首页 > 最新文献

Translational Neurodegeneration最新文献

英文 中文
Neuronal and synaptic adaptations underlying the benefits of deep brain stimulation for Parkinson's disease. 深层脑刺激治疗帕金森病的好处背后的神经元和突触适应。
IF 12.6 1区 医学 Q1 NEUROSCIENCES Pub Date : 2023-11-30 DOI: 10.1186/s40035-023-00390-w
Wenying Xu, Jie Wang, Xin-Ni Li, Jingxue Liang, Lu Song, Yi Wu, Zhenguo Liu, Bomin Sun, Wei-Guang Li

Deep brain stimulation (DBS) is a well-established and effective treatment for patients with advanced Parkinson's disease (PD), yet its underlying mechanisms remain enigmatic. Optogenetics, primarily conducted in animal models, provides a unique approach that allows cell type- and projection-specific modulation that mirrors the frequency-dependent stimulus effects of DBS. Opto-DBS research in animal models plays a pivotal role in unraveling the neuronal and synaptic adaptations that contribute to the efficacy of DBS in PD treatment. DBS-induced neuronal responses rely on a complex interplay between the distributions of presynaptic inputs, frequency-dependent synaptic depression, and the intrinsic excitability of postsynaptic neurons. This orchestration leads to conversion of firing patterns, enabling both antidromic and orthodromic modulation of neural circuits. Understanding these mechanisms is vital for decoding position- and programming-dependent effects of DBS. Furthermore, patterned stimulation is emerging as a promising strategy yielding long-lasting therapeutic benefits. Research on the neuronal and synaptic adaptations to DBS may pave the way for the development of more enduring and precise modulation patterns. Advanced technologies, such as adaptive DBS or directional electrodes, can also be integrated for circuit-specific neuromodulation. These insights hold the potential to greatly improve the effectiveness of DBS and advance PD treatment to new levels.

脑深部电刺激(DBS)是治疗晚期帕金森病(PD)的一种行之有效的治疗方法,但其潜在机制尚不清楚。光遗传学主要在动物模型中进行,提供了一种独特的方法,允许细胞类型和投影特异性调制,反映DBS的频率依赖性刺激效应。动物模型的Opto-DBS研究在揭示DBS治疗PD疗效的神经元和突触适应方面起着关键作用。dbs诱导的神经元反应依赖于突触前输入分布、频率依赖性突触抑制和突触后神经元固有兴奋性之间的复杂相互作用。这种协调导致放电模式的转换,使神经回路的反正交和正正交调制成为可能。了解这些机制对于解码DBS的位置和编程依赖效应至关重要。此外,模式刺激正在成为一种有前景的策略,产生持久的治疗效果。研究神经元和突触对DBS的适应可能为开发更持久和精确的调制模式铺平道路。先进的技术,如自适应脑起搏器或定向电极,也可以用于特定电路的神经调节。这些见解有可能大大提高DBS的有效性,并将PD治疗推向新的水平。
{"title":"Neuronal and synaptic adaptations underlying the benefits of deep brain stimulation for Parkinson's disease.","authors":"Wenying Xu, Jie Wang, Xin-Ni Li, Jingxue Liang, Lu Song, Yi Wu, Zhenguo Liu, Bomin Sun, Wei-Guang Li","doi":"10.1186/s40035-023-00390-w","DOIUrl":"10.1186/s40035-023-00390-w","url":null,"abstract":"<p><p>Deep brain stimulation (DBS) is a well-established and effective treatment for patients with advanced Parkinson's disease (PD), yet its underlying mechanisms remain enigmatic. Optogenetics, primarily conducted in animal models, provides a unique approach that allows cell type- and projection-specific modulation that mirrors the frequency-dependent stimulus effects of DBS. Opto-DBS research in animal models plays a pivotal role in unraveling the neuronal and synaptic adaptations that contribute to the efficacy of DBS in PD treatment. DBS-induced neuronal responses rely on a complex interplay between the distributions of presynaptic inputs, frequency-dependent synaptic depression, and the intrinsic excitability of postsynaptic neurons. This orchestration leads to conversion of firing patterns, enabling both antidromic and orthodromic modulation of neural circuits. Understanding these mechanisms is vital for decoding position- and programming-dependent effects of DBS. Furthermore, patterned stimulation is emerging as a promising strategy yielding long-lasting therapeutic benefits. Research on the neuronal and synaptic adaptations to DBS may pave the way for the development of more enduring and precise modulation patterns. Advanced technologies, such as adaptive DBS or directional electrodes, can also be integrated for circuit-specific neuromodulation. These insights hold the potential to greatly improve the effectiveness of DBS and advance PD treatment to new levels.</p>","PeriodicalId":23269,"journal":{"name":"Translational Neurodegeneration","volume":"12 1","pages":"55"},"PeriodicalIF":12.6,"publicationDate":"2023-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10688037/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138462862","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
N6-methyladenosine-modified circRIMS2 mediates synaptic and memory impairments by activating GluN2B ubiquitination in Alzheimer's disease. n6 -甲基腺苷修饰的circRIMS2通过激活GluN2B泛素化介导阿尔茨海默病的突触和记忆损伤。
IF 12.6 1区 医学 Q1 NEUROSCIENCES Pub Date : 2023-11-28 DOI: 10.1186/s40035-023-00386-6
Xiong Wang, Jiazhao Xie, Lu Tan, Yanjun Lu, Na Shen, Jiaoyuan Li, Hui Hu, Huijun Li, Xiaoguang Li, Liming Cheng

Background: Synaptic degeneration occurs in the early stage of Alzheimer's disease (AD) before devastating symptoms, strongly correlated with cognitive decline. Circular RNAs (circRNAs) are abundantly enriched in neural tissues, and aberrant expression of circRNAs precedes AD symptoms, significantly correlated with clinical dementia severity. However, the direct relationship between circRNA dysregulation and synaptic impairment in the early stage of AD remains poorly understood.

