首页 > 最新文献

Frontiers in Molecular Neuroscience最新文献

英文 中文
The role of Foxo3a in neuron-mediated cognitive impairment. Foxo3a 在神经元介导的认知障碍中的作用
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-19 eCollection Date: 2024-01-01 DOI: 10.3389/fnmol.2024.1424561
Qin-Qin Liu, Gui-Hua Wu, Xiao-Chun Wang, Xiao-Wen Xiong, Rui-Wang, Bao-Le Yao

Cognitive impairment (COI) is a prevalent complication across a spectrum of brain disorders, underpinned by intricate mechanisms yet to be fully elucidated. Neurons, the principal cell population of the nervous system, orchestrate cognitive processes and govern cognitive balance. Extensive inquiry has spotlighted the involvement of Foxo3a in COI. The regulatory cascade of Foxo3a transactivation implicates multiple downstream signaling pathways encompassing mitochondrial function, oxidative stress, autophagy, and apoptosis, collectively affecting neuronal activity. Notably, the expression and activity profile of neuronal Foxo3a are subject to modulation via various modalities, including methylation of promoter, phosphorylation and acetylation of protein. Furthermore, upstream pathways such as PI3K/AKT, the SIRT family, and diverse micro-RNAs intricately interface with Foxo3a, engendering alterations in neuronal function. Through several downstream routes, Foxo3a regulates neuronal dynamics, thereby modulating the onset or amelioration of COI in Alzheimer's disease, stroke, ischemic brain injury, Parkinson's disease, and traumatic brain injury. Foxo3a is a potential therapeutic cognitive target, and clinical drugs or multiple small molecules have been preliminarily shown to have cognitive-enhancing effects that indirectly affect Foxo3a. Particularly noteworthy are multiple randomized, controlled, placebo clinical trials illustrating the significant cognitive enhancement achievable through autophagy modulation. Here, we discussed the role of Foxo3a in neuron-mediated COI and common cognitively impaired diseases.

认知障碍(COI)是一系列脑部疾病中普遍存在的并发症,其复杂的机制尚未完全阐明。神经元是神经系统的主要细胞群,负责协调认知过程并控制认知平衡。广泛的研究表明,Foxo3a 参与了 COI。Foxo3a 转录激活的调控级联涉及多个下游信号通路,包括线粒体功能、氧化应激、自噬和细胞凋亡,共同影响神经元的活动。值得注意的是,神经元 Foxo3a 的表达和活性概况受多种方式的调节,包括启动子甲基化、磷酸化和蛋白质乙酰化。此外,PI3K/AKT、SIRT 家族和各种微 RNA 等上游通路与 Foxo3a 的相互作用错综复杂,导致了神经元功能的改变。Foxo3a 通过几种下游途径调节神经元的动态,从而调节阿尔茨海默病、中风、缺血性脑损伤、帕金森病和创伤性脑损伤中 COI 的发生或改善。Foxo3a 是一个潜在的认知治疗靶点,临床药物或多种小分子药物已初步显示出间接影响 Foxo3a 的认知增强效应。尤其值得注意的是,多项随机对照安慰剂临床试验表明,通过调节自噬可显著增强认知能力。在此,我们讨论了 Foxo3a 在神经元介导的 COI 和常见认知障碍疾病中的作用。
{"title":"The role of Foxo3a in neuron-mediated cognitive impairment.","authors":"Qin-Qin Liu, Gui-Hua Wu, Xiao-Chun Wang, Xiao-Wen Xiong, Rui-Wang, Bao-Le Yao","doi":"10.3389/fnmol.2024.1424561","DOIUrl":"10.3389/fnmol.2024.1424561","url":null,"abstract":"<p><p>Cognitive impairment (COI) is a prevalent complication across a spectrum of brain disorders, underpinned by intricate mechanisms yet to be fully elucidated. Neurons, the principal cell population of the nervous system, orchestrate cognitive processes and govern cognitive balance. Extensive inquiry has spotlighted the involvement of Foxo3a in COI. The regulatory cascade of Foxo3a transactivation implicates multiple downstream signaling pathways encompassing mitochondrial function, oxidative stress, autophagy, and apoptosis, collectively affecting neuronal activity. Notably, the expression and activity profile of neuronal Foxo3a are subject to modulation via various modalities, including methylation of promoter, phosphorylation and acetylation of protein. Furthermore, upstream pathways such as PI3K/AKT, the SIRT family, and diverse micro-RNAs intricately interface with Foxo3a, engendering alterations in neuronal function. Through several downstream routes, Foxo3a regulates neuronal dynamics, thereby modulating the onset or amelioration of COI in Alzheimer's disease, stroke, ischemic brain injury, Parkinson's disease, and traumatic brain injury. Foxo3a is a potential therapeutic cognitive target, and clinical drugs or multiple small molecules have been preliminarily shown to have cognitive-enhancing effects that indirectly affect Foxo3a. Particularly noteworthy are multiple randomized, controlled, placebo clinical trials illustrating the significant cognitive enhancement achievable through autophagy modulation. Here, we discussed the role of Foxo3a in neuron-mediated COI and common cognitively impaired diseases.</p>","PeriodicalId":12630,"journal":{"name":"Frontiers in Molecular Neuroscience","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11220205/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141497816","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
Novel insights into STIM1's role in store-operated calcium entry and its implications for T-cell mediated inflammation in trigeminal neuralgia. STIM1 在钙离子进入贮存器中的作用及其对三叉神经痛中 T 细胞介导的炎症的影响的新见解。
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-19 eCollection Date: 2024-01-01 DOI: 10.3389/fnmol.2024.1391189
Guangyu Cheng, Yu Zhao, Fujia Sun, Qi Zhang

This investigation aims to elucidate the novel role of Stromal Interaction Molecule 1 (STIM1) in modulating store-operated calcium entry (SOCE) and its subsequent impact on inflammatory cytokine release in T lymphocytes, thereby advancing our understanding of trigeminal neuralgia (TN) pathogenesis. Employing the Gene Expression Omnibus (GEO) database, we extracted microarray data pertinent to TN to identify differentially expressed genes (DEGs). A subsequent comparison with SOCE-related genes from the Genecards database helped pinpoint potential target genes. The STRING database facilitated protein-protein interaction (PPI) analysis to spotlight STIM1 as a gene of interest in TN. Through histological staining, transmission electron microscopy (TEM), and behavioral assessments, we probed STIM1's pathological effects on TN in rat models. Additionally, we examined STIM1's influence on the SOCE pathway in trigeminal ganglion cells using techniques like calcium content measurement, patch clamp electrophysiology, and STIM1- ORAI1 co-localization studies. Changes in the expression of inflammatory markers (TNF-α, IL-1β, IL-6) in T cells were quantified using Western blot (WB) and enzyme-linked immunosorbent assay (ELISA) in vitro, while immunohistochemistry and flow cytometry were applied in vivo to assess these cytokines and T cell count alterations. Our bioinformatic approach highlighted STIM1's significant overexpression in TN patients, underscoring its pivotal role in TN's etiology and progression. Experimental findings from both in vitro and in vivo studies corroborated STIM1's regulatory influence on the SOCE pathway. Furthermore, STIM1 was shown to mediate SOCE-induced inflammatory cytokine release in T lymphocytes, a critical factor in TN development. Supportive evidence from histological, ultrastructural, and behavioral analyses reinforced the link between STIM1-mediated SOCE and T lymphocyte-driven inflammation in TN pathogenesis. This study presents novel evidence that STIM1 is a key regulator of SOCE and inflammatory cytokine release in T lymphocytes, contributing significantly to the pathogenesis of trigeminal neuralgia. Our findings not only deepen the understanding of TN's molecular underpinnings but also potentially open new avenues for targeted therapeutic strategies.

