brain organoids

脑类器官
  • 文章类型: Journal Article
    环境问题的普遍存在和人类暴露于环境污染物的风险日益增加已成为全球关注的问题。环境污染的加剧是近年来大多数神经系统相关疾病发病率上升的主要原因之一。然而,直接人类研究的伦理限制和动物模型的种族限制减缓了这一领域的研究进展。本研究的目的是以脑类器官为新模型,综述不同环境污染物对大脑的神经毒性,并得出结论,脑类器官可能在评估环境污染物影响神经发生和引起神经发病机制中起关键作用。为了准确确定环境污染物对神经系统的负面影响,与发育中的大脑高度相似的自组织脑类器官已成为研究环境污染物对人脑发育和疾病影响的新模型系统。本研究以脑类器官为模型,总结不同环境污染物对神经系统的神经毒性,包括脑类器官的结构变化,抑制神经元分化和迁移,线粒体功能受损,细胞纤毛的损伤,以及对信号通路的影响。总之,暴露于环境污染物可能会对神经系统造成不同的神经毒性。因此,了解如何使用脑类器官来改善环境污染引起的神经系统疾病至关重要。
    The prevalence of environmental problems and the increasing risk of human exposure to environmental pollutants have become a global concern. The increasing environmental pollution is one of the main reasons for the rising incidence of most neurological-related diseases in recent years. However, the ethical constraints of direct human research and the racial limitations of animal models have slowed the progress of research in this area. The purpose of this study is to review the neurotoxicity of different environmental pollutants on the brain using brain organoids as a new model and to conclude that brain organoids may play a key role in assessing the mechanisms by which environmental pollutants affect neurogenesis and cause neurological pathogenesis. To accurately determine the negative effects of environmental pollutants on the nervous system, self-organizing brain organoids that are highly similar to the developing brain have become a new model system for studying the effects of environmental pollutants on human brain development and disease. This study uses brain organoids as a model to summarize the neurotoxicity of different environmental pollutants on the nervous system, including structural changes in brain organoids, inhibition of neuronal differentiation and migration, impairment of mitochondrial function, damage to cellular cilia, and influence on signaling pathways. In conclusion, exposure to environmental pollutants may cause different neurotoxicity to the nervous system. Therefore, it is crucial to understand how to use brain organoids to ameliorate neurological disorders caused by environmental pollution.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    中枢神经系统(CNS)神经胶质在维持多发性硬化症(MS)的自我自主炎症和驱动临床进展中的作用正在引起科学兴趣。我们应用了基于单一转录因子(SOX10)的方案来加速人类诱导多能干细胞(hiPSC)衍生的神经前体细胞的少突胶质细胞分化,产生自组织的前脑器官。这些类器官包括神经元,星形胶质细胞,少突胶质细胞,和hiPSC衍生的小胶质细胞来实现免疫能力。超过8周,类器官可重复生成的成熟CNS细胞类型,表现出与成人大脑相似的单细胞转录谱。暴露于MS患者的脑脊液(CSF)发炎,类器官适当地模拟了慢性活动性MS中的大胶质细胞-小胶质细胞神经变性表型和细胞间通讯。少突胶质细胞易损性在MS-CSF暴露后第6天出现,减少近50%。时间分辨的类器官数据支持并扩展了可溶性CSF介质在维持导致少突胶质细胞死亡和炎性神经变性的下游事件中的作用。这些发现支持用于药物筛选以停止炎性神经变性的这种类器官模型的实施。
    The role of central nervous system (CNS) glia in sustaining self-autonomous inflammation and driving clinical progression in multiple sclerosis (MS) is gaining scientific interest. We applied a single transcription factor (SOX10)-based protocol to accelerate oligodendrocyte differentiation from human induced pluripotent stem cell (hiPSC)-derived neural precursor cells, generating self-organizing forebrain organoids. These organoids include neurons, astrocytes, oligodendroglia, and hiPSC-derived microglia to achieve immunocompetence. Over 8 weeks, organoids reproducibly generated mature CNS cell types, exhibiting single-cell transcriptional profiles similar to the adult human brain. Exposed to inflamed cerebrospinal fluid (CSF) from patients with MS, organoids properly mimic macroglia-microglia neurodegenerative phenotypes and intercellular communication seen in chronic active MS. Oligodendrocyte vulnerability emerged by day 6 post-MS-CSF exposure, with nearly 50% reduction. Temporally resolved organoid data support and expand on the role of soluble CSF mediators in sustaining downstream events leading to oligodendrocyte death and inflammatory neurodegeneration. Such findings support the implementation of this organoid model for drug screening to halt inflammatory neurodegeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    人胚胎干细胞和人诱导多能干细胞可用于创建称为脑类器官的3D组织。它们在结构和功能两方面更忠实地复制了人脑组织的生理和病理特征,它们更精确地类似于人类胚胎大脑的形态和细胞结构。这使它们成为有价值的模型,用于药物筛选和关于人脑和相关疾病发展的体外研究。脑类器官实现的技术突破对不同脑区的研究产生了重大影响,大脑发育和疾病,大脑与其他组织和器官之间的联系,和大脑进化。本文讨论了脑类器官的发育,它们在糖尿病研究中的应用,和他们的进步。