Methods: Hippocampal whole-transcriptome sequencing was performed to identify dysregulated circRNAs and miRNAs in 4-month-old wild-type and APP/PS1 mice. RNA antisense purification and mass spectrometry were utilized to unveil interactions between circRIMS2 and methyltransferase 3, N6-adenosine-methyltransferase complex catalytic subunit (METTL3). The roles of circRIMS2/miR-3968 in synaptic targeting of UBE2K-mediated ubiquitination of GluN2B subunit of NMDA receptor were evaluated via numerous lentiviruses followed by morphological staining, co-immunoprecipitation and behavioral testing. Further, a membrane-permeable peptide was used to block the ubiquitination of K1082 on GluN2B in AD mice.

Results: circRIMS2 was significantly upregulated in 4-month-old APP/PS1 mice, which was mediated by METTL3-dependent N6-methyladenosine (m6A) modification. Overexpression of circRIMS2 led to synaptic and memory impairments in 4-month-old C57BL/6 mice. MiR-3968/UBE2K was validated as the downstream of circRIMS2. Elevated UBE2K induced synaptic dysfunction of AD through ubiquitinating K1082 on GluN2B. Silencing METTL3 or blocking the ubiquitination of K1082 on GluN2B with a short membrane-permeable peptide remarkably rescued synaptic dysfunction in AD mice.

Conclusions: In conclusion, our study demonstrated that m6A-modified circRIMS2 mediates the synaptic and memory impairments in AD by activating the UBE2K-dependent ubiquitination and degradation of GluN2B via sponging miR-3968, providing novel therapeutic strategies for AD.

背景:突触变性发生在阿尔茨海默病(AD)的早期,在破坏性症状出现之前,与认知能力下降密切相关。环状rna (circRNAs)在神经组织中大量富集,circRNAs的异常表达先于AD症状,与临床痴呆严重程度显著相关。然而,circRNA失调与AD早期突触损伤之间的直接关系尚不清楚。方法:对4月龄野生型和APP/PS1小鼠进行海马全转录组测序,鉴定异常的环状rna和mirna。利用RNA反义纯化和质谱分析揭示了circRIMS2与甲基转移酶3、n6 -腺苷-甲基转移酶复合物催化亚基(METTL3)之间的相互作用。circRIMS2/miR-3968在突触靶向ube2k介导的NMDA受体GluN2B亚基泛素化中的作用通过多种慢病毒,形态学染色,共免疫沉淀和行为测试来评估。此外,一种膜透性肽被用于阻断AD小鼠GluN2B上K1082的泛素化。结果:circRIMS2在4月龄APP/PS1小鼠中显著上调,这是由mettl3依赖性n6 -甲基腺苷(m6A)修饰介导的。circRIMS2过表达导致4月龄C57BL/6小鼠突触和记忆损伤。证实MiR-3968/UBE2K是circRIMS2的下游。升高的UBE2K通过泛素化GluN2B上的K1082诱导AD的突触功能障碍。沉默METTL3或用短膜可渗透肽阻断GluN2B上K1082的泛素化,可显著拯救AD小鼠的突触功能障碍。结论:总之,我们的研究表明,m6a修饰的circRIMS2通过海绵miR-3968激活ube2k依赖的泛素化和GluN2B的降解,介导AD的突触和记忆损伤,为AD的治疗提供了新的策略。
{"title":"N6-methyladenosine-modified circRIMS2 mediates synaptic and memory impairments by activating GluN2B ubiquitination in Alzheimer's disease.","authors":"Xiong Wang, Jiazhao Xie, Lu Tan, Yanjun Lu, Na Shen, Jiaoyuan Li, Hui Hu, Huijun Li, Xiaoguang Li, Liming Cheng","doi":"10.1186/s40035-023-00386-6","DOIUrl":"10.1186/s40035-023-00386-6","url":null,"abstract":"<p><strong>Background: </strong>Synaptic degeneration occurs in the early stage of Alzheimer's disease (AD) before devastating symptoms, strongly correlated with cognitive decline. Circular RNAs (circRNAs) are abundantly enriched in neural tissues, and aberrant expression of circRNAs precedes AD symptoms, significantly correlated with clinical dementia severity. However, the direct relationship between circRNA dysregulation and synaptic impairment in the early stage of AD remains poorly understood.</p><p><strong>Methods: </strong>Hippocampal whole-transcriptome sequencing was performed to identify dysregulated circRNAs and miRNAs in 4-month-old wild-type and APP/PS1 mice. RNA antisense purification and mass spectrometry were utilized to unveil interactions between circRIMS2 and methyltransferase 3, N6-adenosine-methyltransferase complex catalytic subunit (METTL3). The roles of circRIMS2/miR-3968 in synaptic targeting of UBE2K-mediated ubiquitination of GluN2B subunit of NMDA receptor were evaluated via numerous lentiviruses followed by morphological staining, co-immunoprecipitation and behavioral testing. Further, a membrane-permeable peptide was used to block the ubiquitination of K1082 on GluN2B in AD mice.</p><p><strong>Results: </strong>circRIMS2 was significantly upregulated in 4-month-old APP/PS1 mice, which was mediated by METTL3-dependent N6-methyladenosine (m6A) modification. Overexpression of circRIMS2 led to synaptic and memory impairments in 4-month-old C57BL/6 mice. MiR-3968/UBE2K was validated as the downstream of circRIMS2. Elevated UBE2K induced synaptic dysfunction of AD through ubiquitinating K1082 on GluN2B. Silencing METTL3 or blocking the ubiquitination of K1082 on GluN2B with a short membrane-permeable peptide remarkably rescued synaptic dysfunction in AD mice.</p><p><strong>Conclusions: </strong>In conclusion, our study demonstrated that m6A-modified circRIMS2 mediates the synaptic and memory impairments in AD by activating the UBE2K-dependent ubiquitination and degradation of GluN2B via sponging miR-3968, providing novel therapeutic strategies for AD.</p>","PeriodicalId":23269,"journal":{"name":"Translational Neurodegeneration","volume":"12 1","pages":"53"},"PeriodicalIF":12.6,"publicationDate":"2023-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10683276/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138446299","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
A phase 1 open-label pilot study of low-dose interleukine-2 immunotherapy in patients with Alzheimer's disease. 低剂量白介素-2免疫治疗阿尔茨海默病患者的1期开放标签试点研究
IF 12.6 1区 医学 Q1 NEUROSCIENCES Pub Date : 2023-11-16 DOI: 10.1186/s40035-023-00387-5
Alireza Faridar, Abdulmunaim M Eid, Aaron D Thome, Weihua Zhao, David R Beers, Maria B Pascual, Mohammad O Nakawah, Gustavo C Roman, Charles S Davis, Michael Grundman, Joseph C Masdeu, Stanley H Appel

Trial registration: ClinicalTrials.gov Identifier: NCT05821153, Registered April 20 2023, Retrospectively registered, https://classic.

