,我们研究了成年雌性和雄性小鼠甘氨酸受体α1(Glra1)、α2(Glra2)、α4(Glra4)和β(Glrb)亚基的 mRNA 表达。对Zeisel等人(2018年)数据集进行的单细胞RNA测序数据再分析表明,Glra1、Glra2和Glrb在中枢神经系统中广泛表达,而皮层、纹状体、丘脑、中脑和脊髓中只有少数细胞表达Glra4。Glra1、Glra2和Glrb在脑干的出现率最高。此外,在所研究的亚单位中,Glra1 和 Glrb 在脊髓中的出现率最高。不过,与 Glra1 和 Glra4 相比,Glra2 和 Glrb 在中枢神经系统中的表达更为广泛。大量实时定量 PCR(qRT-PCR)分析显示,Glra1 在下丘脑、丘脑、脑干和脊髓中表达,Glra2 和 Glrb 在中枢神经系统中广泛表达,但表达量较低。此外,在一些内脏器官中也能检测到 Glrb。此外,还发现雌性和雄性在某些脑区(如脑干)的Glra1、Glra2和Glrb表达量不同。Glra4 的表达水平太低,无法通过 qRT-PCR 检测到。最后,RNAscope 从空间上验证了 Glra1、Glra2 和 Glrb 在单细胞和大块分析所显示区域的表达,并进一步发现在大脑皮层、杏仁核、下丘脑、丘脑、脑干(尤其是耳蜗核)和脊髓中可以检测到 Glra4。
{"title":"The mRNA expression profile of glycine receptor subunits alpha 1, alpha 2, alpha 4 and beta in female and male mice.","authors":"Mikaela M Ceder, Kajsa A Magnusson, Hannah M Weman, Katharina Henriksson, Linn Andréasson, Teresa Lindström, Oskar Wiggins, Malin C Lagerström","doi":"10.1016/j.mcn.2024.103976","DOIUrl":"https://doi.org/10.1016/j.mcn.2024.103976","url":null,"abstract":"<p><p>Glycine receptors are ligand-gated chloride-selective channels that control excitability in the central nervous system (CNS). Herein, we have investigated the mRNA expression of the glycine receptor alpha 1 (Glra1), alpha 2 (Glra2), alpha 4 (Glra4) and beta (Glrb) subunits, in adult female and male mice. Single-cell RNA sequencing data re-analysis of the Zeisel et al. (2018) dataset indicated widespread expression of Glra1, Glra2 and Glrb in the CNS, while only a few cells in the cortex, striatum, thalamus, midbrain and spinal cord expressed Glra4. Highest occurrence of Glra1, Glra2 and Glrb were found in the brainstem. Moreover, Glra1 and Glrb were revealed to have the highest occurrences in the spinal cord of the investigated subunits. However, both Glra2 and Glrb had a more widespread expression in the CNS compared with Glra1 and Glra4. Bulk quantitative real-time-PCR (qRT-PCR) analysis revealed Glra1 expression in the hypothalamus, thalamus, brainstem and the spinal cord, and widespread, but low, Glra2 and Glrb expression in the CNS. Moreover, Glrb could be detected in a few visceral organs. Additionally, females and males were found to express Glra1, Glra2 and Glrb differently in certain brain areas such as the brainstem. Expression levels of Glra4 were too low to be detected using qRT-PCR. Lastly, RNAscope spatially validated the expression of Glra1, Glra2 and Glrb in the areas indicated by the single-cell and bulk analyses, and further revealed that Glra4 can be detected in the cortex, amygdala, hypothalamus, thalamus, brainstem, especially the cochlear nucleus, and in the spinal cord.</p>","PeriodicalId":18739,"journal":{"name":"Molecular and Cellular Neuroscience","volume":" ","pages":"103976"},"PeriodicalIF":2.6,"publicationDate":"2024-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142695560","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
TAT-PPA1 protects against oxidative stress-induced loss of dopaminergic neurons TAT-PPA1 可防止氧化应激诱导的多巴胺能神经元丧失。
IF 2.6 3区 医学 Q3 NEUROSCIENCES Pub Date : 2024-10-31 DOI: 10.1016/j.mcn.2024.103978
Hyun Jung Kwon , Hyo Young Jung , Soo Young Choi , In Koo Hwang , Dae Won Kim , Min Jea Shin
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra (SN) of the midbrain, resulting in severe motor impairments. Inorganic pyrophosphatase 1 (PPA1) plays a key role in various biological processes, and this study introduces a cell-penetrating PPA1 fusion protein (TAT-PPA1) to explore its transduction into cells and brain tissues. TAT-PPA1 effectively penetrates SH-SY5Y cells and the SN region of PD animal models without toxicity, exhibiting protective effects against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP-)-induced cell death. TAT-PPA1 revealed an inhibitory influence on the MAPK signaling pathway and MPTP-induced reactive oxygen species (ROS) production. TAT-PPA1 suppresses JNK, AKT, p53, ERK, and p38 phosphorylation, showcasing its multifaceted role in cell survival pathways. In the MPTP-induced PD animal model, TAT-PPA1 prevents dopaminergic cell death and enhances motor function. This study shows that TAT-PPA1 protects against oxidative stress and cell death in neurodegenerative diseases, suggesting potential as a PD treatment.