    Human embryonic stem cells and human induced pluripotent stem cells may be used to create 3D tissues called brain organoids. They duplicate the physiological and pathological characteristics of human brain tissue more faithfully in terms of both structure and function, and they more precisely resemble the morphology and cellular structure of the human embryonic brain. This makes them valuable models for both drug screening and in vitro studies on the development of the human brain and associated disorders. The technical breakthroughs enabled by brain organoids have a significant impact on the research of different brain regions, brain development and sickness, the connections between the brain and other tissues and organs, and brain evolution. This article discusses the development of brain organoids, their use in diabetes research, and their progress.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    性别差异在神经发育过程中普遍存在,并在自闭症等神经精神疾病中发挥作用,男性比女性更普遍。在人类中,男性的大脑体积比女性大,海马体和杏仁核的发育显示出明显的性别差异。机械上,性类固醇和性染色体驱动大脑发育的这些差异,似乎在产前和青春期达到高峰。动物模型在理解性别差异方面发挥了至关重要的作用,但是对人类性别差异的研究需要一个能够概括复杂遗传性状的实验模型。为了填补这个空白,人类诱导的多能干细胞来源的脑类器官现在被用于研究复杂的遗传特征如何影响产前大脑发育。例如,自闭症患者和X染色体连锁Rett综合征和脆性X综合征患者的脑类器官显示了产前细胞增殖的差异,测量大脑体积差异,和兴奋性抑制失衡。脑类器官还揭示了由于雄激素导致的兴奋性神经元的神经发生增加。然而,尽管人们对使用脑类器官越来越感兴趣,几个关键挑战仍然存在,影响其作为模型系统的有效性。在这次审查中,我们讨论了性类固醇和性染色体是如何导致大脑发育中的性别差异的。然后,我们研究了X染色体失活作为驱动性别差异的一个因素的作用.最后,我们讨论了在研究性别差异时需要考虑的X染色体失活建模和脑类器官局限性的综合挑战。
    性别差异是自闭症等神经精神疾病的一个促成因素,这在男性中更为普遍。性别差异是通过雌激素和睾丸激素等性类固醇激素与性染色体(chrX和chrY)之间的相互作用而发生的。人类干细胞衍生的脑类器官是模拟大脑发育的实验室模型。例如,在有神经发育状况的个体中,与神经典型个体相比,脑类器官显示出神经元种群的不平衡。在这次审查中,我们讨论了性类固醇和性染色体对大脑发育的影响,以及研究性别差异时需要考虑的挑战。
    Sex differences are widespread during neurodevelopment and play a role in neuropsychiatric conditions such as autism, which is more prevalent in males than females. In humans, males have been shown to have larger brain volumes than females with development of the hippocampus and amygdala showing prominent sex differences. Mechanistically, sex steroids and sex chromosomes drive these differences in brain development, which seem to peak during prenatal and pubertal stages. Animal models have played a crucial role in understanding sex differences, but the study of human sex differences requires an experimental model that can recapitulate complex genetic traits. To fill this gap, human induced pluripotent stem cell-derived brain organoids are now being used to study how complex genetic traits influence prenatal brain development. For example, brain organoids from individuals with autism and individuals with X chromosome-linked Rett syndrome and fragile X syndrome have revealed prenatal differences in cell proliferation, a measure of brain volume differences, and excitatory-inhibitory imbalances. Brain organoids have also revealed increased neurogenesis of excitatory neurons due to androgens. However, despite growing interest in using brain organoids, several key challenges remain that affect its validity as a model system. In this review, we discuss how sex steroids and the sex chromosomes each contribute to sex differences in brain development. Then, we examine the role of X chromosome inactivation as a factor that drives sex differences. Finally, we discuss the combined challenges of modeling X chromosome inactivation and limitations of brain organoids that need to be taken into consideration when studying sex differences.
