首页 > 最新文献

Frontiers in Cellular Neuroscience最新文献

英文 中文
Impaired olfactory performance and anxiety-like behavior in a rat model of multiple sclerosis are associated with enhanced adenosine signaling in the olfactory bulb via A1R, A2BR, and A3R 多发性硬化症大鼠模型的嗅觉表现和焦虑样行为受损与嗅球中通过 A1R、A2BR 和 A3R 的腺苷信号增强有关
IF 5.3 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-07-30 DOI: 10.3389/fncel.2024.1407975
Andjela Stekic, Milorad Dragic, Jelena Stanojevic, Marina Zaric Kontic, Ivana Stevanovic, Milica Zeljkovic Jovanovic, Katarina Mihajlovic, Nadezda Nedeljkovic
The present study shows that animals with experimental autoimmune encephalomyelitis (EAE) exhibit olfactory dysfunction and impaired general cognitive abilities, as well as anxiety-like behavior. Olfactory dysfunction occurs on average at 2 dpi, well before the onset of the first motor signs of EAE (8–10 dpi). After the initial olfactory dysfunction, the EAE animals show a fluctuation in olfactory performance that resembles the relapsing–remitting course of human MS. The study also shows severe neuroinflammation in the olfactory bulb (OB), with numerous infiltrated CD4+ T cells and peripheral macrophages in the superficial OB layers, marked microgliosis, and massive induction of TNF-α, IL-1β, and IL-6. Reduced tyrosine hydroxylase activity in the glomerular layer, pronounced granule cell atrophy, and reduced numbers of type B neuroblasts in the rostral migratory stream also indicate altered plasticity of the neuronal network in the OB. Considering the exceptionally high purinome expression in the OB, the possible involvement of purinergic signaling was also investigated. The study shows that macrophages infiltrating the OB overexpress A3R, while highly reactive microglia overexpress the adenosine-producing enzyme eN/CD73 as well as A2BR, A3R, and P2X4R. Given the simultaneous induction of complement component C3, the results suggest that the microglial cells develop a functional phenotype of phagocytizing microglia. The study also demonstrates transcriptional and translational upregulation of A1R in mitral and tufted cells, which likely influence resting network activity in OB and likely contribute to olfactory dysfunction in EAE. Overall, our study shows that olfactory dysfunction and altered social and cognitive behavior in EAE are associated with increased adenosine signaling via A1R, A2BR, and A3R.
本研究表明,实验性自身免疫性脑脊髓炎(EAE)动物表现出嗅觉功能障碍、一般认知能力受损以及焦虑样行为。嗅觉功能障碍平均发生在 2 dpi,远远早于 EAE 最初的运动症状(8-10 dpi)。在最初的嗅觉功能障碍之后,EAE 动物的嗅觉表现会出现波动,这与人类多发性硬化症的复发-缓解过程相似。研究还显示,嗅球(OB)存在严重的神经炎症,OB浅层有大量CD4+ T细胞和外周巨噬细胞浸润,小神经胶质增生明显,TNF-α、IL-1β和IL-6大量诱导。肾小球层酪氨酸羟化酶活性降低,颗粒细胞明显萎缩,喙移行流中B型神经细胞数量减少,这些也表明OB神经元网络的可塑性发生了改变。考虑到 OB 中嘌呤基因组的表达量特别高,研究人员还对嘌呤能信号传导可能的参与进行了调查。研究显示,浸润 OB 的巨噬细胞过度表达 A3R,而高反应性小胶质细胞则过度表达腺苷生成酶 eN/CD73、A2BR、A3R 和 P2X4R。鉴于补体成分 C3 的同时诱导,结果表明小胶质细胞形成了吞噬小胶质细胞的功能表型。该研究还证明了有丝分裂细胞和簇细胞中 A1R 的转录和翻译上调,这可能会影响 OB 中的静息网络活动,并可能导致 EAE 中的嗅觉功能障碍。总之,我们的研究表明,EAE 的嗅觉功能障碍以及社会和认知行为的改变与通过 A1R、A2BR 和 A3R 的腺苷信号传导增加有关。
{"title":"Impaired olfactory performance and anxiety-like behavior in a rat model of multiple sclerosis are associated with enhanced adenosine signaling in the olfactory bulb via A1R, A2BR, and A3R","authors":"Andjela Stekic, Milorad Dragic, Jelena Stanojevic, Marina Zaric Kontic, Ivana Stevanovic, Milica Zeljkovic Jovanovic, Katarina Mihajlovic, Nadezda Nedeljkovic","doi":"10.3389/fncel.2024.1407975","DOIUrl":"https://doi.org/10.3389/fncel.2024.1407975","url":null,"abstract":"The present study shows that animals with experimental autoimmune encephalomyelitis (EAE) exhibit olfactory dysfunction and impaired general cognitive abilities, as well as anxiety-like behavior. Olfactory dysfunction occurs on average at 2 dpi, well before the onset of the first motor signs of EAE (8–10 dpi). After the initial olfactory dysfunction, the EAE animals show a fluctuation in olfactory performance that resembles the relapsing–remitting course of human MS. The study also shows severe neuroinflammation in the olfactory bulb (OB), with numerous infiltrated CD4<jats:sup>+</jats:sup> T cells and peripheral macrophages in the superficial OB layers, marked microgliosis, and massive induction of TNF-α, IL-1β, and IL-6. Reduced tyrosine hydroxylase activity in the glomerular layer, pronounced granule cell atrophy, and reduced numbers of type B neuroblasts in the rostral migratory stream also indicate altered plasticity of the neuronal network in the OB. Considering the exceptionally high purinome expression in the OB, the possible involvement of purinergic signaling was also investigated. The study shows that macrophages infiltrating the OB overexpress A<jats:sub>3</jats:sub>R, while highly reactive microglia overexpress the adenosine-producing enzyme eN/CD73 as well as A<jats:sub>2B</jats:sub>R, A<jats:sub>3</jats:sub>R, and P2X<jats:sub>4</jats:sub>R. Given the simultaneous induction of complement component C3, the results suggest that the microglial cells develop a functional phenotype of phagocytizing microglia. The study also demonstrates transcriptional and translational upregulation of A<jats:sub>1</jats:sub>R in mitral and tufted cells, which likely influence resting network activity in OB and likely contribute to olfactory dysfunction in EAE. Overall, our study shows that olfactory dysfunction and altered social and cognitive behavior in EAE are associated with increased adenosine signaling via A<jats:sub>1</jats:sub>R, A<jats:sub>2B</jats:sub>R, and A<jats:sub>3</jats:sub>R.","PeriodicalId":12432,"journal":{"name":"Frontiers in Cellular Neuroscience","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141873114","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
Potassium channels in animal models of post-traumatic stress disorder: mechanistic and therapeutic implications. 创伤后应激障碍动物模型中的钾通道:机理和治疗意义。
IF 4.2 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-07-30 eCollection Date: 2024-01-01 DOI: 10.3389/fncel.2024.1441514
Ravi Philip Rajkumar