本研究旨在阐明基质相互作用分子 1(STIM1)在调节贮存操作钙离子进入(SOCE)及其随后对 T 淋巴细胞释放炎性细胞因子的影响中的新作用,从而加深我们对三叉神经痛(TN)发病机制的理解。我们利用基因表达总库(GEO)数据库提取了与 TN 相关的微阵列数据,以确定差异表达基因(DEGs)。随后与 Genecards 数据库中的 SOCE 相关基因进行比较,帮助确定了潜在的目标基因。STRING 数据库有助于进行蛋白-蛋白相互作用(PPI)分析,从而发现 STIM1 是 TN 中的一个相关基因。通过组织学染色、透射电子显微镜(TEM)和行为评估,我们在大鼠模型中探究了 STIM1 对 TN 的病理影响。此外,我们还利用钙含量测量、膜片钳电生理学和 STIM1- ORAI1 共定位研究等技术,考察了 STIM1 对三叉神经节细胞中 SOCE 通路的影响。在体外,我们使用 Western 印迹(WB)和酶联免疫吸附试验(ELISA)量化了 T 细胞中炎症标志物(TNF-α、IL-1β、IL-6)的表达变化;在体内,我们使用免疫组织化学和流式细胞术评估了这些细胞因子和 T 细胞数量的变化。我们的生物信息学方法强调了STIM1在TN患者中的显著过表达,突出了它在TN的病因和进展中的关键作用。体外和体内研究的实验结果证实了 STIM1 对 SOCE 通路的调节作用。此外,STIM1 还能介导 T 淋巴细胞中由 SOCE 诱导的炎性细胞因子的释放,而这正是 TN 发展过程中的一个关键因素。来自组织学、超微结构和行为分析的支持性证据加强了 STIM1 介导的 SOCE 与 TN 发病过程中 T 淋巴细胞驱动的炎症之间的联系。这项研究提供了新的证据,证明 STIM1 是 T 淋巴细胞 SOCE 和炎症细胞因子释放的关键调节因子,对三叉神经痛的发病机制有重要作用。我们的发现不仅加深了人们对 TN 分子基础的了解,还可能为靶向治疗策略开辟新的途径。
{"title":"Novel insights into STIM1's role in store-operated calcium entry and its implications for T-cell mediated inflammation in trigeminal neuralgia.","authors":"Guangyu Cheng, Yu Zhao, Fujia Sun, Qi Zhang","doi":"10.3389/fnmol.2024.1391189","DOIUrl":"10.3389/fnmol.2024.1391189","url":null,"abstract":"<p><p>This investigation aims to elucidate the novel role of Stromal Interaction Molecule 1 (STIM1) in modulating store-operated calcium entry (SOCE) and its subsequent impact on inflammatory cytokine release in T lymphocytes, thereby advancing our understanding of trigeminal neuralgia (TN) pathogenesis. Employing the Gene Expression Omnibus (GEO) database, we extracted microarray data pertinent to TN to identify differentially expressed genes (DEGs). A subsequent comparison with SOCE-related genes from the Genecards database helped pinpoint potential target genes. The STRING database facilitated protein-protein interaction (PPI) analysis to spotlight STIM1 as a gene of interest in TN. Through histological staining, transmission electron microscopy (TEM), and behavioral assessments, we probed STIM1's pathological effects on TN in rat models. Additionally, we examined STIM1's influence on the SOCE pathway in trigeminal ganglion cells using techniques like calcium content measurement, patch clamp electrophysiology, and STIM1- ORAI1 co-localization studies. Changes in the expression of inflammatory markers (TNF-α, IL-1β, IL-6) in T cells were quantified using Western blot (WB) and enzyme-linked immunosorbent assay (ELISA) <i>in vitro</i>, while immunohistochemistry and flow cytometry were applied <i>in vivo</i> to assess these cytokines and T cell count alterations. Our bioinformatic approach highlighted STIM1's significant overexpression in TN patients, underscoring its pivotal role in TN's etiology and progression. Experimental findings from both <i>in vitro</i> and <i>in vivo</i> studies corroborated STIM1's regulatory influence on the SOCE pathway. Furthermore, STIM1 was shown to mediate SOCE-induced inflammatory cytokine release in T lymphocytes, a critical factor in TN development. Supportive evidence from histological, ultrastructural, and behavioral analyses reinforced the link between STIM1-mediated SOCE and T lymphocyte-driven inflammation in TN pathogenesis. This study presents novel evidence that STIM1 is a key regulator of SOCE and inflammatory cytokine release in T lymphocytes, contributing significantly to the pathogenesis of trigeminal neuralgia. Our findings not only deepen the understanding of TN's molecular underpinnings but also potentially open new avenues for targeted therapeutic strategies.</p>","PeriodicalId":12630,"journal":{"name":"Frontiers in Molecular Neuroscience","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11221526/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141497815","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
Editorial: Molecular and cellular mechanisms of sensory functions in insect models, volume II. 社论:昆虫模型感官功能的分子和细胞机制,第二卷。
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-17 eCollection Date: 2024-01-01 DOI: 10.3389/fnmol.2024.1443041
Takaaki Sokabe, Jia Huang, Youngseok Lee
{"title":"Editorial: Molecular and cellular mechanisms of sensory functions in insect models, volume II.","authors":"Takaaki Sokabe, Jia Huang, Youngseok Lee","doi":"10.3389/fnmol.2024.1443041","DOIUrl":"https://doi.org/10.3389/fnmol.2024.1443041","url":null,"abstract":"","PeriodicalId":12630,"journal":{"name":"Frontiers in Molecular Neuroscience","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11215166/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141476410","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
Editorial: Restoring neural circuits after spinal cord injury. 社论:恢复脊髓损伤后的神经回路。
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-17 eCollection Date: 2024-01-01 DOI: 10.3389/fnmol.2024.1428164
Aikeremujiang Muheremu, Jianjun Wu
{"title":"Editorial: Restoring neural circuits after spinal cord injury.","authors":"Aikeremujiang Muheremu, Jianjun Wu","doi":"10.3389/fnmol.2024.1428164","DOIUrl":"10.3389/fnmol.2024.1428164","url":null,"abstract":"","PeriodicalId":12630,"journal":{"name":"Frontiers in Molecular Neuroscience","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11215169/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141476411","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