    Sex differences are a contributing factor in neuropsychiatric conditions such as autism, which is more prevalent in males. Sex differences occur through interactions between sex steroid hormones such as estrogen and testosterone and sex chromosomes (chrX and chrY). Human stem cell–derived brain organoids are laboratory models that mimic brain development. For example, in individuals with neurodevelopmental conditions, brain organoids have revealed an imbalance of neuron populations compared with neurotypical individuals. In this review, we discuss sex steroid and sex chromosome influences on brain development and challenges of this model that need to be taken into account when studying sex differences.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    人脑类器官产生解剖学相关的细胞结构,并概括体内脑功能的关键方面,它具有模拟神经系统疾病和筛选治疗方法的巨大潜力。然而,3D系统的长生长时间使得脑类器官的培养变得复杂,并且导致样品间的异质性妨碍了它们的应用。我们开发了一个集成平台,以实现3D脑类器官的强大和长期培养。我们设计了一种基于反应-扩散缩放理论的中流生物反应器装置,这实现了强大的培养基交换,以在长期培养中提供足够的营养。我们将该设备与纵向跟踪和基于机器学习的分类工具集成在一起,以实现对类器官的非侵入性质量控制。该集成平台允许样品预选以进行下游分子分析。类器官的转录组分析显示,我们的中流生物反应器促进了类器官的发育,同时减少了细胞死亡。因此,我们的平台提供了一个可推广的工具,可以为3D细胞系统建立可重复的培养标准,用于大脑器官以外的各种应用。
    Human brain organoids produce anatomically relevant cellular structures and recapitulate key aspects of in vivo brain function, which holds great potential to model neurological diseases and screen therapeutics. However, the long growth time of 3D systems complicates the culturing of brain organoids and results in heterogeneity across samples hampering their applications. We developed an integrated platform to enable robust and long-term culturing of 3D brain organoids. We designed a mesofluidic bioreactor device based on a reaction-diffusion scaling theory, which achieves robust media exchange for sufficient nutrient delivery in long-term culture. We integrated this device with longitudinal tracking and machine learning-based classification tools to enable non-invasive quality control of live organoids. This integrated platform allows for sample pre-selection for downstream molecular analysis. Transcriptome analyses of organoids revealed that our mesofluidic bioreactor promoted organoid development while reducing cell death. Our platform thus offers a generalizable tool to establish reproducible culture standards for 3D cellular systems for a variety of applications beyond brain organoids.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    微重力和太空环境与神经肌肉和认知能力的缺陷有关,假设是由于太空中加速老化和神经变性而发生的。虽然具体机制仍在调查中,航天相关的神经病理学是对宇航员和航天游客的重要健康风险,正在积极研究以制定相应的对策。然而,这种空间诱导的神经病理学为加速筛选用于治疗神经退行性疾病的治疗靶标和先导分子提供了机会。这里,我们展示了一个概念验证高通量目标筛选(在地球上),目标验证,使用我们的纳米低聚物平台缓解微重力诱导的神经病理学,在为期43天的SpaceXCRS-29国际空间站任务上。首先,比较3D健康和患病的前额叶皮层(PFC,用于认知)和运动神经元(MN,对于神经肌肉功能)类器官,我们使用与阿尔茨海默病(AD)相关的生物标志物评估空间诱导的病理学,额颞叶痴呆(FTD),和肌萎缩侧索硬化症(ALS)。健康和患病的PFC和MN类器官均显示出明显增强的空间神经变性,通过相关疾病生物标志物测量,与他们各自的地球控制相比。第二,我们测试了前两个铅分子,NI112靶向NF-κB和NI113靶向IL-6。我们观察到这些纳米低聚物显著减轻了AD,FTD,和ALS相关的生物标志物,如淀粉样β-42(Aβ42),磷酸化tau(pTau),Kallikrein(KLK-6),焦油DNA结合蛋白43(TDP-43),和其他人。此外,对这些脑类器官的43天纳米低聚物治疗似乎没有在目标类器官组织中引起任何可观察到的毒性或安全性问题,表明在生理相关剂量下,大脑中这些分子具有良好的耐受性。一起,这些结果表明,NI112和NI113分子的开发和翻译具有巨大的潜力,可以作为更安全的太空旅行的潜在神经保护对策,并证明了太空环境对快速,用于临床翻译的靶标和前导分子的高通量筛选。我们断言,在药物开发和筛选中使用微重力可能最终使地球上数百万患有衰弱神经退行性疾病的患者受益。
    The microgravity and space environment has been linked to deficits in neuromuscular and cognitive capabilities, hypothesized to occur due to accelerated aging and neurodegeneration in space. While the specific mechanisms are still being investigated, spaceflight-associated neuropathology is an important health risk to astronauts and space tourists and is being actively investigated for the development of appropriate countermeasures. However, such space-induced neuropathology offers an opportunity for accelerated screening of therapeutic targets and lead molecules for