{"title":"Potassium channels in animal models of post-traumatic stress disorder: mechanistic and therapeutic implications.","authors":"Ravi Philip Rajkumar","doi":"10.3389/fncel.2024.1441514","DOIUrl":"10.3389/fncel.2024.1441514","url":null,"abstract":"","PeriodicalId":12432,"journal":{"name":"Frontiers in Cellular Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.2,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11319181/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141975513","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 | Potential role of endothelial progenitor cells in the pathogenesis and treatment of cerebral aneurysm 内皮祖细胞在脑动脉瘤发病机制和治疗中的潜在作用
IF 5.3 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-07-30 DOI: 10.3389/fncel.2024.1456775
Jin Yu, Qian Du, Xiang Li, Wei Wei, Yuncun Fan, Jianjian Zhang, Jincao Chen
Cerebral aneurysm (CA) is a significant health concern that results from pathological dilations of blood vessels in the brain and can lead to severe and potentially life-threatening conditions. While the pathogenesis of CA is complex, emerging studies suggest that endothelial progenitor cells (EPCs) play a crucial role. In this paper, we conducted a comprehensive literature review to investigate the potential role of EPCs in the pathogenesis and treatment of CA. Current research indicates that a decreased count and dysfunction of EPCs disrupt the balance between endothelial dysfunction and repair, thus increasing the risk of CA formation. Reversing these EPCs abnormalities may reduce the progression of vascular degeneration after aneurysm induction, indicating EPCs as a promising target for developing new therapeutic strategies to facilitate CA repair. This has motivated researchers to develop novel treatment options, including drug applications, endovascular-combined and tissue engineering therapies. Although preclinical studies have shown promising results, there is still a considerable way to go before clinical translation and eventual benefits for patients. Nonetheless, these findings offer hope for improving the treatment and management of this condition.
脑动脉瘤(CA)是一种严重危害健康的疾病,它是脑血管病理性扩张的结果,可导致严重的、潜在的生命危险。虽然 CA 的发病机制十分复杂,但新的研究表明,内皮祖细胞(EPCs)在其中发挥着至关重要的作用。在本文中,我们进行了全面的文献综述,以研究 EPCs 在 CA 发病机制和治疗中的潜在作用。目前的研究表明,EPCs数量的减少和功能障碍会破坏内皮功能障碍和修复之间的平衡,从而增加CA形成的风险。逆转这些 EPCs 异常可能会减少动脉瘤诱发后血管变性的进展,这表明 EPCs 是开发促进 CA 修复的新治疗策略的一个很有前景的靶点。这促使研究人员开发新的治疗方案,包括药物应用、血管内结合疗法和组织工程疗法。尽管临床前研究已取得了令人鼓舞的成果,但距离临床转化和最终造福患者仍有相当长的路要走。不过,这些发现为改善这种疾病的治疗和管理带来了希望。
{"title":"Frontiers | Potential role of endothelial progenitor cells in the pathogenesis and treatment of cerebral aneurysm","authors":"Jin Yu, Qian Du, Xiang Li, Wei Wei, Yuncun Fan, Jianjian Zhang, Jincao Chen","doi":"10.3389/fncel.2024.1456775","DOIUrl":"https://doi.org/10.3389/fncel.2024.1456775","url":null,"abstract":"Cerebral aneurysm (CA) is a significant health concern that results from pathological dilations of blood vessels in the brain and can lead to severe and potentially life-threatening conditions. While the pathogenesis of CA is complex, emerging studies suggest that endothelial progenitor cells (EPCs) play a crucial role. In this paper, we conducted a comprehensive literature review to investigate the potential role of EPCs in the pathogenesis and treatment of CA. Current research indicates that a decreased count and dysfunction of EPCs disrupt the balance between endothelial dysfunction and repair, thus increasing the risk of CA formation. Reversing these EPCs abnormalities may reduce the progression of vascular degeneration after aneurysm induction, indicating EPCs as a promising target for developing new therapeutic strategies to facilitate CA repair. This has motivated researchers to develop novel treatment options, including drug applications, endovascular-combined and tissue engineering therapies. Although preclinical studies have shown promising results, there is still a considerable way to go before clinical translation and eventual benefits for patients. Nonetheless, these findings offer hope for improving the treatment and management of this condition.","PeriodicalId":12432,"journal":{"name":"Frontiers in Cellular Neuroscience","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141931156","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
A novel autism-associated KCNB1 mutation dramatically slows Kv2.1 potassium channel activation, deactivation and inactivation 一种新型自闭症相关 KCNB1 基因突变可显著减缓 Kv2.1 钾通道的激活、失活和失活速度
IF 5.3 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-07-29 DOI: 10.3389/fncel.2024.1438101
Rían W. Manville, Samantha D. Block, Claire L. Illeck, Jessica Kottmeier, Richard Sidlow, Geoffrey W. Abbott
KCNB1, on human chromosome 20q13.3, encodes the alpha subunit of the Kv2.1 voltage gated potassium channel. Kv2.1 is ubiquitously expressed throughout the brain and is critical in controlling neuronal excitability, including in the hippocampus and pyramidal neurons. Human KCNB1 mutations are known to cause global development delay or plateauing, epilepsy, and behavioral disorders. Here, we report a sibling pair with developmental delay, absence seizures, autism spectrum disorder, hypotonia, and dysmorphic features. Whole exome sequencing revealed a heterozygous variant of uncertain significance (c. 342 C&gt;A), p. (S114R) in KCNB1, encoding a serine to arginine substitution (S114R) in the N-terminal cytoplasmic region of Kv2.1. The siblings’ father demonstrated autistic features and was determined to be an obligate KCNB1 c. 342 C&gt;A carrier based on familial genetic testing results. Functional investigation of Kv2.1-S114R using cellular electrophysiology revealed slowing of channel activation, deactivation, and inactivation, resulting in increased net current after longer membrane depolarizations. To our knowledge, this is the first study of its kind that compares the presentation of siblings each with a KCNB1 disorder. Our study demonstrates that Kv2.1-S114R has profound cellular and phenotypic consequences. Understanding the mechanisms underlying KCNB1-linked disorders aids clinicians in diagnosis and treatment and provides potential therapeutic avenues to pursue.