帕金森病(Parkinson's disease,PD)是一种神经退行性疾病,以中脑黑质(substantia nigra,SN)多巴胺能神经元的进行性丧失为特征,导致严重的运动障碍。无机焦磷酸酶1(PPA1)在多种生物过程中发挥着关键作用,本研究引入了一种细胞穿透性PPA1融合蛋白(TAT-PPA1),以探索其在细胞和脑组织中的转导。TAT-PPA1能有效穿透SH-SY5Y细胞和PD动物模型的SN区域,且无毒性,对1-甲基-4-苯基-1,2,3,6-四氢吡啶(MPTP)诱导的细胞死亡有保护作用。TAT-PPA1 对 MAPK 信号通路和 MPTP 诱导的活性氧(ROS)产生有抑制作用。TAT-PPA1 可抑制 JNK、AKT、p53、ERK 和 p38 的磷酸化,显示了它在细胞存活通路中的多方面作用。在 MPTP 诱导的帕金森病动物模型中,TAT-PPA1 可防止多巴胺能细胞死亡并增强运动功能。这项研究表明,TAT-PPA1 可防止神经退行性疾病中的氧化应激和细胞死亡,具有治疗帕金森病的潜力。
{"title":"TAT-PPA1 protects against oxidative stress-induced loss of dopaminergic neurons","authors":"Hyun Jung Kwon ,&nbsp;Hyo Young Jung ,&nbsp;Soo Young Choi ,&nbsp;In Koo Hwang ,&nbsp;Dae Won Kim ,&nbsp;Min Jea Shin","doi":"10.1016/j.mcn.2024.103978","DOIUrl":"10.1016/j.mcn.2024.103978","url":null,"abstract":"<div><div>Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra (SN) of the midbrain, resulting in severe motor impairments. Inorganic pyrophosphatase 1 (PPA1) plays a key role in various biological processes, and this study introduces a cell-penetrating PPA1 fusion protein (TAT-PPA1) to explore its transduction into cells and brain tissues. TAT-PPA1 effectively penetrates SH-SY5Y cells and the SN region of PD animal models without toxicity, exhibiting protective effects against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP-)-induced cell death. TAT-PPA1 revealed an inhibitory influence on the MAPK signaling pathway and MPTP-induced reactive oxygen species (ROS) production. TAT-PPA1 suppresses JNK, AKT, p53, ERK, and p38 phosphorylation, showcasing its multifaceted role in cell survival pathways. In the MPTP-induced PD animal model, TAT-PPA1 prevents dopaminergic cell death and enhances motor function. This study shows that TAT-PPA1 protects against oxidative stress and cell death in neurodegenerative diseases, suggesting potential as a PD treatment.</div></div>","PeriodicalId":18739,"journal":{"name":"Molecular and Cellular Neuroscience","volume":"131 ","pages":"Article 103978"},"PeriodicalIF":2.6,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142564710","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
Inhibition of phosphodiesterase 10A mitigates neuronal injury by modulating apoptotic pathways in cold-induced traumatic brain injury 抑制磷酸二酯酶10A可通过调节冷诱导的创伤性脑损伤中的细胞凋亡途径减轻神经元损伤。
IF 2.6 3区 医学 Q3 NEUROSCIENCES Pub Date : 2024-10-20 DOI: 10.1016/j.mcn.2024.103977
Mustafa C. Beker , Mehmet O. Altintas , Enes Dogan , Cigdem Bayraktaroglu , Buse Balaban , Aysenur Ozpinar , Nursena Sengun , Serdar Altunay , Ertugrul Kilic
Brain injury develops from a complex series of pathophysiological phases, resulting in acute necrotic or delayed apoptotic cell death after traumatic brain injury (TBI). Inhibition of apoptotic cell death is critical for the treatment of acute neurodegenerative disorders, such as TBI. Here, we investigated the role of phosphodiesterase 10A (PDE10A) in the development of neuronal injury, particularly in apoptotic cell death. Using the PDE10A inhibitor TAK-063, we found that PDE10A inhibition is associated with decreased brain injury, brain swelling, and blood brain barrier disruption 48 h after cold-induced TBI. Furthermore, a particularly notable result was observed with 3 mg/kg TAK-063, which reduced disseminated neuronal injury. Protein abundance analysis revealed that PDE10A inhibition activates survival kinases AKT and ERK-1/-2, which were associated with the decreased activation of MMP-9 and PTEN. Additionally, iNOS and nNOS levels significantly reduced in the TAK-063 group, playing roles in inflammation and apoptosis. A planar surface immunoassay was performed for in-depth analyses of the apoptotic signaling pathways. We observed that inhibition of PDE10A resulted in the decreased expression of TNFRSF1A, TNFRSF10B, and TNFRSF6 receptors, particularly inducing apoptotic cell death. Moreover, these findings correlated with reduced levels of pro-apoptotic proteins, including PTEN, p27, Cytochrome-c, cleaved Caspase-3, Bad, and p53. Interestingly, TAK-063 treatment reduced levels of anti-apoptotic proteins or enzymes, including XIAP, Claspin, and HIF1α, without affecting Bcl-x, MCL-1, SMAC, HO-1, HO-2, HSP27, HSP60, and HSP70. The findings suggest that PDE10A regulates cellular signaling predominantly pro-apoptotic pathways, and inhibition of this protein is a promising approach for the treatment of acute brain injury.