Clinicaltrials: gov/ct2/show/NCT05821153.

试验注册:ClinicalTrials.gov标识符:NCT05821153,注册于2023年4月20日,回顾性注册,https://classic.Clinicaltrials: gov/ct2/show/NCT05821153。
{"title":"A phase 1 open-label pilot study of low-dose interleukine-2 immunotherapy in patients with Alzheimer's disease.","authors":"Alireza Faridar, Abdulmunaim M Eid, Aaron D Thome, Weihua Zhao, David R Beers, Maria B Pascual, Mohammad O Nakawah, Gustavo C Roman, Charles S Davis, Michael Grundman, Joseph C Masdeu, Stanley H Appel","doi":"10.1186/s40035-023-00387-5","DOIUrl":"10.1186/s40035-023-00387-5","url":null,"abstract":"<p><strong>Trial registration: </strong>ClinicalTrials.gov Identifier: NCT05821153, Registered April 20 2023, Retrospectively registered, https://classic.</p><p><strong>Clinicaltrials: </strong>gov/ct2/show/NCT05821153.</p>","PeriodicalId":23269,"journal":{"name":"Translational Neurodegeneration","volume":"12 1","pages":"54"},"PeriodicalIF":12.6,"publicationDate":"2023-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10652426/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"134649851","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Oligodendrocyte progenitor cells in Alzheimer's disease: from physiology to pathology. 阿尔茨海默病少突胶质细胞祖细胞:从生理学到病理学。
IF 12.6 1区 医学 Q1 NEUROSCIENCES Pub Date : 2023-11-14 DOI: 10.1186/s40035-023-00385-7
Peibin Zou, Chongyun Wu, Timon Cheng-Yi Liu, Rui Duan, Luodan Yang

Oligodendrocyte progenitor cells (OPCs) play pivotal roles in myelin formation and phagocytosis, communicating with neighboring cells and contributing to the integrity of the blood-brain barrier (BBB). However, under the pathological circumstances of Alzheimer's disease (AD), the brain's microenvironment undergoes detrimental changes that significantly impact OPCs and their functions. Starting with OPC functions, we delve into the transformation of OPCs to myelin-producing oligodendrocytes, the intricate signaling interactions with other cells in the central nervous system (CNS), and the fascinating process of phagocytosis, which influences the function of OPCs and affects CNS homeostasis. Moreover, we discuss the essential role of OPCs in BBB formation and highlight the critical contribution of OPCs in forming CNS-protective barriers. In the context of AD, the deterioration of the local microenvironment in the brain is discussed, mainly focusing on neuroinflammation, oxidative stress, and the accumulation of toxic proteins. The detrimental changes disturb the delicate balance in the brain, impacting the regenerative capacity of OPCs and compromising myelin integrity. Under pathological conditions, OPCs experience significant alterations in migration and proliferation, leading to impaired differentiation and a reduced ability to produce mature oligodendrocytes. Moreover, myelin degeneration and formation become increasingly active in AD, contributing to progressive neurodegeneration. Finally, we summarize the current therapeutic approaches targeting OPCs in AD. Strategies to revitalize OPC senescence, modulate signaling pathways to enhance OPC differentiation, and explore other potential therapeutic avenues are promising in alleviating the impact of AD on OPCs and CNS function. In conclusion, this review highlights the indispensable role of OPCs in CNS function and their involvement in the pathogenesis of AD. The intricate interplay between OPCs and the AD brain microenvironment underscores the complexity of neurodegenerative diseases. Insights from studying OPCs under pathological conditions provide a foundation for innovative therapeutic strategies targeting OPCs and fostering neurodegeneration. Future research will advance our understanding and management of neurodegenerative diseases, ultimately offering hope for effective treatments and improved quality of life for those affected by AD and related disorders.