KCNB1 位于人类染色体 20q13.3 上,编码 Kv2.1 电压门控钾通道的 alpha 亚基。Kv2.1 在整个大脑中普遍表达,是控制神经元兴奋性的关键,包括在海马和锥体神经元中。已知人类 KCNB1 基因突变会导致整体发育延迟或停滞、癫痫和行为紊乱。在此,我们报告了一对患有发育迟缓、失神发作、自闭症谱系障碍、肌张力低下和畸形特征的兄弟姐妹。全外显子组测序发现了 KCNB1 中的一个意义不确定的杂合变异(c. 342 C&gt;A),p. (S114R),编码 Kv2.1 N 端胞质区域中丝氨酸到精氨酸的置换 (S114R)。这对兄妹的父亲表现出自闭症特征,并根据家族遗传检测结果被确定为 KCNB1 c. 342 C&gt;A 的强制性携带者。利用细胞电生理学对 Kv2.1-S114R 进行的功能研究发现,通道激活、失活和失活的速度减慢,导致较长时间膜去极化后的净电流增加。据我们所知,这是第一项对患有 KCNB1 紊乱的同胞兄弟姐妹的表现进行比较的同类研究。我们的研究表明,Kv2.1-S114R 会对细胞和表型产生深远的影响。了解 KCNB1 相关疾病的发病机制有助于临床医生进行诊断和治疗,并提供了潜在的治疗途径。
{"title":"A novel autism-associated KCNB1 mutation dramatically slows Kv2.1 potassium channel activation, deactivation and inactivation","authors":"Rían W. Manville, Samantha D. Block, Claire L. Illeck, Jessica Kottmeier, Richard Sidlow, Geoffrey W. Abbott","doi":"10.3389/fncel.2024.1438101","DOIUrl":"https://doi.org/10.3389/fncel.2024.1438101","url":null,"abstract":"<jats:italic>KCNB1</jats:italic>, on human chromosome 20q13.3, encodes the alpha subunit of the Kv2.1 voltage gated potassium channel. Kv2.1 is ubiquitously expressed throughout the brain and is critical in controlling neuronal excitability, including in the hippocampus and pyramidal neurons. Human <jats:italic>KCNB1</jats:italic> mutations are known to cause global development delay or plateauing, epilepsy, and behavioral disorders. Here, we report a sibling pair with developmental delay, absence seizures, autism spectrum disorder, hypotonia, and dysmorphic features. Whole exome sequencing revealed a heterozygous variant of uncertain significance (c. 342 C&amp;gt;A), p. (S114R) in <jats:italic>KCNB1</jats:italic>, encoding a serine to arginine substitution (S114R) in the N-terminal cytoplasmic region of Kv2.1. The siblings’ father demonstrated autistic features and was determined to be an obligate <jats:italic>KCNB1</jats:italic> c. 342 C&amp;gt;A carrier based on familial genetic testing results. Functional investigation of Kv2.1-S114R using cellular electrophysiology revealed slowing of channel activation, deactivation, and inactivation, resulting in increased net current after longer membrane depolarizations. To our knowledge, this is the first study of its kind that compares the presentation of siblings each with a <jats:italic>KCNB1</jats:italic> disorder. Our study demonstrates that Kv2.1-S114R has profound cellular and phenotypic consequences. Understanding the mechanisms underlying <jats:italic>KCNB1</jats:italic>-linked disorders aids clinicians in diagnosis and treatment and provides potential therapeutic avenues to pursue.","PeriodicalId":12432,"journal":{"name":"Frontiers in Cellular Neuroscience","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141869133","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
Neuroinflammation in Parkinson’s disease: focus on the relationship between miRNAs and microglia 帕金森病的神经炎症:关注 miRNA 与小胶质细胞之间的关系
IF 5.3 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-07-26 DOI: 10.3389/fncel.2024.1429977
Ke Xu, Yuan Li, Yan Zhou, Yu Zhang, Yue Shi, Chengguang Zhang, Yan Bai, Shun Wang
Parkinson’s disease (PD) is a prevalent neurodegenerative disorder that affects the central nervous system (CNS). Neuroinflammation is a crucial factor in the pathological advancement of PD. PD is characterized by the presence of activated microglia and increased levels of proinflammatory factors, which play a crucial role in its pathology. During the immune response of PD, microglia regulation is significantly influenced by microRNA (miRNA). The excessive activation of microglia, persistent neuroinflammation, and abnormal polarization of macrophages in the brain can be attributed to the dysregulation of certain miRNAs. Additionally, there are miRNAs that possess the ability to inhibit neuroinflammation. miRNAs, which are small non-coding epigenetic regulators, have the ability to modulate microglial activity in both normal and abnormal conditions. They also have a significant impact on promoting communication between neurons and microglia.