脑损伤是由一系列复杂的病理生理阶段发展而来的,导致创伤性脑损伤(TBI)后细胞急性坏死或延迟凋亡。抑制细胞凋亡对于治疗创伤性脑损伤等急性神经退行性疾病至关重要。在这里,我们研究了磷酸二酯酶 10A(PDE10A)在神经元损伤的发展过程中,尤其是在细胞凋亡中的作用。通过使用 PDE10A 抑制剂 TAK-063,我们发现 PDE10A 抑制与冷诱导创伤性脑损伤 48 小时后脑损伤、脑肿胀和血脑屏障破坏的减少有关。此外,3 毫克/千克的 TAK-063 还能减少弥散性神经元损伤,效果尤为显著。蛋白质丰度分析表明,PDE10A 抑制可激活存活激酶 AKT 和 ERK-1/-2,这与 MMP-9 和 PTEN 的激活减少有关。此外,TAK-063 组的 iNOS 和 nNOS 水平明显降低,它们在炎症和细胞凋亡中发挥作用。为了深入分析细胞凋亡信号通路,我们进行了平面表面免疫测定。我们观察到,抑制 PDE10A 会导致 TNFRSF1A、TNFRSF10B 和 TNFRSF6 受体的表达减少,尤其会诱导细胞凋亡。此外,这些发现与促凋亡蛋白水平的降低有关,包括 PTEN、p27、细胞色素-c、裂解的 Caspase-3、Bad 和 p53。有趣的是,TAK-063 处理降低了抗凋亡蛋白或酶的水平,包括 XIAP、Claspin 和 HIF1α,但不影响 Bcl-x、MCL-1、SMAC、HO-1、HO-2、HSP27、HSP60 和 HSP70。研究结果表明,PDE10A主要调节细胞信号传导的促凋亡通路,抑制该蛋白是治疗急性脑损伤的一种很有前景的方法。
{"title":"Inhibition of phosphodiesterase 10A mitigates neuronal injury by modulating apoptotic pathways in cold-induced traumatic brain injury","authors":"Mustafa C. Beker ,&nbsp;Mehmet O. Altintas ,&nbsp;Enes Dogan ,&nbsp;Cigdem Bayraktaroglu ,&nbsp;Buse Balaban ,&nbsp;Aysenur Ozpinar ,&nbsp;Nursena Sengun ,&nbsp;Serdar Altunay ,&nbsp;Ertugrul Kilic","doi":"10.1016/j.mcn.2024.103977","DOIUrl":"10.1016/j.mcn.2024.103977","url":null,"abstract":"<div><div>Brain injury develops from a complex series of pathophysiological phases, resulting in acute necrotic or delayed apoptotic cell death after traumatic brain injury (TBI). Inhibition of apoptotic cell death is critical for the treatment of acute neurodegenerative disorders, such as TBI. Here, we investigated the role of phosphodiesterase 10A (PDE10A) in the development of neuronal injury, particularly in apoptotic cell death. Using the PDE10A inhibitor TAK-063, we found that PDE10A inhibition is associated with decreased brain injury, brain swelling, and blood brain barrier disruption 48 h after cold-induced TBI. Furthermore, a particularly notable result was observed with 3 mg/kg TAK-063, which reduced disseminated neuronal injury. Protein abundance analysis revealed that PDE10A inhibition activates survival kinases AKT and ERK-1/-2, which were associated with the decreased activation of MMP-9 and PTEN. Additionally, iNOS and nNOS levels significantly reduced in the TAK-063 group, playing roles in inflammation and apoptosis. A planar surface immunoassay was performed for in-depth analyses of the apoptotic signaling pathways. We observed that inhibition of PDE10A resulted in the decreased expression of TNFRSF1A, TNFRSF10B, and TNFRSF6 receptors, particularly inducing apoptotic cell death. Moreover, these findings correlated with reduced levels of pro-apoptotic proteins, including PTEN, p27, Cytochrome-c, cleaved Caspase-3, Bad, and p53. Interestingly, TAK-063 treatment reduced levels of anti-apoptotic proteins or enzymes, including XIAP, Claspin, and HIF1α, without affecting Bcl-x, MCL-1, SMAC, HO-1, HO-2, HSP27, HSP60, and HSP70. The findings suggest that PDE10A regulates cellular signaling predominantly pro-apoptotic pathways, and inhibition of this protein is a promising approach for the treatment of acute brain injury.</div></div>","PeriodicalId":18739,"journal":{"name":"Molecular and Cellular Neuroscience","volume":"131 ","pages":"Article 103977"},"PeriodicalIF":2.6,"publicationDate":"2024-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504325","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
Vulnerability of neurofilament-expressing neurons in frontotemporal dementia 额颞叶痴呆症中神经丝表达神经元的脆弱性。
IF 2.6 3区 医学 Q3 NEUROSCIENCES Pub Date : 2024-10-05 DOI: 10.1016/j.mcn.2024.103974
Nina Daniels, Aidan D. Bindoff, James C. Vickers, Anna E. King , Jessica M. Collins
Frontotemporal dementia (FTD) is an umbrella term for several early onset dementias, that are caused by frontotemporal lobar degeneration (FTLD), which involves the atrophy of the frontal and temporal lobes of the brain. Neuron loss in the frontal and temporal lobes is a characteristic feature of FTLD, however the selective vulnerability of different neuronal populations in this group of diseases is not fully understood. Neurofilament-expressing neurons have been shown to be selectively vulnerable in other neurodegenerative diseases, including Alzheimer's disease and amyotrophic lateral sclerosis, therefore we sought to investigate whether this neuronal population is vulnerable in FTLD. We also examined whether neuronal sub-type vulnerability differed between FTLD with TDP-43 inclusions (FTLD-TDP) and FTLD with tau inclusions (FTLD-Tau). Post-mortem human tissue from the superior frontal gyrus (SFG) of FTLD-TDP (n = 15), FTLD-Tau (n = 8) and aged Control cases (n = 6) was immunolabelled using antibodies against non-phosphorylated neurofilaments (SMI32 antibody), calretinin and NeuN, to explore neuronal cell loss. The presence of non-phosphorylated neurofilament immunolabelling in axons of the SFG white matter was also quantified as a measure of axon pathology, as axonal neurofilaments are normally phosphorylated. We demonstrate the selective loss of neurofilament-expressing neurons in both FTLD-TDP and FTLD-Tau cases compared to aged Controls. We also show that non-phosphorylated neurofilament axonal pathology in the SFG white matter was associated with increasing age, but not FTLD. This data suggests neurofilament-expressing neurons are vulnerable in both FTLD-TDP and FTLD-Tau.