少突胶质细胞祖细胞(OPCs)在髓磷脂形成和吞噬中起关键作用,与邻近细胞沟通,并有助于血脑屏障(BBB)的完整性。然而,在阿尔茨海默病(AD)的病理情况下,大脑的微环境发生了有害的变化,显著影响OPCs及其功能。从OPC的功能开始,我们深入探讨了OPC向产生髓磷脂的少突胶质细胞的转化,与中枢神经系统(CNS)中其他细胞复杂的信号相互作用,以及吞噬的奇妙过程,这些过程影响OPC的功能和影响中枢神经系统的稳态。此外,我们讨论了OPCs在血脑屏障形成中的重要作用,并强调了OPCs在形成cns保护性屏障中的重要贡献。在AD的背景下,讨论了大脑局部微环境的恶化,主要集中在神经炎症,氧化应激和有毒蛋白的积累。这些有害的变化扰乱了大脑的微妙平衡,影响了OPCs的再生能力,损害了髓磷脂的完整性。病理条件下,OPCs在迁移和增殖方面发生显著改变,导致分化受损,产生成熟少突胶质细胞的能力降低。此外,髓鞘变性和形成在AD中变得越来越活跃,有助于进行性神经变性。最后,我们总结了目前针对OPCs治疗AD的治疗方法。激活OPC衰老,调节信号通路以增强OPC分化,以及探索其他潜在的治疗途径有望减轻AD对OPC和中枢神经系统功能的影响。综上所述,本综述强调了OPCs在中枢神经系统功能中不可或缺的作用及其在AD发病机制中的作用。OPCs与AD脑微环境之间复杂的相互作用凸显了神经退行性疾病的复杂性。病理条件下对OPCs的研究为针对OPCs和促进神经变性的创新治疗策略提供了基础。未来的研究将促进我们对神经退行性疾病的理解和管理,最终为阿尔茨海默病和相关疾病患者提供有效治疗和改善生活质量的希望。
{"title":"Oligodendrocyte progenitor cells in Alzheimer's disease: from physiology to pathology.","authors":"Peibin Zou, Chongyun Wu, Timon Cheng-Yi Liu, Rui Duan, Luodan Yang","doi":"10.1186/s40035-023-00385-7","DOIUrl":"10.1186/s40035-023-00385-7","url":null,"abstract":"<p><p>Oligodendrocyte progenitor cells (OPCs) play pivotal roles in myelin formation and phagocytosis, communicating with neighboring cells and contributing to the integrity of the blood-brain barrier (BBB). However, under the pathological circumstances of Alzheimer's disease (AD), the brain's microenvironment undergoes detrimental changes that significantly impact OPCs and their functions. Starting with OPC functions, we delve into the transformation of OPCs to myelin-producing oligodendrocytes, the intricate signaling interactions with other cells in the central nervous system (CNS), and the fascinating process of phagocytosis, which influences the function of OPCs and affects CNS homeostasis. Moreover, we discuss the essential role of OPCs in BBB formation and highlight the critical contribution of OPCs in forming CNS-protective barriers. In the context of AD, the deterioration of the local microenvironment in the brain is discussed, mainly focusing on neuroinflammation, oxidative stress, and the accumulation of toxic proteins. The detrimental changes disturb the delicate balance in the brain, impacting the regenerative capacity of OPCs and compromising myelin integrity. Under pathological conditions, OPCs experience significant alterations in migration and proliferation, leading to impaired differentiation and a reduced ability to produce mature oligodendrocytes. Moreover, myelin degeneration and formation become increasingly active in AD, contributing to progressive neurodegeneration. Finally, we summarize the current therapeutic approaches targeting OPCs in AD. Strategies to revitalize OPC senescence, modulate signaling pathways to enhance OPC differentiation, and explore other potential therapeutic avenues are promising in alleviating the impact of AD on OPCs and CNS function. In conclusion, this review highlights the indispensable role of OPCs in CNS function and their involvement in the pathogenesis of AD. The intricate interplay between OPCs and the AD brain microenvironment underscores the complexity of neurodegenerative diseases. Insights from studying OPCs under pathological conditions provide a foundation for innovative therapeutic strategies targeting OPCs and fostering neurodegeneration. Future research will advance our understanding and management of neurodegenerative diseases, ultimately offering hope for effective treatments and improved quality of life for those affected by AD and related disorders.</p>","PeriodicalId":23269,"journal":{"name":"Translational Neurodegeneration","volume":"12 1","pages":"52"},"PeriodicalIF":12.6,"publicationDate":"2023-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10644503/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"107592281","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
A novel transgenic mouse line with hippocampus-dominant and inducible expression of truncated human tau. 一种新的转基因小鼠系,具有截短的人tau的海马显性和诱导表达。
IF 12.6 1区 医学 Q1 NEUROSCIENCES Pub Date : 2023-11-10 DOI: 10.1186/s40035-023-00379-5
Yang Gao, Yuying Wang, Huiyang Lei, Zhendong Xu, Shihong Li, Haitao Yu, Jiazhao Xie, Zhentao Zhang, Gongping Liu, Yao Zhang, Jie Zheng, Jian-Zhi Wang

Background: Intraneuronal accumulation of hyperphosphorylated tau is a defining hallmark of Alzheimer's disease (AD). However, mouse models imitating AD-exclusive neuronal tau pathologies are lacking.

Methods: We generated a new tet-on transgenic mouse model expressing truncated human tau N1-368 (termed hTau368), a tau fragment increased in the brains of AD patients and aged mouse brains. Doxycycline (dox) was administered in drinking water to induce hTau368 expression. Immunostaining and Western blotting were performed to measure the tau level. RNA sequencing was performed to evaluate gene expression, and several behavioral tests were conducted to evaluate mouse cognitive functions, emotion and locomotion.

Results: Dox treatment for 1-2 months at a young age induced overt and reversible human tau accumulation in the brains of hTau368 transgenic mice, predominantly in the hippocampus. Meanwhile, the transgenic mice exhibited AD-like high level of tau phosphorylation, glial activation, loss of mature neurons, impaired hippocampal neurogenesis, synaptic degeneration and cognitive deficits.

Conclusions: This study developed a well-characterized and easy-to-use tool for the investigations and drug development for AD and other tauopathies.