帕金森病(PD)是一种影响中枢神经系统(CNS)的流行性神经退行性疾病。神经炎症是帕金森病病理发展的关键因素。脊髓灰质炎的特点是存在活化的小胶质细胞和促炎因子水平升高,这在其病理过程中起着至关重要的作用。在帕金森病的免疫反应过程中,小胶质细胞的调节受到微小核糖核酸(miRNA)的显著影响。小胶质细胞的过度激活、持续的神经炎症以及大脑中巨噬细胞的异常极化都可归因于某些 miRNA 的失调。此外,还有一些 miRNAs 具有抑制神经炎症的能力。miRNAs 是一种小型非编码表观遗传调节因子,能够在正常和异常情况下调节小胶质细胞的活性。它们对促进神经元和小胶质细胞之间的交流也有重要影响。
{"title":"Neuroinflammation in Parkinson’s disease: focus on the relationship between miRNAs and microglia","authors":"Ke Xu, Yuan Li, Yan Zhou, Yu Zhang, Yue Shi, Chengguang Zhang, Yan Bai, Shun Wang","doi":"10.3389/fncel.2024.1429977","DOIUrl":"https://doi.org/10.3389/fncel.2024.1429977","url":null,"abstract":"Parkinson’s disease (PD) is a prevalent neurodegenerative disorder that affects the central nervous system (CNS). Neuroinflammation is a crucial factor in the pathological advancement of PD. PD is characterized by the presence of activated microglia and increased levels of proinflammatory factors, which play a crucial role in its pathology. During the immune response of PD, microglia regulation is significantly influenced by microRNA (miRNA). The excessive activation of microglia, persistent neuroinflammation, and abnormal polarization of macrophages in the brain can be attributed to the dysregulation of certain miRNAs. Additionally, there are miRNAs that possess the ability to inhibit neuroinflammation. miRNAs, which are small non-coding epigenetic regulators, have the ability to modulate microglial activity in both normal and abnormal conditions. They also have a significant impact on promoting communication between neurons and microglia.","PeriodicalId":12432,"journal":{"name":"Frontiers in Cellular Neuroscience","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141772122","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
A decrease in Fkbp52 alters autophagosome maturation and A152T-tau clearance in vivo Fkbp52的减少会改变体内自噬体的成熟和A152T-tau的清除
IF 5.3 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-07-25 DOI: 10.3389/fncel.2024.1425222
Emilie Lesport, Lucie Commeau, Mélanie Genet, Etienne-Emile Baulieu, Marcel Tawk, Julien Giustiniani
The failure of the autophagy-lysosomal pathway to clear the pathogenic forms of Tau exacerbates the pathogenesis of tauopathies. We have previously shown that the immunophilin FKBP52 interacts both physically and functionally with Tau, and that a decrease in FKBP52 protein levels is associated with Tau deposition in affected human brains. We have also shown that FKBP52 is physiologically present within the lysosomal system in healthy human neurons and that a decrease in FKBP52 expression alters perinuclear lysosomal positioning and Tau clearance during Tau-induced proteotoxic stress in vitro. In this study, we generate a zebrafish fkbp4 loss of function mutant and show that axonal retrograde trafficking of Lamp1 vesicles is altered in this mutant. Moreover, using our transgenic HuC::mCherry-EGFP-LC3 line, we demonstrate that the autophagic flux is impaired in fkbp4 mutant embryos, suggesting a role for Fkbp52 in the maturation of autophagic vesicles. Alterations in both axonal transport and autophagic flux are more evident in heterozygous rather than homozygous fkbp4 mutants. Finally, taking advantage of the previously described A152T-Tau transgenic fish, we show that the clearance of pathogenic A152T-Tau mutant proteins is slower in fkbp4+/− mutants in comparison to fkbp4+/+ larvae. Altogether, these results indicate that Fkbp52 is required for the normal trafficking and maturation of lysosomes and autophagic vacuoles along axons, and that its decrease is sufficient to hinder the clearance of pathogenic Tau in vivo.