Tackling neurodegeneration in vitro with omics: a path towards new targets and drugs. 利用 omics 解决体外神经变性问题:通往新目标和新药物之路。
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-17 eCollection Date: 2024-01-01 DOI: 10.3389/fnmol.2024.1414886
Caterina Carraro, Jessica V Montgomery, Julien Klimmt, Dominik Paquet, Joachim L Schultze, Marc D Beyer

Drug discovery is a generally inefficient and capital-intensive process. For neurodegenerative diseases (NDDs), the development of novel therapeutics is particularly urgent considering the long list of late-stage drug candidate failures. Although our knowledge on the pathogenic mechanisms driving neurodegeneration is growing, additional efforts are required to achieve a better and ultimately complete understanding of the pathophysiological underpinnings of NDDs. Beyond the etiology of NDDs being heterogeneous and multifactorial, this process is further complicated by the fact that current experimental models only partially recapitulate the major phenotypes observed in humans. In such a scenario, multi-omic approaches have the potential to accelerate the identification of new or repurposed drugs against a multitude of the underlying mechanisms driving NDDs. One major advantage for the implementation of multi-omic approaches in the drug discovery process is that these overarching tools are able to disentangle disease states and model perturbations through the comprehensive characterization of distinct molecular layers (i.e., genome, transcriptome, proteome) up to a single-cell resolution. Because of recent advances increasing their affordability and scalability, the use of omics technologies to drive drug discovery is nascent, but rapidly expanding in the neuroscience field. Combined with increasingly advanced in vitro models, which particularly benefited from the introduction of human iPSCs, multi-omics are shaping a new paradigm in drug discovery for NDDs, from disease characterization to therapeutics prediction and experimental screening. In this review, we discuss examples, main advantages and open challenges in the use of multi-omic approaches for the in vitro discovery of targets and therapies against NDDs.

药物研发通常是一个低效和资本密集型的过程。对于神经退行性疾病(NDDs)而言,考虑到后期候选药物失败的案例不胜枚举,开发新型疗法显得尤为迫切。尽管我们对驱动神经退行性变的致病机制的了解在不断加深,但要更好地、最终全面地了解 NDD 的病理生理学基础,还需要付出更多的努力。除了 NDD 的病因是多因素的异质性病因外,目前的实验模型只能部分再现在人类身上观察到的主要表型这一事实也使这一过程变得更加复杂。在这种情况下,多组学方法有可能加快鉴定针对驱动 NDDs 的多种潜在机制的新药或改用药物。在药物发现过程中采用多组学方法的一个主要优势是,这些总体工具能够通过对不同分子层(即基因组、转录组、蛋白质组)的全面表征来分解疾病状态和模型扰动,直至单细胞分辨率。由于最近的进步提高了这些技术的可负担性和可扩展性,利用 omics 技术推动药物发现的工作刚刚起步,但在神经科学领域正在迅速扩展。结合日益先进的体外模型,特别是受益于人类 iPSCs 的引入,多组学正在塑造一种新的 NDD 药物发现范式,从疾病特征描述到治疗预测和实验筛选。在这篇综述中,我们将讨论使用多组学方法体外发现 NDDs 靶点和疗法的实例、主要优势和面临的挑战。
{"title":"Tackling neurodegeneration <i>in vitro</i> with omics: a path towards new targets and drugs.","authors":"Caterina Carraro, Jessica V Montgomery, Julien Klimmt, Dominik Paquet, Joachim L Schultze, Marc D Beyer","doi":"10.3389/fnmol.2024.1414886","DOIUrl":"10.3389/fnmol.2024.1414886","url":null,"abstract":"<p><p>Drug discovery is a generally inefficient and capital-intensive process. For neurodegenerative diseases (NDDs), the development of novel therapeutics is particularly urgent considering the long list of late-stage drug candidate failures. Although our knowledge on the pathogenic mechanisms driving neurodegeneration is growing, additional efforts are required to achieve a better and ultimately complete understanding of the pathophysiological underpinnings of NDDs. Beyond the etiology of NDDs being heterogeneous and multifactorial, this process is further complicated by the fact that current experimental models only partially recapitulate the major phenotypes observed in humans. In such a scenario, multi-omic approaches have the potential to accelerate the identification of new or repurposed drugs against a multitude of the underlying mechanisms driving NDDs. One major advantage for the implementation of multi-omic approaches in the drug discovery process is that these overarching tools are able to disentangle disease states and model perturbations through the comprehensive characterization of distinct molecular layers (i.e., genome, transcriptome, proteome) up to a single-cell resolution. Because of recent advances increasing their affordability and scalability, the use of omics technologies to drive drug discovery is nascent, but rapidly expanding in the neuroscience field. Combined with increasingly advanced <i>in vitro</i> models, which particularly benefited from the introduction of human iPSCs, multi-omics are shaping a new paradigm in drug discovery for NDDs, from disease characterization to therapeutics prediction and experimental screening. In this review, we discuss examples, main advantages and open challenges in the use of multi-omic approaches for the <i>in vitro</i> discovery of targets and therapies against NDDs.</p>","PeriodicalId":12630,"journal":{"name":"Frontiers in Molecular Neuroscience","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11215216/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141476440","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
Recovery kinetics of dual AAV-mediated human otoferlin expression. 双 AAV 介导的人类奥托费林表达的恢复动力学。
IF 3.5 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-17 eCollection Date: 2024-01-01 DOI: 10.3389/fnmol.2024.1376128