额颞叶痴呆症(FTD)是多种早发性痴呆症的总称,由额颞叶变性(FTLD)引起,涉及大脑额叶和颞叶的萎缩。额叶和颞叶的神经元缺失是额颞叶变性的一个特征,但这组疾病中不同神经元群的选择性易感性尚不完全清楚。在其他神经退行性疾病(包括阿尔茨海默病和肌萎缩侧索硬化症)中,神经丝表达的神经元已被证明具有选择性易损性,因此我们试图研究这一神经元群在 FTLD 中是否易损。我们还研究了带有 TDP-43 包涵体的 FTLD(FTLD-TDP)和带有 tau 包涵体的 FTLD(FTLD-Tau)之间神经元亚型的易损性是否存在差异。使用针对非磷酸化神经丝(SMI32 抗体)、钙视蛋白和 NeuN 的抗体对 FTLD-TDP(n = 15)、FTLD-Tau(n = 8)和老年对照病例(n = 6)的额叶上回(SFG)死后人体组织进行免疫标记,以检测神经细胞丢失情况。由于轴突神经丝通常是磷酸化的,因此还对 SFG 白质轴突中存在的非磷酸化神经丝免疫标记进行了量化,作为衡量轴突病理学的一个指标。我们发现,与老年对照组相比,FTLD-TDP 和 FTLD-Tau 病例中神经丝表达神经元的选择性缺失。我们还发现,SFG 白质中的非磷酸化神经丝轴突病变与年龄增长有关,但与 FTLD 无关。这些数据表明,表达神经丝的神经元在FTLD-TDP和FTLD-Tau中都很脆弱。
{"title":"Vulnerability of neurofilament-expressing neurons in frontotemporal dementia","authors":"Nina Daniels,&nbsp;Aidan D. Bindoff,&nbsp;James C. Vickers,&nbsp;Anna E. King ,&nbsp;Jessica M. Collins","doi":"10.1016/j.mcn.2024.103974","DOIUrl":"10.1016/j.mcn.2024.103974","url":null,"abstract":"<div><div>Frontotemporal dementia (FTD) is an umbrella term for several early onset dementias, that are caused by frontotemporal lobar degeneration (FTLD), which involves the atrophy of the frontal and temporal lobes of the brain. Neuron loss in the frontal and temporal lobes is a characteristic feature of FTLD, however the selective vulnerability of different neuronal populations in this group of diseases is not fully understood. Neurofilament-expressing neurons have been shown to be selectively vulnerable in other neurodegenerative diseases, including Alzheimer's disease and amyotrophic lateral sclerosis, therefore we sought to investigate whether this neuronal population is vulnerable in FTLD. We also examined whether neuronal sub-type vulnerability differed between FTLD with TDP-43 inclusions (FTLD-TDP) and FTLD with tau inclusions (FTLD-Tau). Post-mortem human tissue from the superior frontal gyrus (SFG) of FTLD-TDP (n = 15), FTLD-Tau (n = 8) and aged Control cases (n = 6) was immunolabelled using antibodies against non-phosphorylated neurofilaments (SMI32 antibody), calretinin and NeuN, to explore neuronal cell loss. The presence of non-phosphorylated neurofilament immunolabelling in axons of the SFG white matter was also quantified as a measure of axon pathology, as axonal neurofilaments are normally phosphorylated. We demonstrate the selective loss of neurofilament-expressing neurons in both FTLD-TDP and FTLD-Tau cases compared to aged Controls. We also show that non-phosphorylated neurofilament axonal pathology in the SFG white matter was associated with increasing age, but not FTLD. This data suggests neurofilament-expressing neurons are vulnerable in both FTLD-TDP and FTLD-Tau.</div></div>","PeriodicalId":18739,"journal":{"name":"Molecular and Cellular Neuroscience","volume":"131 ","pages":"Article 103974"},"PeriodicalIF":2.6,"publicationDate":"2024-10-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142381270","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
Astrocytes initiate autophagic flux and maintain cell viability after internalizing non-active native extracellular α-synuclein 星形胶质细胞在内化非活性原生细胞外α-突触核蛋白后启动自噬通量并维持细胞活力。
IF 2.6 3区 医学 Q3 NEUROSCIENCES Pub Date : 2024-10-04 DOI: 10.1016/j.mcn.2024.103975
Fotis Andromidas , Brooke E. Mackinnon , Abigail J. Myers , Melanie M. Shaffer , Ayat Brahimi , Saeid Atashpanjeh , Tiana L. Vazquez , Timmy Le , Evan R. Jellison , Susan Staurovsky , Andrew O. Koob
Astrocytes are tasked with regulating the synaptic environment. Early stages of various neurodegenerative diseases are characterized by synapse loss, and astrocytic atrophy and dysfunction has been proposed as a possible cause. α-Synuclein (αS) is a highly expressed neuronal protein located in the synapse that can be released in the extracellular space. Evidence points to astrocytes as being responsible for uptake and degradation of extracellular αS. Therefore, misfolded active fibrillized αS resulting in protein inclusions and aggregates could be due to astrocytic dysfunction. Despite these pathological hallmarks and lines of evidence, the autophagic function of astrocytes in response to monomeric non-active αS to model healthy conditions has not been investigated. Human primary cortical astrocytes were treated with 100 nM of extracellular monomeric non-active αS alone, and in combination with N-terminal binding monomeric γ-synuclein (γS) as a control. Western blot analysis and super resolution imaging of HiLyte-488 labeled αS confirmed successful internalization of αS at 12, 24 and 48 h after treatment, while αS dimers were only observed at 48 h. Western blot analysis also confirmed αS's ability to induce autophagic flux by 48 h. Annexin V/PI flow cytometry results revealed increased early apoptosis at 24 h, but which resolved itself by 48 h, indicating no cell death in cortical astrocytes at all time points, suggesting astrocytes can manage the protein degradation demand of monomeric αS in healthy physiological conditions. Likewise, astrocytes reduced secretion of apolipoprotein (ApoE), a protein involved in pro-inflammatory pathways, synapse regulation, and autophagy by 12 h. Similarly, total c-JUN protein levels, a transcription factor involved in pro-inflammatory pathways increased by 12 h in the nuclear fraction. Therefore, astrocytes are able to respond and degrade αS in healthy physiological conditions, and astrocyte dysfunction could precede detrimental αS accumulation.