自噬-溶酶体途径无法清除致病形式的 Tau,这加剧了 Tau 病的发病机理。我们之前已经证明,免疫嗜蛋白FKBP52与Tau存在物理和功能上的相互作用,而且FKBP52蛋白水平的降低与受影响人脑中Tau的沉积有关。我们还发现,FKBP52生理性地存在于健康人类神经元的溶酶体系统中,FKBP52表达的减少会改变核周溶酶体的定位,并在体外Tau诱导的蛋白毒性应激过程中改变Tau的清除。在本研究中,我们产生了一个斑马鱼 fkbp4 功能缺失突变体,并表明在该突变体中,Lamp1 小泡的轴突逆向贩运发生了改变。此外,我们利用转基因 HuC::mCherry-EGFP-LC3 株系证明,fkbp4 突变体胚胎的自噬通量受损,这表明 Fkbp52 在自噬囊泡的成熟过程中发挥作用。轴突运输和自噬通量的改变在杂合子而非同合子的fkbp4突变体中更为明显。最后,利用之前描述的 A152T-Tau 转基因鱼,我们发现与 fkbp4+/+ 幼体相比,fkbp4+/- 突变体中致病性 A152T-Tau 突变蛋白的清除速度较慢。总之,这些结果表明,Fkbp52是轴突溶酶体和自噬泡正常运输和成熟所必需的,它的减少足以阻碍体内致病性Tau的清除。
{"title":"A decrease in Fkbp52 alters autophagosome maturation and A152T-tau clearance in vivo","authors":"Emilie Lesport, Lucie Commeau, Mélanie Genet, Etienne-Emile Baulieu, Marcel Tawk, Julien Giustiniani","doi":"10.3389/fncel.2024.1425222","DOIUrl":"https://doi.org/10.3389/fncel.2024.1425222","url":null,"abstract":"The failure of the autophagy-lysosomal pathway to clear the pathogenic forms of Tau exacerbates the pathogenesis of tauopathies. We have previously shown that the immunophilin FKBP52 interacts both physically and functionally with Tau, and that a decrease in FKBP52 protein levels is associated with Tau deposition in affected human brains. We have also shown that FKBP52 is physiologically present within the lysosomal system in healthy human neurons and that a decrease in FKBP52 expression alters perinuclear lysosomal positioning and Tau clearance during Tau-induced proteotoxic stress <jats:italic>in vitro</jats:italic>. In this study, we generate a zebrafish <jats:italic>fkbp4</jats:italic> loss of function mutant and show that axonal retrograde trafficking of Lamp1 vesicles is altered in this mutant. Moreover, using our transgenic <jats:italic>HuC::mCherry-EGFP-LC3</jats:italic> line, we demonstrate that the autophagic flux is impaired in <jats:italic>fkbp4</jats:italic> mutant embryos, suggesting a role for Fkbp52 in the maturation of autophagic vesicles. Alterations in both axonal transport and autophagic flux are more evident in heterozygous rather than homozygous <jats:italic>fkbp4</jats:italic> mutants. Finally, taking advantage of the previously described A152T-Tau transgenic fish, we show that the clearance of pathogenic A152T-Tau mutant proteins is slower in <jats:italic>fkbp4</jats:italic><jats:sup>+/−</jats:sup> mutants in comparison to <jats:italic>fkbp4</jats:italic><jats:sup>+/+</jats:sup> larvae. Altogether, these results indicate that Fkbp52 is required for the normal trafficking and maturation of lysosomes and autophagic vacuoles along axons, and that its decrease is sufficient to hinder the clearance of pathogenic Tau <jats:italic>in vivo</jats:italic>.","PeriodicalId":12432,"journal":{"name":"Frontiers in Cellular Neuroscience","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141772123","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
Modeling riboflavin transporter deficiency type 2: from iPSC-derived motoneurons to iPSC-derived astrocytes 核黄素转运体缺乏症 2 型建模:从 iPSC 衍生的运动神经元到 iPSC 衍生的星形胶质细胞
IF 5.3 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-07-24 DOI: 10.3389/fncel.2024.1440555
Valentina Magliocca, Angela Lanciotti, Elena Ambrosini, Lorena Travaglini, Veronica D’Ezio, Valentina D’Oria, Stefania Petrini, Michela Catteruccia, Keith Massey, Marco Tartaglia, Enrico Bertini, Tiziana Persichini, Claudia Compagnucci
IntroductionRiboflavin transporter deficiency type 2 (RTD2) is a rare neurodegenerative autosomal recessive disease caused by mutations in the SLC52A2 gene encoding the riboflavin transporters, RFVT2. Riboflavin (Rf) is the precursor of FAD (flavin adenine dinucleotide) and FMN (flavin mononucleotide), which are involved in different redox reactions, including the energetic metabolism processes occurring in mitochondria. To date, human induced pluripotent stem cells (iPSCs) have given the opportunity to characterize RTD2 motoneurons, which reflect the most affected cell type. Previous works have demonstrated mitochondrial and peroxisomal altered energy metabolism as well as cytoskeletal derangement in RTD2 iPSCs and iPSC-derived motoneurons. So far, no attention has been dedicated to astrocytes.Results and discussionHere, we demonstrate that in vitro differentiation of astrocytes, which guarantee trophic and metabolic support to neurons, from RTD2 iPSCs is not compromised. These cells do not exhibit evident morphological differences nor significant changes in the survival rate when compared to astrocytes derived from iPSCs of healthy individuals. These findings indicate that differently from what had previously been documented for neurons, RTD2 does not compromise the morpho-functional features of astrocytes.
核黄素转运体缺乏症 2 型(RTD2)是一种罕见的神经退行性常染色体隐性遗传病,由编码核黄素转运体 RFVT2 的 SLC52A2 基因突变引起。核黄素(Rf)是黄素腺嘌呤二核苷酸(FAD)和黄素单核苷酸(FMN)的前体,它们参与不同的氧化还原反应,包括线粒体中发生的能量代谢过程。迄今为止,人类诱导多能干细胞(iPSCs)为研究 RTD2 运动神经元的特征提供了机会,而 RTD2 运动神经元是受影响最严重的细胞类型。之前的研究表明,线粒体和过氧化物酶体改变了能量代谢,RTD2 iPSC 和 iPSC 衍生的运动神经元的细胞骨架也发生了变化。结果与讨论在这里,我们证明了从 RTD2 iPSCs 体外分化出的星形胶质细胞并没有受到损害,这些细胞为神经元提供营养和代谢支持。与从健康人的 iPSCs 中提取的星形胶质细胞相比,这些细胞没有表现出明显的形态差异,存活率也没有显著变化。这些研究结果表明,与之前记录的神经元不同,RTD2 不会损害星形胶质细胞的形态功能特征。
{"title":"Modeling riboflavin transporter deficiency type 2: from iPSC-derived motoneurons to iPSC-derived astrocytes","authors":"Valentina Magliocca, Angela Lanciotti, Elena Ambrosini, Lorena Travaglini, Veronica D’Ezio, Valentina D’Oria, Stefania Petrini, Michela Catteruccia, Keith Massey, Marco Tartaglia, Enrico Bertini, Tiziana Persichini, Claudia Compagnucci","doi":"10.3389/fncel.2024.1440555","DOIUrl":"https://doi.org/10.3389/fncel.2024.1440555","url":null,"abstract":"IntroductionRiboflavin transporter deficiency type 2 (RTD2) is a rare neurodegenerative autosomal recessive disease caused by mutations in the SLC52A2 gene encoding the riboflavin transporters, RFVT2. Riboflavin (Rf) is the precursor of FAD (flavin adenine dinucleotide) and FMN (flavin mononucleotide), which are involved in different redox reactions, including the energetic metabolism processes occurring in mitochondria. To date, human induced pluripotent stem cells (iPSCs) have given the opportunity to characterize RTD2 motoneurons, which reflect the most affected cell type. Previous works have demonstrated mitochondrial and peroxisomal altered energy metabolism as well as cytoskeletal derangement in RTD2 iPSCs and iPSC-derived motoneurons. So far, no attention has been dedicated to astrocytes.Results and discussionHere, we demonstrate that <jats:italic>in vitro</jats:italic> differentiation of astrocytes, which guarantee trophic and metabolic support to neurons, from RTD2 iPSCs is not compromised. These cells do not exhibit evident morphological differences nor significant changes in the survival rate when compared to astrocytes derived from iPSCs of healthy individuals. These findings indicate that differently from what had previously been documented for neurons, RTD2 does not compromise the morpho-functional features of astrocytes.","PeriodicalId":12432,"journal":{"name":"Frontiers in Cellular Neuroscience","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141772233","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