Jonathan B Sellon, Kathy S So, Andrew D'Arcangelo, Sarah Cancelarich, Meghan C Drummond, Peter G Slade, Ning Pan, Tyler M Gibson, Tian Yang, Joseph C Burns, Adam T Palermo, Lars Becker

Deafness-causing deficiencies in otoferlin (OTOF) have been addressed preclinically using dual adeno-associated virus (AAV)-based approaches. However, timing of transduction, recombination of mRNA, and protein expression with dual hybrid AAV methods methods have not previously been characterized. Here, we have established an ex vivo assay to determine the kinetics of dual-AAV mediated expression of OTOF in hair cells of the mouse utricle. We utilized two different recombinant vectors that comprise DB-OTO, one containing the 5' portion of OTOF under the control of the hair cell-specific Myo15 promoter, and the other the 3' portion of OTOF. We explored specificity of the Myo15 promoter in hair cells of the mouse utricle, established dose response characteristics of DB-OTO ex vivo in an OTOF-deficient mouse model, and demonstrated tolerability of AAV1 in utricular hair cells. Furthermore, we established deviations from a one-to-one ratio of 5' to 3' vectors with little impact on recombined OTOF. Finally, we established a plateau in quantity of recombined OTOF mRNA and protein expression by 14 to 21 days ex vivo with comparable recovery timing to that in vivo model. These findings demonstrate the utility of an ex vivo model system for exploring expression kinetics and establish in vivo and ex vivo recovery timing of dual AAV-mediated OTOF expression.

基于双重腺相关病毒(AAV)的方法已在临床前解决了奥托费林(OTOF)致聋缺陷的问题。然而,使用双杂交 AAV 方法进行转导、mRNA 重组和蛋白质表达的时机尚未确定。在此,我们建立了一种体内外试验,以确定双 AAV 介导的 OTOF 在小鼠胞顶部毛细胞中的表达动力学。我们使用了两种不同的重组载体,其中一种包含在毛细胞特异性 Myo15 启动子控制下的 OTOF 的 5' 部分,另一种包含 OTOF 的 3' 部分。我们探索了小鼠胞顶部毛细胞中 Myo15 启动子的特异性,在 OTOF 缺失的小鼠模型中确定了 DB-OTO 的体内外剂量反应特性,并证明了 AAV1 在胞顶部毛细胞中的耐受性。此外,我们还确定了 5' 与 3' 载体一一对应比例的偏差对重组 OTOF 的影响很小。最后,我们确定了重组 OTOF mRNA 和蛋白表达量在体外 14 到 21 天达到高峰,恢复时间与体内模型相当。这些发现证明了体内外模型系统对探索表达动力学的实用性,并确定了体内外双 AAV 介导的 OTOF 表达的恢复时间。
{"title":"Recovery kinetics of dual AAV-mediated human otoferlin expression.","authors":"Jonathan B Sellon, Kathy S So, Andrew D'Arcangelo, Sarah Cancelarich, Meghan C Drummond, Peter G Slade, Ning Pan, Tyler M Gibson, Tian Yang, Joseph C Burns, Adam T Palermo, Lars Becker","doi":"10.3389/fnmol.2024.1376128","DOIUrl":"10.3389/fnmol.2024.1376128","url":null,"abstract":"<p><p>Deafness-causing deficiencies in <i>otoferlin</i> (<i>OTOF</i>) have been addressed preclinically using dual adeno-associated virus (AAV)-based approaches. However, timing of transduction, recombination of mRNA, and protein expression with dual hybrid AAV methods methods have not previously been characterized. Here, we have established an <i>ex vivo</i> assay to determine the kinetics of dual-AAV mediated expression of <i>OTOF</i> in hair cells of the mouse utricle. We utilized two different recombinant vectors that comprise DB-OTO, one containing the 5' portion of <i>OTOF</i> under the control of the hair cell-specific <i>Myo15</i> promoter, and the other the 3' portion of <i>OTOF</i>. We explored specificity of the <i>Myo15</i> promoter in hair cells of the mouse utricle, established dose response characteristics of DB-OTO <i>ex vivo</i> in an OTOF-deficient mouse model, and demonstrated tolerability of AAV1 in utricular hair cells. Furthermore, we established deviations from a one-to-one ratio of 5' to 3' vectors with little impact on recombined <i>OTOF</i>. Finally, we established a plateau in quantity of recombined <i>OTOF</i> mRNA and protein expression by 14 to 21 days <i>ex vivo</i> with comparable recovery timing to that <i>in vivo</i> model. These findings demonstrate the utility of an <i>ex vivo</i> model system for exploring expression kinetics and establish <i>in vivo</i> and <i>ex vivo</i> recovery timing of dual AAV-mediated <i>OTOF</i> expression.</p>","PeriodicalId":12630,"journal":{"name":"Frontiers in Molecular Neuroscience","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11215969/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141476412","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
Frontiers | hnRNPs: roles in neurodevelopment and implication for brain disorders hnRNPs前沿:在神经发育中的作用及对脑部疾病的影响
IF 4.8 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-17 DOI: 10.3389/fnmol.2024.1411639
Pierre Tilliole, Simon Fix, Juliette D. GODIN
Heterogeneous nuclear ribonucleoproteins (hnRNPs) constitute a family of multifunctional RNA-binding proteins able to process nuclear pre-mRNAs into mature mRNAs and regulate gene expression in multiple ways. They comprise at least 20 different members in mammals, named from A (HNRNP A1) to U (HNRNP U). Many of these proteins are components of the spliceosome complex and can modulate alternative splicing in a tissue-specific manner. Notably, while genes encoding hnRNPs exhibit ubiquitous expression, increasing evidence associate these proteins to various neurodevelopmental and neurodegenerative disorders, such as intellectual disability, epilepsy, microcephaly, amyotrophic lateral sclerosis, or dementias, highlighting their crucial role in the central nervous system. This review explores the evolution of the hnRNPs family, highlighting the emergence of numerous new members within this family, and sheds light on their implications for brain development.