背景:神经元内过度磷酸化tau的积累是阿尔茨海默病(AD)的一个明确标志。然而,缺乏模拟AD专属神经元tau病理的小鼠模型。方法:我们产生了一种新的tet-on转基因小鼠模型,表达截短的人类tau N1-368(称为hTau368),一种在AD患者和老年小鼠大脑中增加的tau片段。在饮用水中施用多西环素(dox)以诱导hTau368的表达。进行免疫染色和蛋白质印迹以测量tau水平。进行RNA测序以评估基因表达,并进行多项行为测试以评估小鼠的认知功能、情绪和运动。结果:年轻时接受Dox治疗1-2个月,在hTau368转基因小鼠的大脑中诱导了明显且可逆的人类tau积累,主要在海马中。同时,转基因小鼠表现出AD样高水平的tau磷酸化、胶质细胞活化、成熟神经元丧失、海马神经发生受损、突触变性和认知缺陷。结论:本研究为AD和其他tau病的研究和药物开发开发开发了一种功能齐全、易于使用的工具。
{"title":"A novel transgenic mouse line with hippocampus-dominant and inducible expression of truncated human tau.","authors":"Yang Gao, Yuying Wang, Huiyang Lei, Zhendong Xu, Shihong Li, Haitao Yu, Jiazhao Xie, Zhentao Zhang, Gongping Liu, Yao Zhang, Jie Zheng, Jian-Zhi Wang","doi":"10.1186/s40035-023-00379-5","DOIUrl":"10.1186/s40035-023-00379-5","url":null,"abstract":"<p><strong>Background: </strong>Intraneuronal accumulation of hyperphosphorylated tau is a defining hallmark of Alzheimer's disease (AD). However, mouse models imitating AD-exclusive neuronal tau pathologies are lacking.</p><p><strong>Methods: </strong>We generated a new tet-on transgenic mouse model expressing truncated human tau N1-368 (termed hTau368), a tau fragment increased in the brains of AD patients and aged mouse brains. Doxycycline (dox) was administered in drinking water to induce hTau368 expression. Immunostaining and Western blotting were performed to measure the tau level. RNA sequencing was performed to evaluate gene expression, and several behavioral tests were conducted to evaluate mouse cognitive functions, emotion and locomotion.</p><p><strong>Results: </strong>Dox treatment for 1-2 months at a young age induced overt and reversible human tau accumulation in the brains of hTau368 transgenic mice, predominantly in the hippocampus. Meanwhile, the transgenic mice exhibited AD-like high level of tau phosphorylation, glial activation, loss of mature neurons, impaired hippocampal neurogenesis, synaptic degeneration and cognitive deficits.</p><p><strong>Conclusions: </strong>This study developed a well-characterized and easy-to-use tool for the investigations and drug development for AD and other tauopathies.</p>","PeriodicalId":23269,"journal":{"name":"Translational Neurodegeneration","volume":"12 1","pages":"51"},"PeriodicalIF":12.6,"publicationDate":"2023-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10637005/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"72210935","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Intranasal administration of induced pluripotent stem cell-derived cortical neural stem cell-secretome as a treatment option for Alzheimer's disease. 鼻内给药诱导多能干细胞衍生的皮层神经干细胞分泌组作为阿尔茨海默病的治疗选择。
IF 12.6 1区 医学 Q1 NEUROSCIENCES Pub Date : 2023-11-09 DOI: 10.1186/s40035-023-00384-8
Hyunkyung Mo, Juryun Kim, Jennifer Yejean Kim, Jang Woon Kim, Heeju Han, Si Hwa Choi, Yeri Alice Rim, Ji Hyeon Ju

Background: Alzheimer's disease (AD) is the most common neurodegenerative disorder in the elderly, resulting in gradual destruction of cognitive abilities. Research on the development of various AD treatments is underway; however, no definitive treatment has been developed yet. Herein, we present induced pluripotent stem cell (iPSC)-derived cortical neural stem cell secretome (CNSC-SE) as a new treatment candidate for AD and explore its efficacy.

Methods: We first assessed the effects of CNSC-SE treatment on neural maturation and electromagnetic signal during cortical nerve cell differentiation. Then to confirm the efficacy in vivo, CNSC-SE was administered to the 5×FAD mouse model through the nasal cavity (5 μg/g, once a week, 4 weeks). The cell-mediated effects on nerve recovery, amyloid beta (Aβ) plaque aggregation, microglial and astrocyte detection in the brain, and neuroinflammatory responses were investigated. Metabolomics analysis of iPSC-derived CNSC-SE revealed that it contained components that could exert neuro-protective effects or amplify cognitive restorative effects.

Results: Human iPSC-derived CNSC-SE increased neuronal proliferation and dendritic structure formation in vitro. Furthermore, CNSC-SE-treated iPSC-derived cortical neurons acquired electrical network activity and action potential bursts. The 5×FAD mice treated with CNSC-SE showed memory restoration and reduced Aβ plaque accumulation.

Conclusions: Our findings suggest that the iPSC-derived CNSC-SE may serve as a potential, non-invasive therapeutic option for AD in reducing amyloid infiltration and restoring memory.

背景:阿尔茨海默病(AD)是老年人最常见的神经退行性疾病,导致认知能力逐渐受损。各种AD治疗方法的开发研究正在进行中;然而,目前还没有明确的治疗方法。在此,我们提出了诱导多能干细胞(iPSC)衍生的皮层神经干细胞分泌组(CNSC-SE)作为AD的一种新的候选治疗方法,并探索其疗效。方法:我们首先评估了CNSC-SE处理对皮层神经细胞分化过程中神经成熟和电磁信号的影响。然后,为了确认体内疗效,通过鼻腔向5×FAD小鼠模型施用CNSC-SE(5μg/g,每周一次,4周)。研究了细胞介导的对神经恢复、淀粉样蛋白β(Aβ)斑块聚集、脑中小胶质细胞和星形胶质细胞检测以及神经炎症反应的影响。iPSC衍生的CNSC-SE的代谢组学分析表明,它含有可以发挥神经保护作用或增强认知恢复作用的成分。结果:人iPSC衍生的CNSC-SE在体外增加了神经元增殖和树突结构的形成。此外,CNSC SE处理的iPSC衍生的皮层神经元获得了电网络活动和动作电位爆发。CNSC-SE处理的5×FAD小鼠表现出记忆恢复和Aβ斑块积聚减少。结论:我们的研究结果表明,iPSC衍生的CNSC-SE可能是AD减少淀粉样蛋白浸润和恢复记忆的一种潜在的非侵入性治疗选择。
{"title":"Intranasal administration of induced pluripotent stem cell-derived cortical neural stem cell-secretome as a treatment option for Alzheimer's disease.","authors":"Hyunkyung Mo, Juryun Kim, Jennifer Yejean Kim, Jang Woon Kim, Heeju Han, Si Hwa Choi, Yeri Alice Rim, Ji Hyeon Ju","doi":"10.1186/s40035-023-00384-8","DOIUrl":"10.1186/s40035-023-00384-8","url":null,"abstract":"<p><strong>Background: </strong>Alzheimer's disease (AD) is the most common neurodegenerative disorder in the elderly, resulting in gradual destruction of cognitive abilities. Research on the development of various AD treatments is underway; however, no definitive treatment has been developed yet. Herein, we present induced pluripotent stem cell (iPSC)-derived cortical neural stem cell secretome (CNSC-SE) as a new treatment candidate for AD and explore its efficacy.</p><p><strong>Methods: </strong>We first assessed the effects of CNSC-SE treatment on neural maturation and electromagnetic signal during cortical nerve cell differentiation. Then to confirm the efficacy in vivo, CNSC-SE was administered to the 5×FAD mouse model through the nasal cavity (5 μg/g, once a week, 4 weeks). The cell-mediated effects on nerve recovery, amyloid beta (Aβ) plaque aggregation, microglial and astrocyte detection in the brain, and neuroinflammatory responses were investigated. Metabolomics analysis of iPSC-derived CNSC-SE revealed that it contained components that could exert neuro-protective effects or amplify cognitive restorative effects.</p><p><strong>Results: </strong>Human iPSC-derived CNSC-SE increased neuronal proliferation and dendritic structure formation in vitro. Furthermore, CNSC-SE-treated iPSC-derived cortical neurons acquired electrical network activity and action potential bursts. The 5×FAD mice treated with CNSC-SE showed memory restoration and reduced Aβ plaque accumulation.</p><p><strong>Conclusions: </strong>Our findings suggest that the iPSC-derived CNSC-SE may serve as a potential, non-invasive therapeutic option for AD in reducing amyloid infiltration and restoring memory.</p>","PeriodicalId":23269,"journal":{"name":"Translational Neurodegeneration","volume":"12 1","pages":"50"},"PeriodicalIF":12.6,"publicationDate":"2023-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10634159/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"72015542","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
NLRP3 inflammasome in cognitive impairment and pharmacological properties of its inhibitors. 认知障碍中的NLRP3炎症小体及其抑制剂的药理学特性。
IF 12.6 1区 医学 Q1 NEUROSCIENCES Pub Date : 2023-11-02 DOI: 10.1186/s40035-023-00381-x
Yi Xu, Yanling Yang, Xi Chen, Danling Jiang, Fei Zhang, Yao Guo, Bin Hu, Guohai Xu, Shengliang Peng, Lidong Wu, Jialing Hu