Genetic approaches to elucidating cortical and hippocampal GABAergic interneuron diversity 阐明大脑皮层和海马 GABA 能中间神经元多样性的遗传方法
IF 5.3 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-07-24 DOI: 10.3389/fncel.2024.1414955
Robert Machold, Bernardo Rudy
GABAergic interneurons (INs) in the mammalian forebrain represent a diverse population of cells that provide specialized forms of local inhibition to regulate neural circuit activity. Over the last few decades, the development of a palette of genetic tools along with the generation of single-cell transcriptomic data has begun to reveal the molecular basis of IN diversity, thereby providing deep insights into how different IN subtypes function in the forebrain. In this review, we outline the emerging picture of cortical and hippocampal IN speciation as defined by transcriptomics and developmental origin and summarize the genetic strategies that have been utilized to target specific IN subtypes, along with the technical considerations inherent to each approach. Collectively, these methods have greatly facilitated our understanding of how IN subtypes regulate forebrain circuitry via cell type and compartment-specific inhibition and thus have illuminated a path toward potential therapeutic interventions for a variety of neurocognitive disorders.
哺乳动物前脑中的 GABA 能中间神经元(IN)代表了一个多样化的细胞群体,它们提供专门的局部抑制形式来调节神经回路的活动。在过去的几十年里,随着单细胞转录组数据的产生,一系列遗传学工具的开发开始揭示 IN 多样性的分子基础,从而为深入了解不同 IN 亚型如何在前脑中发挥作用提供了依据。在这篇综述中,我们概述了由转录组学和发育起源所定义的大脑皮层和海马 IN 种群的新情况,并总结了针对特定 IN 亚型所采用的遗传策略,以及每种方法固有的技术注意事项。总之,这些方法极大地促进了我们对 IN 亚型如何通过细胞类型和区室特异性抑制调节前脑回路的理解,从而为各种神经认知障碍的潜在治疗干预指明了道路。
{"title":"Genetic approaches to elucidating cortical and hippocampal GABAergic interneuron diversity","authors":"Robert Machold, Bernardo Rudy","doi":"10.3389/fncel.2024.1414955","DOIUrl":"https://doi.org/10.3389/fncel.2024.1414955","url":null,"abstract":"GABAergic interneurons (INs) in the mammalian forebrain represent a diverse population of cells that provide specialized forms of local inhibition to regulate neural circuit activity. Over the last few decades, the development of a palette of genetic tools along with the generation of single-cell transcriptomic data has begun to reveal the molecular basis of IN diversity, thereby providing deep insights into how different IN subtypes function in the forebrain. In this review, we outline the emerging picture of cortical and hippocampal IN speciation as defined by transcriptomics and developmental origin and summarize the genetic strategies that have been utilized to target specific IN subtypes, along with the technical considerations inherent to each approach. Collectively, these methods have greatly facilitated our understanding of how IN subtypes regulate forebrain circuitry via cell type and compartment-specific inhibition and thus have illuminated a path toward potential therapeutic interventions for a variety of neurocognitive disorders.","PeriodicalId":12432,"journal":{"name":"Frontiers in Cellular Neuroscience","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141772124","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
Oligodendrocyte precursor cell-derived exosomes combined with cell therapy promote clinical recovery by immunomodulation and gliosis attenuation 少突胶质前体细胞衍生的外泌体与细胞疗法相结合,通过免疫调节和减轻胶质病变促进临床康复
IF 5.3 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-07-23 DOI: 10.3389/fncel.2024.1413843
Sarah Ingrid Pinto Santos, Santiago José Ortiz-Peñuela, Alessandro de Paula Filho, Ana Laura Midori Rossi Tomiyama, Lilian de Oliveira Coser, Juliano Coelho da Silveira, Daniele dos Santos Martins, Adriano Polican Ciena, Alexandre Leite Rodrigues de Oliveira, Carlos Eduardo Ambrósio
Multiple sclerosis is a chronic inflammatory disease of the central nervous system characterized by autoimmune destruction of the myelin sheath, leading to irreversible and progressive functional deficits in patients. Pre-clinical studies involving the use of neural stem cells (NSCs) have already demonstrated their potential in neuronal regeneration and remyelination. However, the exclusive application of cell therapy has not proved sufficient to achieve satisfactory therapeutic levels. Recognizing these limitations, there is a need to combine cell therapy with other adjuvant protocols. In this context, extracellular vesicles (EVs) can contribute to intercellular communication, stimulating the production of proteins and lipids associated with remyelination and providing trophic support to axons. This study aimed to evaluate the therapeutic efficacy of the combination of NSCs and EVs derived from oligodendrocyte precursor cells (OPCs) in an animal model of multiple sclerosis. OPCs were differentiated from NSCs and had their identity confirmed by gene expression analysis and immunocytochemistry. Exosomes were isolated by differential ultracentrifugation and characterized by Western, transmission electron microscopy and nanoparticle tracking analysis. Experimental therapy of C57BL/6 mice induced with experimental autoimmune encephalomyelitis (EAE) were grouped in control, treated with NSCs, treated with OPC-derived EVs and treated with a combination of both. The treatments were evaluated clinically using scores and body weight, microscopically using immunohistochemistry and immunological profile by flow cytometry. The animals showed significant clinical improvement and weight gain with the treatments. However, only the treatments involving EVs led to immune modulation, changing the profile from Th1 to Th2 lymphocytes. Fifteen days after treatment revealed a reduction in reactive microgliosis and astrogliosis in the groups treated with EVs. However, there was no reduction in demyelination. The results indicate the potential therapeutic use of OPC-derived EVs to attenuate inflammation and promote recovery in EAE, especially when combined with cell therapy.