异质核核糖核蛋白(hnRNPs)是一个多功能 RNA 结合蛋白家族,能够将核前 mRNA 处理成成熟的 mRNA,并以多种方式调控基因表达。哺乳动物中至少有 20 个不同的成员,从 A(HNRNP A1)到 U(HNRNP U)依次命名。其中许多蛋白是剪接体复合物的组成成分,能以组织特异性的方式调节替代剪接。值得注意的是,虽然编码 hnRNPs 的基因无处不在,但越来越多的证据表明,这些蛋白与各种神经发育和神经退行性疾病有关,如智力障碍、癫痫、小头畸形、肌萎缩性脊髓侧索硬化症或痴呆症等,突显了它们在中枢神经系统中的关键作用。这篇综述探讨了 hnRNPs 家族的进化,强调了该家族中出现的众多新成员,并揭示了它们对大脑发育的影响。
{"title":"Frontiers | hnRNPs: roles in neurodevelopment and implication for brain disorders","authors":"Pierre Tilliole, Simon Fix, Juliette D. GODIN","doi":"10.3389/fnmol.2024.1411639","DOIUrl":"https://doi.org/10.3389/fnmol.2024.1411639","url":null,"abstract":"Heterogeneous nuclear ribonucleoproteins (hnRNPs) constitute a family of multifunctional RNA-binding proteins able to process nuclear pre-mRNAs into mature mRNAs and regulate gene expression in multiple ways. They comprise at least 20 different members in mammals, named from A (HNRNP A1) to U (HNRNP U). Many of these proteins are components of the spliceosome complex and can modulate alternative splicing in a tissue-specific manner. Notably, while genes encoding hnRNPs exhibit ubiquitous expression, increasing evidence associate these proteins to various neurodevelopmental and neurodegenerative disorders, such as intellectual disability, epilepsy, microcephaly, amyotrophic lateral sclerosis, or dementias, highlighting their crucial role in the central nervous system. This review explores the evolution of the hnRNPs family, highlighting the emergence of numerous new members within this family, and sheds light on their implications for brain development.","PeriodicalId":12630,"journal":{"name":"Frontiers in Molecular Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.8,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141721582","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
Frontiers | Copper Toxicity and Deficiency: The Vicious Cycle at the Core of Protein Aggregation in ALS 铜的毒性和缺乏:ALS蛋白聚集核心的恶性循环
IF 4.8 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-14 DOI: 10.3389/fnmol.2024.1408159
Jin-Hong Min, Robert A. Harris, Heela Sarlus
The pathophysiology of ALS involves many signs of a disruption in copper homeostasis, with both excess free levels and functional deficiency likely occurring simultaneously. This is crucial, as many important physiological functions are performed by cuproenzymes. While it is unsurprising that many ALS symptoms are related to signs of copper deficiency, resulting in vascular, antioxidant system and mitochondrial oxidative respiration deficiencies, there are also signs of copper toxicity such as ROS generation and enhanced protein aggregation. We discuss how copper also plays a key role in proteostasis and interacts either directly or indirectly with many of the key aggregate-prone proteins implicated in ALS, such as TDP-43, C9ORF72, SOD1 and FUS as well as the effect of their aggregation on copper homeostasis. We suggest that loss of cuproprotein function is at the core of ALS pathology, a condition that is driven by a combination of unbound copper and ROS that can either initiate and/or accelerate protein aggregation. This could trigger a positive feedback cycle whereby protein aggregates trigger the aggregation of other proteins in a chain reaction that eventually captures elements of the proteostatic mechanisms in place to counteract them. The end result is an abundance of aggregated non-functional cuproproteins and chaperones alongside depleted intracellular copper stores, resulting in a general lack of cuproenzyme function. We then discuss the possible aetiology of ALS and illustrate how strong risk factors including environmental toxins such as BMAA and heavy metals can functionally behave to promote protein aggregation and disturb copper metabolism that likely drives this vicious cycle in sporadic ALS. From this synthesis, we propose restoration of copper balance using copper delivery agents in combination with chaperones/chaperone mimetics, perhaps in conjunction with the neuroprotective amino acid serine, as a promising strategy in the treatment of this incurable disease.