Cognitive impairment is a multifactorial and multi-step pathological process that places a heavy burden on patients and the society. Neuroinflammation is one of the main factors leading to cognitive impairment. The inflammasomes are multi-protein complexes that respond to various microorganisms and endogenous danger signals, helping to initiate innate protective responses in inflammatory diseases. NLRP3 inflammasomes produce proinflammatory cytokines (interleukin IL-1β and IL-18) by activating caspase-1. In this review, we comprehensively describe the structure and functions of the NLRP3 inflammasome. We also explore the intrinsic relationship between the NLRP3 inflammasome and cognitive impairment, which involves immune cell activation, cell apoptosis, oxidative stress, mitochondrial autophagy, and neuroinflammation. Finally, we describe NLRP3 inflammasome antagonists as targeted therapies to improve cognitive impairment.

认知障碍是一个多因素、多步骤的病理过程,给患者和社会带来了沉重的负担。神经炎症是导致认知障碍的主要因素之一。炎症小体是一种多蛋白复合物,对各种微生物和内源性危险信号作出反应,有助于在炎症性疾病中启动先天保护反应。NLRP3炎症小体通过激活胱天蛋白酶1产生促炎细胞因子(白细胞介素IL-1β和IL-18)。在这篇综述中,我们全面描述了NLRP3炎症小体的结构和功能。我们还探讨了NLRP3炎症小体与认知障碍之间的内在关系,认知障碍涉及免疫细胞活化、细胞凋亡、氧化应激、线粒体自噬和神经炎症。最后,我们将NLRP3炎症小体拮抗剂描述为改善认知障碍的靶向疗法。
{"title":"NLRP3 inflammasome in cognitive impairment and pharmacological properties of its inhibitors.","authors":"Yi Xu, Yanling Yang, Xi Chen, Danling Jiang, Fei Zhang, Yao Guo, Bin Hu, Guohai Xu, Shengliang Peng, Lidong Wu, Jialing Hu","doi":"10.1186/s40035-023-00381-x","DOIUrl":"10.1186/s40035-023-00381-x","url":null,"abstract":"<p><p>Cognitive impairment is a multifactorial and multi-step pathological process that places a heavy burden on patients and the society. Neuroinflammation is one of the main factors leading to cognitive impairment. The inflammasomes are multi-protein complexes that respond to various microorganisms and endogenous danger signals, helping to initiate innate protective responses in inflammatory diseases. NLRP3 inflammasomes produce proinflammatory cytokines (interleukin IL-1β and IL-18) by activating caspase-1. In this review, we comprehensively describe the structure and functions of the NLRP3 inflammasome. We also explore the intrinsic relationship between the NLRP3 inflammasome and cognitive impairment, which involves immune cell activation, cell apoptosis, oxidative stress, mitochondrial autophagy, and neuroinflammation. Finally, we describe NLRP3 inflammasome antagonists as targeted therapies to improve cognitive impairment.</p>","PeriodicalId":23269,"journal":{"name":"Translational Neurodegeneration","volume":"12 1","pages":"49"},"PeriodicalIF":12.6,"publicationDate":"2023-11-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10621314/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71427161","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Metabolic regulation of microglial phagocytosis: Implications for Alzheimer's disease therapeutics. 小胶质细胞吞噬作用的代谢调节:对阿尔茨海默病治疗的启示。
IF 12.6 1区 医学 Q1 NEUROSCIENCES Pub Date : 2023-10-31 DOI: 10.1186/s40035-023-00382-w
Izabela Lepiarz-Raba, Ismail Gbadamosi, Roberta Florea, Rosa Chiara Paolicelli, Ali Jawaid