多发性硬化症是中枢神经系统的一种慢性炎症性疾病,其特点是髓鞘受到自身免疫性破坏,导致患者出现不可逆转的进行性功能障碍。使用神经干细胞(NSCs)进行的临床前研究已经证明了它们在神经元再生和髓鞘再形成方面的潜力。然而,仅应用细胞疗法还不足以达到令人满意的治疗水平。认识到这些局限性,有必要将细胞疗法与其他辅助方案相结合。在这种情况下,细胞外囊泡(EVs)可以促进细胞间的交流,刺激与再髓鞘化相关的蛋白质和脂质的产生,并为轴突提供营养支持。本研究旨在评估在多发性硬化症动物模型中,将少突胶质前体细胞(OPCs)提取的NSCs和EVs结合使用的疗效。OPCs 从 NSCs 分化而来,并通过基因表达分析和免疫细胞化学确认了其身份。通过差速超速离心法分离出外泌体,并通过Western、透射电子显微镜和纳米颗粒追踪分析对其进行鉴定。对诱发实验性自身免疫性脑脊髓炎(EAE)的 C57BL/6 小鼠的实验治疗分为对照组、NSCs 治疗组、OPC 衍生 EVs 治疗组和两者结合治疗组。临床上使用评分和体重对治疗进行评估,显微镜下使用免疫组化,流式细胞术对免疫学特征进行评估。动物的临床症状明显改善,体重增加。不过,只有使用 EVs 的治疗才会导致免疫调节,使 Th1 淋巴细胞变为 Th2 淋巴细胞。治疗十五天后发现,在使用EVs治疗的组中,反应性小胶质细胞和星形胶质细胞减少。不过,脱髓鞘现象并未减少。这些结果表明,OPC衍生的EVs具有潜在的治疗作用,可减轻炎症反应并促进EAE的恢复,尤其是在与细胞疗法相结合时。
{"title":"Oligodendrocyte precursor cell-derived exosomes combined with cell therapy promote clinical recovery by immunomodulation and gliosis attenuation","authors":"Sarah Ingrid Pinto Santos, Santiago José Ortiz-Peñuela, Alessandro de Paula Filho, Ana Laura Midori Rossi Tomiyama, Lilian de Oliveira Coser, Juliano Coelho da Silveira, Daniele dos Santos Martins, Adriano Polican Ciena, Alexandre Leite Rodrigues de Oliveira, Carlos Eduardo Ambrósio","doi":"10.3389/fncel.2024.1413843","DOIUrl":"https://doi.org/10.3389/fncel.2024.1413843","url":null,"abstract":"Multiple sclerosis is a chronic inflammatory disease of the central nervous system characterized by autoimmune destruction of the myelin sheath, leading to irreversible and progressive functional deficits in patients. Pre-clinical studies involving the use of neural stem cells (NSCs) have already demonstrated their potential in neuronal regeneration and remyelination. However, the exclusive application of cell therapy has not proved sufficient to achieve satisfactory therapeutic levels. Recognizing these limitations, there is a need to combine cell therapy with other adjuvant protocols. In this context, extracellular vesicles (EVs) can contribute to intercellular communication, stimulating the production of proteins and lipids associated with remyelination and providing trophic support to axons. This study aimed to evaluate the therapeutic efficacy of the combination of NSCs and EVs derived from oligodendrocyte precursor cells (OPCs) in an animal model of multiple sclerosis. OPCs were differentiated from NSCs and had their identity confirmed by gene expression analysis and immunocytochemistry. Exosomes were isolated by differential ultracentrifugation and characterized by Western, transmission electron microscopy and nanoparticle tracking analysis. Experimental therapy of C57BL/6 mice induced with experimental autoimmune encephalomyelitis (EAE) were grouped in control, treated with NSCs, treated with OPC-derived EVs and treated with a combination of both. The treatments were evaluated clinically using scores and body weight, microscopically using immunohistochemistry and immunological profile by flow cytometry. The animals showed significant clinical improvement and weight gain with the treatments. However, only the treatments involving EVs led to immune modulation, changing the profile from Th1 to Th2 lymphocytes. Fifteen days after treatment revealed a reduction in reactive microgliosis and astrogliosis in the groups treated with EVs. However, there was no reduction in demyelination. The results indicate the potential therapeutic use of OPC-derived EVs to attenuate inflammation and promote recovery in EAE, especially when combined with cell therapy.","PeriodicalId":12432,"journal":{"name":"Frontiers in Cellular Neuroscience","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141772126","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
A novel histone deacetylase inhibitor W2A-16 improves the barrier integrity in brain vascular endothelial cells 新型组蛋白去乙酰化酶抑制剂 W2A-16 可改善脑血管内皮细胞屏障的完整性
IF 5.3 3区 医学 Q2 NEUROSCIENCES Pub Date : 2024-07-19 DOI: 10.3389/fncel.2024.1368018
Yasuteru Inoue, Yingxue Ren, Shuwen Zhang, Michael Bamkole, Naeyma N. Islam, Manikandan Selvaraj, Wenyan Lu, Thomas R. Caulfield, Yonghe Li, Takahisa Kanekiyo