渐冻人症的病理生理学涉及铜平衡紊乱的许多迹象,游离水平过高和功能性缺乏可能同时发生。这一点至关重要,因为许多重要的生理功能都是由铜酵素实现的。许多渐冻人症的症状与铜缺乏有关,导致血管、抗氧化系统和线粒体氧化呼吸不足,这一点不足为奇,但同时也存在铜中毒的迹象,如 ROS 生成和蛋白质聚集增强。我们讨论了铜如何在蛋白稳态中发挥关键作用,如何直接或间接与许多与 ALS 有关的易聚集的关键蛋白(如 TDP-43、C9ORF72、SOD1 和 FUS)相互作用,以及它们的聚集对铜稳态的影响。我们认为,铜蛋白功能的丧失是渐冻症病理学的核心,这种病症是由未结合的铜和 ROS 共同驱动的,而 ROS 可以启动和/或加速蛋白质的聚集。这可能会引发一种正反馈循环,即蛋白质聚集引发其他蛋白质的聚集,这种连锁反应最终会捕捉到蛋白质静态机制中的元素来抵消它们。最终的结果是,大量聚集的无功能铜蛋白和伴侣蛋白以及细胞内铜储存耗尽,导致铜酵素功能普遍缺乏。然后,我们讨论了渐冻症的可能病因,并说明了包括环境毒素(如 BMAA 和重金属)在内的强风险因素如何在功能上促进蛋白质聚集和扰乱铜代谢,从而可能在散发性渐冻症中推动这一恶性循环。综上所述,我们建议将铜输送剂与伴侣蛋白/伴侣蛋白仿制药结合使用,或许与神经保护性氨基酸丝氨酸结合使用,以恢复铜平衡,作为治疗这种不治之症的一种有前途的策略。
{"title":"Frontiers | Copper Toxicity and Deficiency: The Vicious Cycle at the Core of Protein Aggregation in ALS","authors":"Jin-Hong Min, Robert A. Harris, Heela Sarlus","doi":"10.3389/fnmol.2024.1408159","DOIUrl":"https://doi.org/10.3389/fnmol.2024.1408159","url":null,"abstract":"The pathophysiology of ALS involves many signs of a disruption in copper homeostasis, with both excess free levels and functional deficiency likely occurring simultaneously. This is crucial, as many important physiological functions are performed by cuproenzymes. While it is unsurprising that many ALS symptoms are related to signs of copper deficiency, resulting in vascular, antioxidant system and mitochondrial oxidative respiration deficiencies, there are also signs of copper toxicity such as ROS generation and enhanced protein aggregation. We discuss how copper also plays a key role in proteostasis and interacts either directly or indirectly with many of the key aggregate-prone proteins implicated in ALS, such as TDP-43, C9ORF72, SOD1 and FUS as well as the effect of their aggregation on copper homeostasis. We suggest that loss of cuproprotein function is at the core of ALS pathology, a condition that is driven by a combination of unbound copper and ROS that can either initiate and/or accelerate protein aggregation. This could trigger a positive feedback cycle whereby protein aggregates trigger the aggregation of other proteins in a chain reaction that eventually captures elements of the proteostatic mechanisms in place to counteract them. The end result is an abundance of aggregated non-functional cuproproteins and chaperones alongside depleted intracellular copper stores, resulting in a general lack of cuproenzyme function. We then discuss the possible aetiology of ALS and illustrate how strong risk factors including environmental toxins such as BMAA and heavy metals can functionally behave to promote protein aggregation and disturb copper metabolism that likely drives this vicious cycle in sporadic ALS. From this synthesis, we propose restoration of copper balance using copper delivery agents in combination with chaperones/chaperone mimetics, perhaps in conjunction with the neuroprotective amino acid serine, as a promising strategy in the treatment of this incurable disease.","PeriodicalId":12630,"journal":{"name":"Frontiers in Molecular Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.8,"publicationDate":"2024-06-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141576867","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
Frontiers | Disease-Modifying rdHSV-CA8* Non-Opioid Analgesic Gene Therapy Treats Chronic Osteoarthritis Pain by Activating Kv7 Voltage-Gated Potassium Channels 疾病修饰rdHSV-CA8*非阿片类镇痛剂基因疗法通过激活Kv7电压门控钾通道治疗慢性骨关节炎疼痛
IF 4.8 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-13 DOI: 10.3389/fnmol.2024.1416148
Gerald Z. Zhuang, William F. Goins, Munal Kandel, Marco Marzulli, Mingdi Zhang, Joseph C. Glorioso, Yuan Kang, Alexandra E. Levitt, Konstantinos D. Sarantopoulos, Roy C. Levitt
Chronic pain is common in our population, and most of these patients are inadequately treated, making the development of safer analgesics a high priority. Knee osteoarthritis (OA) is a primary cause of chronic pain and disability worldwide, and lower extremity OA is a major contributor to loss of quality-adjusted life-years. In this study we tested the hypothesis that a novel JDNI8 replication-defective herpes simplex-1 viral vector (rdHSV) incorporating a modified carbonic anhydrase-8 transgene (CA8*) produces analgesia and treats monoiodoacetate-induced (MIA) chronic knee pain due to OA. We observed transduction of lumbar DRG sensory neurons with these viral constructs (vHCA8*) (~40% of advillin-positive cells and ~ 50% of TrkA-positive cells colocalized with V5-positive cells) using the intra-articular (IA) knee joint (KJ) route of administration. vHCA8* inhibited chronic mechanical OA knee pain induced by MIA was dose- and time-dependent. Mechanical thresholds returned to Baseline by D17 after IA KJ vHCA8* treatment, and exceeded Baseline (analgesia) through D65, whereas negative controls failed to reach Baseline responses. Weight-bearing and automated voluntary wheel running were improved by vHCA8*, but not negative controls. Kv7 voltage-gated potassium channel-specific inhibitor XE-991 reversed vHCA8*-induced analgesia. Using IHC, IA KJ of vHCA8* activated DRG Kv7 channels via dephosphorylation, but negative controls failed to impact Kv7 channels. XE-991 stimulated Kv7.2–7.5 and Kv7.3 phosphorylation using western blotting of differentiated SH-SY5Y cells, which was inhibited by vHCA8* but not by negative controls. The observed prolonged dose-dependent therapeutic effects of IA KJ administration of vHCA8* on MIA-induced chronic KJ pain due to OA is consistent with the specific activation of Kv7 channels in small DRG sensory neurons. Together, these data demonstrate for the first-time local IA KJ administration of vHCA8* produces opioid-independent analgesia in this MIA-induced OA chronic pain model, supporting further therapeutic development.