Microglia, the resident immune cells of the brain, are increasingly implicated in the regulation of brain health and disease. Microglia perform multiple functions in the central nervous system, including surveillance, phagocytosis and release of a variety of soluble factors. Importantly, a majority of their functions are closely related to changes in their metabolism. This natural inter-dependency between core microglial properties and metabolism offers a unique opportunity to modulate microglial activities via nutritional or metabolic interventions. In this review, we examine the existing scientific literature to synthesize the hypothesis that microglial phagocytosis of amyloid beta (Aβ) aggregates in Alzheimer's disease (AD) can be selectively enhanced via metabolic interventions. We first review the basics of microglial metabolism and the effects of common metabolites, such as glucose, lipids, ketone bodies, glutamine, pyruvate and lactate, on microglial inflammatory and phagocytic properties. Next, we examine the evidence for dysregulation of microglial metabolism in AD. This is followed by a review of in vivo studies on metabolic manipulation of microglial functions to ascertain their therapeutic potential in AD. Finally, we discuss the effects of metabolic factors on microglial phagocytosis of healthy synapses, a pathological process that also contributes to the progression of AD. We conclude by enlisting the current challenges that need to be addressed before strategies to harness microglial phagocytosis to clear pathological protein deposits in AD and other neurodegenerative disorders can be widely adopted.

小胶质细胞是大脑的固有免疫细胞,越来越多地参与大脑健康和疾病的调节。小胶质细胞在中枢神经系统中发挥多种功能,包括监测、吞噬和释放各种可溶性因子。重要的是,它们的大部分功能与新陈代谢的变化密切相关。核心小胶质细胞特性和代谢之间的这种自然相互依赖性为通过营养或代谢干预调节小胶质细胞活性提供了独特的机会。在这篇综述中,我们检查了现有的科学文献,以综合以下假设:阿尔茨海默病(AD)中淀粉样蛋白β(Aβ)聚集体的小胶质细胞吞噬作用可以通过代谢干预选择性增强。我们首先综述了小胶质细胞代谢的基础以及常见代谢产物,如葡萄糖、脂质、酮体、谷氨酰胺、丙酮酸盐和乳酸,对小胶质细胞炎症和吞噬特性的影响。接下来,我们研究了AD中小胶质细胞代谢失调的证据。随后,我们对小胶质细胞功能代谢调控的体内研究进行了综述,以确定其在AD中的治疗潜力。最后,我们讨论了代谢因子对健康突触的小胶质细胞吞噬作用的影响,这是一个也有助于AD进展的病理过程。在广泛采用利用小胶质细胞吞噬作用清除AD和其他神经退行性疾病病理性蛋白质沉积的策略之前,我们总结了当前需要解决的挑战。
{"title":"Metabolic regulation of microglial phagocytosis: Implications for Alzheimer's disease therapeutics.","authors":"Izabela Lepiarz-Raba, Ismail Gbadamosi, Roberta Florea, Rosa Chiara Paolicelli, Ali Jawaid","doi":"10.1186/s40035-023-00382-w","DOIUrl":"10.1186/s40035-023-00382-w","url":null,"abstract":"<p><p>Microglia, the resident immune cells of the brain, are increasingly implicated in the regulation of brain health and disease. Microglia perform multiple functions in the central nervous system, including surveillance, phagocytosis and release of a variety of soluble factors. Importantly, a majority of their functions are closely related to changes in their metabolism. This natural inter-dependency between core microglial properties and metabolism offers a unique opportunity to modulate microglial activities via nutritional or metabolic interventions. In this review, we examine the existing scientific literature to synthesize the hypothesis that microglial phagocytosis of amyloid beta (Aβ) aggregates in Alzheimer's disease (AD) can be selectively enhanced via metabolic interventions. We first review the basics of microglial metabolism and the effects of common metabolites, such as glucose, lipids, ketone bodies, glutamine, pyruvate and lactate, on microglial inflammatory and phagocytic properties. Next, we examine the evidence for dysregulation of microglial metabolism in AD. This is followed by a review of in vivo studies on metabolic manipulation of microglial functions to ascertain their therapeutic potential in AD. Finally, we discuss the effects of metabolic factors on microglial phagocytosis of healthy synapses, a pathological process that also contributes to the progression of AD. We conclude by enlisting the current challenges that need to be addressed before strategies to harness microglial phagocytosis to clear pathological protein deposits in AD and other neurodegenerative disorders can be widely adopted.</p>","PeriodicalId":23269,"journal":{"name":"Translational Neurodegeneration","volume":"12 1","pages":"48"},"PeriodicalIF":12.6,"publicationDate":"2023-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10617244/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71427160","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Janus kinase inhibitors are potential therapeutics for amyotrophic lateral sclerosis. Janus激酶抑制剂是肌萎缩侧索硬化症的潜在治疗方法。
IF 10.8 1区 医学 Q1 NEUROSCIENCES Pub Date : 2023-10-12 DOI: 10.1186/s40035-023-00380-y
Peter J Richardson, Daniel P Smith, Alex de Giorgio, Xenia Snetkov, Joshua Almond-Thynne, Sara Cronin, Richard J Mead, Christopher J McDermott, Pamela J Shaw

Amyotrophic lateral sclerosis (ALS) is a poorly treated multifactorial neurodegenerative disease associated with multiple cell types and subcellular organelles. As with other multifactorial diseases, it is likely that drugs will need to target multiple disease processes and cell types to be effective. We review here the role of Janus kinase (JAK)/Signal transducer and activator of transcription (STAT) signalling in ALS, confirm the association of this signalling with fundamental ALS disease processes using the BenevolentAI Knowledge Graph, and demonstrate that inhibitors of this pathway could reduce the ALS pathophysiology in neurons, glia, muscle fibres, and blood cells. Specifically, we suggest that inhibition of the JAK enzymes by approved inhibitors known as Jakinibs could reduce STAT3 activation and modify the progress of this disease. Analysis of the Jakinibs highlights baricitinib as a suitable candidate due to its ability to penetrate the central nervous system and exert beneficial effects on the immune system. Therefore, we recommend that this drug be tested in appropriately designed clinical trials for ALS.