星形胶质细胞的任务是调节突触环境。各种神经退行性疾病的早期阶段都以突触丧失为特征,而星形胶质细胞萎缩和功能障碍被认为是可能的原因之一。α-突触核蛋白(αS)是位于突触中的一种高表达神经元蛋白,可释放到细胞外空间。有证据表明,星形胶质细胞负责吸收和降解细胞外的αS。因此,星形胶质细胞功能障碍可能导致折叠错误的活性纤维化 αS,从而导致蛋白质内含物和聚集。尽管有这些病理特征和证据,但尚未研究星形胶质细胞对单体非活性αS的自噬功能,以模拟健康状况。用 100 nM 细胞外单体非活性 αS 单独处理人类原代皮质星形胶质细胞,并结合 N 端结合单体 γ-突触核蛋白(γS)作为对照。对HiLyte-488标记的αS进行Western印迹分析和超分辨率成像,证实αS在处理后12、24和48小时成功内化,而αS二聚体只在48小时才被观察到。Western 印迹分析也证实了 αS 在 48 小时前诱导自噬通量的能力。Annexin V/PI 流式细胞术结果显示,24 小时后早期细胞凋亡增加,但到 48 小时后凋亡消失,这表明皮质星形胶质细胞在所有时间点都没有细胞死亡,这表明星形胶质细胞在健康的生理条件下可以处理单体 αS 的蛋白质降解需求。同样,12 小时后,星形胶质细胞减少了载脂蛋白(ApoE)的分泌,载脂蛋白是一种参与促炎途径、突触调节和自噬的蛋白质。同样,参与促炎通路的转录因子 c-JUN 蛋白的总含量在 12 小时后也在核部分有所增加。因此,在健康的生理条件下,星形胶质细胞能够对αS做出反应并降解αS,而星形胶质细胞功能障碍可能先于有害的αS积累。
{"title":"Astrocytes initiate autophagic flux and maintain cell viability after internalizing non-active native extracellular α-synuclein","authors":"Fotis Andromidas ,&nbsp;Brooke E. Mackinnon ,&nbsp;Abigail J. Myers ,&nbsp;Melanie M. Shaffer ,&nbsp;Ayat Brahimi ,&nbsp;Saeid Atashpanjeh ,&nbsp;Tiana L. Vazquez ,&nbsp;Timmy Le ,&nbsp;Evan R. Jellison ,&nbsp;Susan Staurovsky ,&nbsp;Andrew O. Koob","doi":"10.1016/j.mcn.2024.103975","DOIUrl":"10.1016/j.mcn.2024.103975","url":null,"abstract":"<div><div>Astrocytes are tasked with regulating the synaptic environment. Early stages of various neurodegenerative diseases are characterized by synapse loss, and astrocytic atrophy and dysfunction has been proposed as a possible cause. α-Synuclein (αS) is a highly expressed neuronal protein located in the synapse that can be released in the extracellular space. Evidence points to astrocytes as being responsible for uptake and degradation of extracellular αS. Therefore, misfolded active fibrillized αS resulting in protein inclusions and aggregates could be due to astrocytic dysfunction. Despite these pathological hallmarks and lines of evidence, the autophagic function of astrocytes in response to monomeric non-active αS to model healthy conditions has not been investigated. Human primary cortical astrocytes were treated with 100 nM of extracellular monomeric non-active αS alone, and in combination with N-terminal binding monomeric γ-synuclein (γS) as a control. Western blot analysis and super resolution imaging of HiLyte-488 labeled αS confirmed successful internalization of αS at 12, 24 and 48 h after treatment, while αS dimers were only observed at 48 h. Western blot analysis also confirmed αS's ability to induce autophagic flux by 48 h. Annexin V/PI flow cytometry results revealed increased early apoptosis at 24 h, but which resolved itself by 48 h, indicating no cell death in cortical astrocytes at all time points, suggesting astrocytes can manage the protein degradation demand of monomeric αS in healthy physiological conditions. Likewise, astrocytes reduced secretion of apolipoprotein (ApoE), a protein involved in pro-inflammatory pathways, synapse regulation, and autophagy by 12 h. Similarly, total c-JUN protein levels, a transcription factor involved in pro-inflammatory pathways increased by 12 h in the nuclear fraction. Therefore, astrocytes are able to respond and degrade αS in healthy physiological conditions, and astrocyte dysfunction could precede detrimental αS accumulation.</div></div>","PeriodicalId":18739,"journal":{"name":"Molecular and Cellular Neuroscience","volume":"131 ","pages":"Article 103975"},"PeriodicalIF":2.6,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142378099","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
The neuroprotective effect of short-chain fatty acids against hypoxia-reperfusion injury 短链脂肪酸对缺氧再灌注损伤的神经保护作用
IF 2.6 3区 医学 Q3 NEUROSCIENCES Pub Date : 2024-09-28 DOI: 10.1016/j.mcn.2024.103972
Anjit K. Harijan , Retnamony Kalaiarasan , Amit Kumar Ghosh , Ruchi P. Jain , Amal Kanti Bera
Gut microbe-derived short-chain fatty acids (SCFAs) are known to have a profound impact on various brain functions, including cognition, mood, and overall neurological health. However, their role, if any, in protecting against hypoxic injury and ischemic stroke has not been extensively studied. In this study, we investigated the effects of two major SCFAs abundant in the gut, propionate (P) and butyrate (B), on hypoxia-reperfusion injury using a neuronal cell line and a zebrafish model. Neuro 2a (N2a) cells treated with P and B exhibited reduced levels of mitochondrial and cytosolic reactive oxygen species (ROS), diminished loss of mitochondrial membrane potential, suppressed caspase activation, and lower rates of cell death when exposed to CoCl2, a chemical commonly used to simulate