慢性疼痛在我国人口中很常见,但大多数患者都没有得到适当的治疗,因此开发更安全的镇痛药成为当务之急。膝关节骨关节炎(OA)是全球慢性疼痛和残疾的主要原因,下肢OA是造成质量调整生命年损失的主要因素。在这项研究中,我们测试了这样一个假设:一种新型的 JDNI8 复制缺陷单纯疱疹-1 病毒载体(rdHSV)含有改良的碳酸酐酶-8 转基因(CA8*),它能产生镇痛作用并治疗单碘乙酸盐诱导的慢性膝关节疼痛。我们通过膝关节内(IA)给药途径观察到腰椎DRG感觉神经元转导了这些病毒构建体(vHCA8*)(约40%的Advillin阳性细胞和约50%的TrkA阳性细胞与V5阳性细胞共定位)。经IA KJ vHCA8*治疗后,机械阈值在D17时恢复到基线,并在D65时超过基线(镇痛),而阴性对照组未能达到基线反应。vHCA8*能改善负重和自动自主轮跑,而阴性对照组则不能。Kv7 电压门控钾通道特异性抑制剂 XE-991 逆转了 vHCA8* 诱导的镇痛。通过 IHC 检测,vHCA8* 的 IA KJ 可通过去磷酸化激活 DRG Kv7 通道,但阴性对照组未能影响 Kv7 通道。通过对分化的SH-SY5Y细胞进行Western印迹,XE-991可刺激Kv7.2-7.5和Kv7.3磷酸化,vHCA8*可抑制磷酸化,而阴性对照组则不能抑制磷酸化。vHCA8* 的 IA KJ 给药对 OA 引起的 MIA 诱导的慢性 KJ 疼痛具有长期剂量依赖性治疗效果,这与 Kv7 通道在小 DRG 感觉神经元中的特异性激活是一致的。总之,这些数据首次证明了在MIA诱导的OA慢性疼痛模型中,局部IA KJ给药vHCA8*可产生阿片类药物依赖性镇痛,支持进一步的治疗开发。
{"title":"Frontiers | Disease-Modifying rdHSV-CA8* Non-Opioid Analgesic Gene Therapy Treats Chronic Osteoarthritis Pain by Activating Kv7 Voltage-Gated Potassium Channels","authors":"Gerald Z. Zhuang, William F. Goins, Munal Kandel, Marco Marzulli, Mingdi Zhang, Joseph C. Glorioso, Yuan Kang, Alexandra E. Levitt, Konstantinos D. Sarantopoulos, Roy C. Levitt","doi":"10.3389/fnmol.2024.1416148","DOIUrl":"https://doi.org/10.3389/fnmol.2024.1416148","url":null,"abstract":"Chronic pain is common in our population, and most of these patients are inadequately treated, making the development of safer analgesics a high priority. Knee osteoarthritis (OA) is a primary cause of chronic pain and disability worldwide, and lower extremity OA is a major contributor to loss of quality-adjusted life-years. In this study we tested the hypothesis that a novel JDNI8 replication-defective herpes simplex-1 viral vector (rdHSV) incorporating a modified carbonic anhydrase-8 transgene (CA8*) produces analgesia and treats monoiodoacetate-induced (MIA) chronic knee pain due to OA. We observed transduction of lumbar DRG sensory neurons with these viral constructs (vHCA8*) (~40% of advillin-positive cells and ~ 50% of TrkA-positive cells colocalized with V5-positive cells) using the intra-articular (IA) knee joint (KJ) route of administration. vHCA8* inhibited chronic mechanical OA knee pain induced by MIA was dose- and time-dependent. Mechanical thresholds returned to Baseline by D17 after IA KJ vHCA8* treatment, and exceeded Baseline (analgesia) through D65, whereas negative controls failed to reach Baseline responses. Weight-bearing and automated voluntary wheel running were improved by vHCA8*, but not negative controls. Kv7 voltage-gated potassium channel-specific inhibitor XE-991 reversed vHCA8*-induced analgesia. Using IHC, IA KJ of vHCA8* activated DRG Kv7 channels via dephosphorylation, but negative controls failed to impact Kv7 channels. XE-991 stimulated Kv7.2–7.5 and Kv7.3 phosphorylation using western blotting of differentiated SH-SY5Y cells, which was inhibited by vHCA8* but not by negative controls. The observed prolonged dose-dependent therapeutic effects of IA KJ administration of vHCA8* on MIA-induced chronic KJ pain due to OA is consistent with the specific activation of Kv7 channels in small DRG sensory neurons. Together, these data demonstrate for the first-time local IA KJ administration of vHCA8* produces opioid-independent analgesia in this MIA-induced OA chronic pain model, supporting further therapeutic development.","PeriodicalId":12630,"journal":{"name":"Frontiers in Molecular Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.8,"publicationDate":"2024-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141721581","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
Frontiers | Context-dependent hyperactivity in syngap1a and syngap1b zebrafish models of SYNGAP1-related disorder 前沿 | SYNGAP1相关障碍的syngap1a和syngap1b斑马鱼模型中的内涵依赖性多动症
IF 4.8 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-06-04 DOI: 10.3389/fnmol.2024.1401746
Sureni H. Sumathipala, Suha Khan, Robert A. Kozol, Yoichi Araki, Sheyum Syed, Richard L. Huganir, Julia E. Dallman
Background and aimsSYNGAP1-related disorder (SYNGAP1-RD) is a prevalent genetic form of Autism Spectrum Disorder and Intellectual Disability (ASD/ID) and is caused by de novo or inherited mutations in one copy of the SYNGAP1 gene. In addition to ASD/ID, SYNGAP1 disorder is associated with comorbid symptoms including treatment-resistant-epilepsy, sleep disturbances, and gastrointestinal distress. Mechanistic links between these diverse symptoms and SYNGAP1 variants remain obscure, therefore, our goal was to generate a zebrafish model in which this range of symptoms can be studied.MethodsWe used CRISPR/Cas9 to introduce frameshift mutations in the syngap1a and syngap1b zebrafish duplicates (syngap1ab) and validated these stable models for Syngap1 loss-of-function. Because SYNGAP1 is extensively spliced, we mapped splice variants to the two zebrafish syngap1a and b genes and identified mammalian-like isoforms. We then quantified locomotory behaviors in zebrafish syngap1ab larvae under three conditions that normally evoke different arousal states in wild-type larvae: aversive, high-arousal acoustic, medium-arousal dark, and low-arousal light stimuli.ResultsWe show that CRISPR/Cas9 indels in zebrafish syngap1a and syngap1b produced loss-of-function alleles at RNA and protein levels. Our analyses of zebrafish Syngap1 isoforms showed that, as in mammals, zebrafish Syngap1 N- and C-termini are extensively spliced. We identified a zebrafish syngap1 α1-like variant that maps exclusively to the syngap1b gene. Quantifying locomotor behaviors showed that syngap1ab mutant larvae are hyperactive compared to wild-type but to differing degrees depending on the stimulus. Hyperactivity was most pronounced in low arousal settings, and hyperactivity was proportional to the number of mutant syngap1 alleles.LimitationsSyngap1 loss-of-function mutations produce relatively subtle phenotypes in zebrafish compared to mammals. For example, while mouse Syngap1 homozygotes die at birth, zebrafish syngap1ab−/− survive to adulthood and are fertile, thus some aspects of symptoms in people with SYNGAP1-Related Disorder are not likely to be reflected in zebrafish.ConclusionOur data support mutations in zebrafish syngap1ab as causal for hyperactivity associated with elevated arousal that is especially pronounced in low-arousal environments.