The maturation of brain microvascular endothelial cells leads to the formation of a tightly sealed monolayer, known as the blood–brain barrier (BBB). The BBB damage is associated with the pathogenesis of age-related neurodegenerative diseases including vascular cognitive impairment and Alzheimer’s disease. Growing knowledge in the field of epigenetics can enhance the understanding of molecular profile of the BBB and has great potential for the development of novel therapeutic strategies or targets to repair a disrupted BBB. Histone deacetylases (HDACs) inhibitors are epigenetic regulators that can induce acetylation of histones and induce open chromatin conformation, promoting gene expression by enhancing the binding of DNA with transcription factors. We investigated how HDAC inhibition influences the barrier integrity using immortalized human endothelial cells (HCMEC/D3) and the human induced pluripotent stem cell (iPSC)-derived brain vascular endothelial cells. The endothelial cells were treated with or without a novel compound named W2A-16. W2A-16 not only activates Wnt/β-catenin signaling but also functions as a class I HDAC inhibitor. We demonstrated that the administration with W2A-16 sustained barrier properties of the monolayer of endothelial cells, as evidenced by increased trans-endothelial electrical resistance (TEER). The BBB-related genes and protein expression were also increased compared with non-treated controls. Analysis of transcript profiles through RNA-sequencing in hCMEC/D3 cells indicated that W2A-16 potentially enhances BBB integrity by influencing genes associated with the regulation of the extracellular microenvironment. These findings collectively propose that the HDAC inhibition by W2A-16 plays a facilitating role in the formation of the BBB. Pharmacological approaches to inhibit HDAC may be a potential therapeutic strategy to boost and/or restore BBB integrity.
脑微血管内皮细胞成熟后会形成一个紧密密封的单层,即血脑屏障(BBB)。血脑屏障损伤与血管性认知障碍和阿尔茨海默病等与年龄相关的神经退行性疾病的发病机制有关。表观遗传学领域不断增长的知识可以加深人们对 BBB 分子特征的了解,并具有开发新型治疗策略或靶点以修复被破坏的 BBB 的巨大潜力。组蛋白去乙酰化酶(HDACs)抑制剂是一种表观遗传调节剂,可诱导组蛋白乙酰化并诱导染色质开放构象,通过增强 DNA 与转录因子的结合促进基因表达。我们使用永生化人内皮细胞(HCMEC/D3)和诱导多能干细胞(iPSC)衍生的脑血管内皮细胞研究了 HDAC 抑制如何影响屏障完整性。使用或不使用一种名为 W2A-16 的新型化合物处理内皮细胞。W2A-16 不仅能激活 Wnt/β-catenin 信号转导,而且还是一种 I 类 HDAC 抑制剂。我们证实,服用 W2A-16 能维持内皮细胞单层的屏障特性,表现为跨内皮电阻(TEER)的增加。与未经处理的对照组相比,与 BBB 相关的基因和蛋白质表达也有所增加。通过对 hCMEC/D3 细胞中的 RNA 序列进行转录谱分析表明,W2A-16 有可能通过影响与细胞外微环境调控相关的基因来增强 BBB 的完整性。这些发现共同表明,W2A-16 对 HDAC 的抑制在 BBB 的形成过程中起到了促进作用。抑制 HDAC 的药理方法可能是促进和/或恢复 BBB 完整性的潜在治疗策略。
{"title":"A novel histone deacetylase inhibitor W2A-16 improves the barrier integrity in brain vascular endothelial cells","authors":"Yasuteru Inoue, Yingxue Ren, Shuwen Zhang, Michael Bamkole, Naeyma N. Islam, Manikandan Selvaraj, Wenyan Lu, Thomas R. Caulfield, Yonghe Li, Takahisa Kanekiyo","doi":"10.3389/fncel.2024.1368018","DOIUrl":"https://doi.org/10.3389/fncel.2024.1368018","url":null,"abstract":"The maturation of brain microvascular endothelial cells leads to the formation of a tightly sealed monolayer, known as the blood–brain barrier (BBB). The BBB damage is associated with the pathogenesis of age-related neurodegenerative diseases including vascular cognitive impairment and Alzheimer’s disease. Growing knowledge in the field of epigenetics can enhance the understanding of molecular profile of the BBB and has great potential for the development of novel therapeutic strategies or targets to repair a disrupted BBB. Histone deacetylases (HDACs) inhibitors are epigenetic regulators that can induce acetylation of histones and induce open chromatin conformation, promoting gene expression by enhancing the binding of DNA with transcription factors. We investigated how HDAC inhibition influences the barrier integrity using immortalized human endothelial cells (HCMEC/D3) and the human induced pluripotent stem cell (iPSC)-derived brain vascular endothelial cells. The endothelial cells were treated with or without a novel compound named W2A-16. W2A-16 not only activates Wnt/β-catenin signaling but also functions as a class I HDAC inhibitor. We demonstrated that the administration with W2A-16 sustained barrier properties of the monolayer of endothelial cells, as evidenced by increased trans-endothelial electrical resistance (TEER). The BBB-related genes and protein expression were also increased compared with non-treated controls. Analysis of transcript profiles through RNA-sequencing in hCMEC/D3 cells indicated that W2A-16 potentially enhances BBB integrity by influencing genes associated with the regulation of the extracellular microenvironment. These findings collectively propose that the HDAC inhibition by W2A-16 plays a facilitating role in the formation of the BBB. Pharmacological approaches to inhibit HDAC may be a potential therapeutic strategy to boost and/or restore BBB integrity.","PeriodicalId":12432,"journal":{"name":"Frontiers in Cellular Neuroscience","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141744313","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 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