hypoxia. Furthermore, adult zebrafish fed SCFA-supplemented feeds showed less susceptibility to hypoxic conditions compared to the control group, as indicated by multiple behavioral measures. Histological analysis of 2,3,5-Triphenyltetrazolium chloride (TTC) stained brain sections revealed less damage in the SCFA-fed group. We also found that Fatty Acid Binding Protein 7 (FABP7), also known as Brain Lipid Binding Protein (BLBP), a neuroprotective fatty acid binding protein, was upregulated in the brains of the SCFA-fed group. Additionally, when FABP7 was overexpressed in N2a cells, it protected the cells from injury caused by CoCl2 treatment. Overall, our data provide evidence for a neuroprotective role of P and B against hypoxic brain injury and suggest the potential of dietary supplementation with SCFAs to mitigate stroke-induced brain damage.
众所周知,肠道微生物衍生的短链脂肪酸(SCFAs)对包括认知、情绪和整体神经系统健康在内的各种大脑功能有着深远的影响。然而,它们在保护大脑免受缺氧损伤和缺血性中风方面的作用尚未得到广泛研究。在这项研究中,我们利用神经细胞系和斑马鱼模型研究了肠道中富含的两种主要 SCFAs--丙酸盐(P)和丁酸盐(B)--对缺氧再灌注损伤的影响。当神经 2a(N2a)细胞暴露于常用于模拟缺氧的化学物质 CoCl2 时,经 P 和 B 处理的细胞表现出线粒体和细胞膜活性氧(ROS)水平降低、线粒体膜电位丧失减少、caspase 激活被抑制以及细胞死亡率降低。此外,与对照组相比,喂食添加了 SCFA 的成年斑马鱼在缺氧条件下表现出更低的易感性,这体现在多种行为测量上。2,3,5-三苯基氯化四氮唑(TTC)染色脑切片的组织学分析表明,喂食 SCFA 的斑马鱼损伤较小。我们还发现,脂肪酸结合蛋白 7 (FABP7),又称脑脂结合蛋白 (BLBP),是一种具有神经保护作用的脂肪酸结合蛋白,在 SCFA 饲喂组的大脑中上调。此外,当 FABP7 在 N2a 细胞中过表达时,它能保护细胞免受 CoCl2 处理造成的损伤。总之,我们的数据清楚地证明了 P 和 B 对缺氧性脑损伤的神经保护作用,并表明膳食中补充 SCFAs 有可能减轻中风引起的脑损伤。
{"title":"The neuroprotective effect of short-chain fatty acids against hypoxia-reperfusion injury","authors":"Anjit K. Harijan ,&nbsp;Retnamony Kalaiarasan ,&nbsp;Amit Kumar Ghosh ,&nbsp;Ruchi P. Jain ,&nbsp;Amal Kanti Bera","doi":"10.1016/j.mcn.2024.103972","DOIUrl":"10.1016/j.mcn.2024.103972","url":null,"abstract":"<div><div>Gut microbe-derived short-chain fatty acids (SCFAs) are known to have a profound impact on various brain functions, including cognition, mood, and overall neurological health. However, their role, if any, in protecting against hypoxic injury and ischemic stroke has not been extensively studied. In this study, we investigated the effects of two major SCFAs abundant in the gut, propionate (P) and butyrate (B), on hypoxia-reperfusion injury using a neuronal cell line and a zebrafish model. Neuro 2a (N2a) cells treated with P and B exhibited reduced levels of mitochondrial and cytosolic reactive oxygen species (ROS), diminished loss of mitochondrial membrane potential, suppressed caspase activation, and lower rates of cell death when exposed to CoCl<sub>2</sub>, a chemical commonly used to simulate hypoxia. Furthermore, adult zebrafish fed SCFA-supplemented feeds showed less susceptibility to hypoxic conditions compared to the control group, as indicated by multiple behavioral measures. Histological analysis of 2,3,5-Triphenyltetrazolium chloride (TTC) stained brain sections revealed less damage in the SCFA-fed group. We also found that Fatty Acid Binding Protein 7 (FABP7), also known as Brain Lipid Binding Protein (BLBP), a neuroprotective fatty acid binding protein, was upregulated in the brains of the SCFA-fed group. Additionally, when FABP7 was overexpressed in N2a cells, it protected the cells from injury caused by CoCl<sub>2</sub> treatment. Overall, our data provide evidence for a neuroprotective role of P and B against hypoxic brain injury and suggest the potential of dietary supplementation with SCFAs to mitigate stroke-induced brain damage.</div></div>","PeriodicalId":18739,"journal":{"name":"Molecular and Cellular Neuroscience","volume":"131 ","pages":"Article 103972"},"PeriodicalIF":2.6,"publicationDate":"2024-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142350267","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
Knockdown of BMP7 induced oligodendrocyte apoptosis, demyelination and motor function loss 敲除 BMP7 会诱导少突胶质细胞凋亡、脱髓鞘和运动功能丧失。
IF 2.6 3区 医学 Q3 NEUROSCIENCES Pub Date : 2024-09-25 DOI: 10.1016/j.mcn.2024.103973
Xiaojin Wei , Shuxin Liu , Kai Chen , Meng Wang , Yaping Wang , Dingquan Zou , Yanying Xiao

Background

Demyelinating diseases, including multiple sclerosis (MS) and spinal cord injury (SCI), lead to significant neurological deficits primarily due to the loss of oligodendrocytes (OLs). Bone Morphogenetic Protein 7 (BMP7) is expressed abundantly in the central nervous system and previous studies showed its protective effect in reducing OL loss. In this study, we aim to explore BMP7's potential as a biomarker and therapeutic target for demyelinating diseases by investigating its expression and effects on OLs and myelin sheath integrity.