背景和目的SYNGAP1相关障碍(SYNGAP1-RD)是自闭症谱系障碍和智力障碍(ASD/ID)的一种常见遗传形式,由SYNGAP1基因的一个拷贝发生新突变或遗传突变引起。除 ASD/ID 外,SYNGAP1 紊乱症还伴有一些合并症状,包括抗药性癫痫、睡眠障碍和肠胃不适。我们使用 CRISPR/Cas9 在 syngap1a 和 syngap1b 斑马鱼复制品(syngap1ab)中引入框移突变,并验证了这些稳定的 Syngap1 功能缺失模型。由于 SYNGAP1 的剪接范围很广,我们将剪接变体映射到两个斑马鱼 syngap1a 和 b 基因上,并确定了类似哺乳动物的同工型。然后,我们对斑马鱼 syngap1ab 幼体在三种条件下的运动行为进行了量化,这三种条件通常会诱发野生型幼体的不同唤醒状态:厌恶、高唤醒声刺激、中唤醒暗刺激和低唤醒光刺激。我们对斑马鱼 Syngap1 异构体的分析表明,与哺乳动物一样,斑马鱼 Syngap1 的 N 端和 C 端也是广泛剪接的。我们发现了一种斑马鱼 Syngap1 α1样变体,它只映射到 syngap1b 基因上。定量运动行为显示,与野生型相比,syngap1ab突变体幼体运动亢进,但亢进程度因刺激而异。与哺乳动物相比,斑马鱼的Syngap1功能缺失突变会产生相对微妙的表型。例如,小鼠Syngap1同源基因型的斑马鱼在出生时就会死亡,而syngap1ab-/-斑马鱼却能存活到成年并具有生育能力,因此SYNGAP1相关障碍患者的某些症状在斑马鱼中可能无法反映出来。结论我们的数据支持斑马鱼syngap1ab突变是导致唤醒水平升高相关多动的原因,这种多动在低唤醒水平环境中尤为明显。
{"title":"Frontiers | Context-dependent hyperactivity in syngap1a and syngap1b zebrafish models of SYNGAP1-related disorder","authors":"Sureni H. Sumathipala, Suha Khan, Robert A. Kozol, Yoichi Araki, Sheyum Syed, Richard L. Huganir, Julia E. Dallman","doi":"10.3389/fnmol.2024.1401746","DOIUrl":"https://doi.org/10.3389/fnmol.2024.1401746","url":null,"abstract":"Background and aimsSYNGAP1-related disorder (SYNGAP1-RD) is a prevalent genetic form of Autism Spectrum Disorder and Intellectual Disability (ASD/ID) and is caused by de novo or inherited mutations in one copy of the SYNGAP1 gene. In addition to ASD/ID, SYNGAP1 disorder is associated with comorbid symptoms including treatment-resistant-epilepsy, sleep disturbances, and gastrointestinal distress. Mechanistic links between these diverse symptoms and SYNGAP1 variants remain obscure, therefore, our goal was to generate a zebrafish model in which this range of symptoms can be studied.MethodsWe used CRISPR/Cas9 to introduce frameshift mutations in the syngap1a and syngap1b zebrafish duplicates (syngap1ab) and validated these stable models for Syngap1 loss-of-function. Because SYNGAP1 is extensively spliced, we mapped splice variants to the two zebrafish syngap1a and b genes and identified mammalian-like isoforms. We then quantified locomotory behaviors in zebrafish syngap1ab larvae under three conditions that normally evoke different arousal states in wild-type larvae: aversive, high-arousal acoustic, medium-arousal dark, and low-arousal light stimuli.ResultsWe show that CRISPR/Cas9 indels in zebrafish syngap1a and syngap1b produced loss-of-function alleles at RNA and protein levels. Our analyses of zebrafish Syngap1 isoforms showed that, as in mammals, zebrafish Syngap1 N- and C-termini are extensively spliced. We identified a zebrafish syngap1 α1-like variant that maps exclusively to the syngap1b gene. Quantifying locomotor behaviors showed that syngap1ab mutant larvae are hyperactive compared to wild-type but to differing degrees depending on the stimulus. Hyperactivity was most pronounced in low arousal settings, and hyperactivity was proportional to the number of mutant syngap1 alleles.LimitationsSyngap1 loss-of-function mutations produce relatively subtle phenotypes in zebrafish compared to mammals. For example, while mouse Syngap1 homozygotes die at birth, zebrafish syngap1ab−/− survive to adulthood and are fertile, thus some aspects of symptoms in people with SYNGAP1-Related Disorder are not likely to be reflected in zebrafish.ConclusionOur data support mutations in zebrafish syngap1ab as causal for hyperactivity associated with elevated arousal that is especially pronounced in low-arousal environments.","PeriodicalId":12630,"journal":{"name":"Frontiers in Molecular Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.8,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141576868","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
Frontiers in Molecular 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