肌萎缩侧索硬化症(ALS)是一种治疗不力的多因素神经退行性疾病,与多种细胞类型和亚细胞器有关。与其他多因素疾病一样,药物可能需要针对多种疾病过程和细胞类型才能有效。我们在此综述了Janus激酶(JAK)/信号转导子和转录激活子(STAT)信号在ALS中的作用,使用BenevolentAI知识图证实了这种信号与ALS基本疾病过程的关联,并证明了这种途径的抑制剂可以减少神经元、神经胶质、肌肉纤维和血细胞中的ALS病理生理学。具体而言,我们认为,被批准的Jakinibs抑制剂对JAK酶的抑制可以减少STAT3的激活,并改变这种疾病的进展。对雅替尼的分析强调,巴里西替尼是一种合适的候选药物,因为它能够穿透中枢神经系统并对免疫系统产生有益影响。因此,我们建议在适当设计的ALS临床试验中测试这种药物。
{"title":"Janus kinase inhibitors are potential therapeutics for amyotrophic lateral sclerosis.","authors":"Peter J Richardson, Daniel P Smith, Alex de Giorgio, Xenia Snetkov, Joshua Almond-Thynne, Sara Cronin, Richard J Mead, Christopher J McDermott, Pamela J Shaw","doi":"10.1186/s40035-023-00380-y","DOIUrl":"10.1186/s40035-023-00380-y","url":null,"abstract":"<p><p>Amyotrophic lateral sclerosis (ALS) is a poorly treated multifactorial neurodegenerative disease associated with multiple cell types and subcellular organelles. As with other multifactorial diseases, it is likely that drugs will need to target multiple disease processes and cell types to be effective. We review here the role of Janus kinase (JAK)/Signal transducer and activator of transcription (STAT) signalling in ALS, confirm the association of this signalling with fundamental ALS disease processes using the BenevolentAI Knowledge Graph, and demonstrate that inhibitors of this pathway could reduce the ALS pathophysiology in neurons, glia, muscle fibres, and blood cells. Specifically, we suggest that inhibition of the JAK enzymes by approved inhibitors known as Jakinibs could reduce STAT3 activation and modify the progress of this disease. Analysis of the Jakinibs highlights baricitinib as a suitable candidate due to its ability to penetrate the central nervous system and exert beneficial effects on the immune system. Therefore, we recommend that this drug be tested in appropriately designed clinical trials for ALS.</p>","PeriodicalId":23269,"journal":{"name":"Translational Neurodegeneration","volume":"12 1","pages":"47"},"PeriodicalIF":10.8,"publicationDate":"2023-10-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10568794/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41213960","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Patient-derived iPSC models of Friedreich ataxia: a new frontier for understanding disease mechanisms and therapeutic application. Friedreich共济失调的患者衍生iPSC模型:理解疾病机制和治疗应用的新前沿。
IF 12.6 1区 医学 Q1 NEUROSCIENCES Pub Date : 2023-09-20 DOI: 10.1186/s40035-023-00376-8
Saumya Maheshwari, Gabriela Vilema-Enríquez, Richard Wade-Martins

Friedreich ataxia (FRDA) is a rare genetic multisystem disorder caused by a pathological GAA trinucleotide repeat expansion in the FXN gene. The numerous drawbacks of historical cellular and rodent models of FRDA have caused difficulty in performing effective mechanistic and translational studies to investigate the disease. The recent discovery and subsequent development of induced pluripotent stem cell (iPSC) technology provides an exciting platform to enable enhanced disease modelling for studies of rare genetic diseases. Utilising iPSCs, researchers have created phenotypically relevant and previously inaccessible cellular models of FRDA. These models enable studies of the molecular mechanisms underlying GAA-induced pathology, as well as providing an exciting tool for the screening and testing of novel disease-modifying therapies. This review explores how the use of iPSCs to study FRDA has developed over the past decade, as well as discussing the enormous therapeutic potentials of iPSC-derived models, their current limitations and their future direction within the field of FRDA research.

弗里德里希共济失调(FRDA)是一种罕见的遗传性多系统疾病,由FXN基因中病理性GAA三核苷酸重复扩增引起。FRDA的历史细胞和啮齿动物模型的许多缺点导致难以进行有效的机制和转化研究来研究该疾病。诱导多能干细胞(iPSC)技术的最新发现和随后的发展为罕见遗传疾病的研究提供了一个令人兴奋的平台。利用iPSC,研究人员创建了与表型相关且以前无法访问的FRDA细胞模型。这些模型能够研究GAA诱导的病理学的分子机制,并为筛选和测试新的疾病修饰疗法提供了一个令人兴奋的工具。这篇综述探讨了在过去十年中使用iPSC研究FRDA的进展,并讨论了iPSC衍生模型的巨大治疗潜力、其目前的局限性及其在FRDA研究领域的未来方向。
{"title":"Patient-derived iPSC models of Friedreich ataxia: a new frontier for understanding disease mechanisms and therapeutic application.","authors":"Saumya Maheshwari, Gabriela Vilema-Enríquez, Richard Wade-Martins","doi":"10.1186/s40035-023-00376-8","DOIUrl":"10.1186/s40035-023-00376-8","url":null,"abstract":"<p><p>Friedreich ataxia (FRDA) is a rare genetic multisystem disorder caused by a pathological GAA trinucleotide repeat expansion in the FXN gene. The numerous drawbacks of historical cellular and rodent models of FRDA have caused difficulty in performing effective mechanistic and translational studies to investigate the disease. The recent discovery and subsequent development of induced pluripotent stem cell (iPSC) technology provides an exciting platform to enable enhanced disease modelling for studies of rare genetic diseases. Utilising iPSCs, researchers have created phenotypically relevant and previously inaccessible cellular models of FRDA. These models enable studies of the molecular mechanisms underlying GAA-induced pathology, as well as providing an exciting tool for the screening and testing of novel disease-modifying therapies. This review explores how the use of iPSCs to study FRDA has developed over the past decade, as well as discussing the enormous therapeutic potentials of iPSC-derived models, their current limitations and their future direction within the field of FRDA research.</p>","PeriodicalId":23269,"journal":{"name":"Translational Neurodegeneration","volume":"12 1","pages":"45"},"PeriodicalIF":12.6,"publicationDate":"2023-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10510273/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41166481","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Translational Neurodegeneration
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