Method

We analyzed multiple Gene Expression Omnibus datasets for BMP7 expression profiles in demyelinating conditions such as MS and SCI. Experimentally, we employed a BMP7 knockdown model in rat spinal cords using adeno-associated virus8 vectors to specifically reduce BMP7 expression. Western blotting, immunofluorescence, and Nissl staining were used to assess the effect on OL and other types of cells. The structure of myelin sheath and locomotor function were evaluated using transmission electron microscopy and BBB scores, and statistical analysis included ROC curves and ANOVA to evaluate BMP7's diagnostic and therapeutic potential.

Results

BMP7 expression consistently decreased across various demyelinating models, and BMP7 knockdown led to increased OL apoptosis through the Smad1/5/9 pathway, with no apparent effect on other cell types. This reduction in OLs was associated with myelin degeneration, axonal damage, and impaired motor function.

Conclusion

The study confirms BMP7's significant involvement in the pathophysiology of demyelinating diseases and supports its potential as a therapeutic target or biomarker. Future research should focus on therapeutic strategies to enhance BMP7 function and further investigate the mechanisms by which BMP7 supports myelin integrity.
背景:脱髓鞘疾病,包括多发性硬化症(MS)和脊髓损伤(SCI),主要由于少突胶质细胞(OL)的丧失而导致严重的神经功能缺损。骨形态发生蛋白 7(BMP7)在中枢神经系统中大量表达,先前的研究表明它在减少少突胶质细胞丢失方面具有保护作用。在本研究中,我们旨在通过研究 BMP7 的表达及其对 OL 和髓鞘完整性的影响,探索其作为脱髓鞘疾病的生物标记物和治疗靶点的潜力:我们分析了多个基因表达总库数据集,以了解 BMP7 在 MS 和 SCI 等脱髓鞘疾病中的表达情况。实验中,我们使用腺相关病毒8载体在大鼠脊髓中建立了BMP7基因敲除模型,以特异性降低BMP7的表达。我们使用 Western 印迹、免疫荧光和 Nissl 染色来评估其对 OL 和其他类型细胞的影响。统计分析包括ROC曲线和方差分析,以评估BMP7的诊断和治疗潜力:结果:在各种脱髓鞘模型中,BMP7的表达量持续下降,BMP7基因敲除导致OL通过Smad1/5/9途径凋亡增加,但对其他细胞类型没有明显影响。OL的减少与髓鞘变性、轴突损伤和运动功能受损有关:该研究证实了 BMP7 在脱髓鞘疾病的病理生理学中的重要作用,并支持其作为治疗靶点或生物标志物的潜力。未来的研究应侧重于增强 BMP7 功能的治疗策略,并进一步研究 BMP7 支持髓鞘完整性的机制。
{"title":"Knockdown of BMP7 induced oligodendrocyte apoptosis, demyelination and motor function loss","authors":"Xiaojin Wei ,&nbsp;Shuxin Liu ,&nbsp;Kai Chen ,&nbsp;Meng Wang ,&nbsp;Yaping Wang ,&nbsp;Dingquan Zou ,&nbsp;Yanying Xiao","doi":"10.1016/j.mcn.2024.103973","DOIUrl":"10.1016/j.mcn.2024.103973","url":null,"abstract":"<div><h3>Background</h3><div>Demyelinating diseases, including multiple sclerosis (MS) and spinal cord injury (SCI), lead to significant neurological deficits primarily due to the loss of oligodendrocytes (OLs). Bone Morphogenetic Protein 7 (BMP7) is expressed abundantly in the central nervous system and previous studies showed its protective effect in reducing OL loss. In this study, we aim to explore BMP7's potential as a biomarker and therapeutic target for demyelinating diseases by investigating its expression and effects on OLs and myelin sheath integrity.</div></div><div><h3>Method</h3><div>We analyzed multiple Gene Expression Omnibus datasets for BMP7 expression profiles in demyelinating conditions such as MS and SCI. Experimentally, we employed a BMP7 knockdown model in rat spinal cords using adeno-associated virus8 vectors to specifically reduce BMP7 expression. Western blotting, immunofluorescence, and Nissl staining were used to assess the effect on OL and other types of cells. The structure of myelin sheath and locomotor function were evaluated using transmission electron microscopy and BBB scores, and statistical analysis included ROC curves and ANOVA to evaluate BMP7's diagnostic and therapeutic potential.</div></div><div><h3>Results</h3><div>BMP7 expression consistently decreased across various demyelinating models, and BMP7 knockdown led to increased OL apoptosis through the Smad1/5/9 pathway, with no apparent effect on other cell types. This reduction in OLs was associated with myelin degeneration, axonal damage, and impaired motor function.</div></div><div><h3>Conclusion</h3><div>The study confirms BMP7's significant involvement in the pathophysiology of demyelinating diseases and supports its potential as a therapeutic target or biomarker. Future research should focus on therapeutic strategies to enhance BMP7 function and further investigate the mechanisms by which BMP7 supports myelin integrity.</div></div>","PeriodicalId":18739,"journal":{"name":"Molecular and Cellular Neuroscience","volume":"131 ","pages":"Article 103973"},"PeriodicalIF":2.6,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142350268","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
SUMOylation modulates mitochondrial dynamics in an in vitro rotenone model of Parkinson's disease SUMOylation 在帕金森病的离体鱼藤酮模型中调节线粒体动力学
IF 2.6 3区 医学 Q3 NEUROSCIENCES Pub Date : 2024-09-10 DOI: 10.1016/j.mcn.2024.103969
Ericks Sousa Soares , Letícia Yoshitome Queiroz , Ellen Gerhardt , Rui Daniel S. Prediger , Tiago Fleming Outeiro , Helena Iturvides Cimarosti

SUMOylation is a post-translational modification essential for various biological processes. SUMO proteins bind to target substrates in a three-step enzymatic pathway, which is rapidly reversible by the action of specific proteases, known as SENPs. Studies have shown that SUMOylation is dysregulated in several human disorders, including neurodegenerative diseases that are characterized by the progressive loss of neurons, mitochondrial dysfunction, deficits in autophagy, and oxidative stress. Considering the potential neuroprotective roles of SUMOylation, the aim of this study was to investigate the effects of SENP3 knockdown in H4 neuroglioma cells exposed to rotenone, an in vitro model of cytotoxicity that mimics dopaminergic loss in Parkinson's disease (PD). The current data show that SENP3 knockdown increases SUMO-2/3 conjugates, which is accompanied by reduced levels of the mitochondrial fission protein Drp1 and increased levels of the mitochondrial fusion protein OPA1. Of high interest, SENP3 knockdown prevented rotenone-induced superoxide production and cellular death. Taken together, these findings highlight the importance of SUMOylation in maintaining mitochondrial homeostasis and the neuroprotective potential of this modification in PD.

SUMOylation 是一种对各种生物过程至关重要的翻译后修饰。SUMO 蛋白通过三步酶解途径与目标底物结合,在特异性蛋白酶(即 SENPs)的作用下可快速逆转。研究表明,SUMOylation 在多种人类疾病中出现失调,包括神经退行性疾病,这些疾病的特征是神经元的逐渐丧失、线粒体功能障碍、自噬缺陷和氧化应激。考虑到 SUMOylation 的潜在神经保护作用,本研究旨在调查 SENP3 敲除对暴露于鱼藤酮的 H4 神经胶质瘤细胞的影响,鱼藤酮是一种模拟帕金森病(PD)中多巴胺能丧失的体外细胞毒性模型。目前的数据显示,SENP3敲除会增加SUMO-2/3共轭物,同时线粒体裂变蛋白Drp1水平降低,线粒体融合蛋白OPA1水平升高。更令人感兴趣的是,SENP3 基因敲除可防止鱼藤酮诱导的超氧化物生成和细胞死亡。综上所述,这些发现突出了 SUMOylation 在维持线粒体稳态中的重要性,以及这种修饰在帕金森病中的神经保护潜力。
{"title":"SUMOylation modulates mitochondrial dynamics in an in vitro rotenone model of Parkinson's disease","authors":"Ericks Sousa Soares ,&nbsp;Letícia Yoshitome Queiroz ,&nbsp;Ellen Gerhardt ,&nbsp;Rui Daniel S. Prediger ,&nbsp;Tiago Fleming Outeiro ,&nbsp;Helena Iturvides Cimarosti","doi":"10.1016/j.mcn.2024.103969","DOIUrl":"10.1016/j.mcn.2024.103969","url":null,"abstract":"<div><p>SUMOylation is a post-translational modification essential for various biological processes. SUMO proteins bind to target substrates in a three-step enzymatic pathway, which is rapidly reversible by the action of specific proteases, known as SENPs. Studies have shown that SUMOylation is dysregulated in several human disorders, including neurodegenerative diseases that are characterized by the progressive loss of neurons, mitochondrial dysfunction, deficits in autophagy, and oxidative stress. Considering the potential neuroprotective roles of SUMOylation, the aim of this study was to investigate the effects of SENP3 knockdown in H4 neuroglioma cells exposed to rotenone, an <em>in vitro</em> model of cytotoxicity that mimics dopaminergic loss in Parkinson's disease (PD). The current data show that SENP3 knockdown increases SUMO-2/3 conjugates, which is accompanied by reduced levels of the mitochondrial fission protein Drp1 and increased levels of the mitochondrial fusion protein OPA1. Of high interest, SENP3 knockdown prevented rotenone-induced superoxide production and cellular death. Taken together, these findings highlight the importance of SUMOylation in maintaining mitochondrial homeostasis and the neuroprotective potential of this modification in PD.</p></div>","PeriodicalId":18739,"journal":{"name":"Molecular and Cellular Neuroscience","volume":"131 ","pages":"Article 103969"},"PeriodicalIF":2.6,"publicationDate":"2024-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142171877","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
期刊
Molecular and Cellular Neuroscience
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