treating neurodegenerative diseases. Here, we show a proof-of-concept high-throughput target screening (on Earth), target validation, and mitigation of microgravity-induced neuropathology using our Nanoligomer platform, onboard the 43-day SpaceX CRS-29 mission to the International Space Station. First, comparing 3D healthy and diseased prefrontal cortex (PFC, for cognition) and motor neuron (MN, for neuromuscular function) organoids, we assessed space-induced pathology using biomarkers relevant to Alzheimer\'s disease (AD), frontotemporal dementia (FTD), and amyotrophic lateral sclerosis (ALS). Both healthy and diseased PFC and MN organoids showed significantly enhanced neurodegeneration in space, as measured through relevant disease biomarkers, when compared to their respective Earth controls. Second, we tested the top two lead molecules, NI112 that targeted NF-κB and NI113 that targeted IL-6. We observed that these Nanoligomers significantly mitigate the AD, FTD, and ALS relevant biomarkers like amyloid beta-42 (Aβ42), phosphorylated tau (pTau), Kallikrein (KLK-6), Tar DNA-binding protein 43 (TDP-43), and others. Moreover, the 43-day Nanoligomer treatment of these brain organoids did not appear to cause any observable toxicity or safety issues in the target organoid tissue, suggesting good tolerability for these molecules in the brain at physiologically relevant doses. Together, these results show significant potential for both the development and translation of NI112 and NI113 molecules as potential neuroprotective countermeasures for safer space travel and demonstrate the usefulness of the space environment for rapid, high-throughput screening of targets and lead molecules for clinical translation. We assert that the use of microgravity in drug development and screening may ultimately benefit millions of patients suffering from debilitating neurodegenerative diseases on Earth.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    2013年,M.Lancaster描述了第一个获得人脑类器官的方案。这些类器官,通常由人类诱导的多能干细胞产生,可以模仿人脑的三维结构。虽然他们概括了人类大脑的显著发育阶段,它们用于研究神经退行性疾病的发病和机制仍然面临着严重的局限性。在这次审查中,我们的目标是强调这些局限性,这阻碍了脑类器官成为研究神经退行性疾病如阿尔茨海默病(AD)的可靠模型,帕金森病(PD),和肌萎缩侧索硬化症(ALS)。具体来说,我们将描述结构和生物障碍,包括缺乏老化的足迹,血管生成,髓鞘形成,以及包含功能和免疫活性小胶质细胞-AD中神经变性发作的所有重要因素,PD,和ALS。此外,我们将讨论监测这些类器官的显微解剖学和电生理学的技术限制。并行,我们将提出解决方案来克服当前的局限性,从而使人脑类器官成为模拟神经变性的更可靠的工具。
    In 2013, M. Lancaster described the first protocol to obtain human brain organoids. These organoids, usually generated from human-induced pluripotent stem cells, can mimic the three-dimensional structure of the human brain. While they recapitulate the salient developmental stages of the human brain, their use to investigate the onset and mechanisms of neurodegenerative diseases still faces crucial limitations. In this review, we aim to highlight these limitations, which hinder brain organoids from becoming reliable models to study neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS). Specifically, we will describe structural and biological impediments, including the lack of an aging footprint, angiogenesis, myelination, and the inclusion of functional and immunocompetent microglia—all important factors in the onset of neurodegeneration in AD, PD, and ALS. Additionally, we will discuss technical limitations for monitoring the microanatomy and electrophysiology of these organoids. In parallel, we will propose solutions to overcome the current limitations, thereby making human brain organoids a more reliable tool to model neurodegeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    癫痫在新生儿至幼儿时期具有高峰发病率。这些早发性癫痫可能是严重的疾病,通常与发育缺陷和智力障碍等合并症相关,在相当比例的患者中,可能是药物抵抗。使用成年啮齿动物模型探索早期癫痫的机制和治疗具有挑战性,因为它忽略了显著的特定年龄的发育差异。最近,在未成熟动物中开发的模型,比如啮齿动物,或在三维类器官中可能更接近地模拟未成熟大脑的各个方面,并可能导致更可翻译的发现。虽然模型并不完美,它们可以在机制和治疗研究中提供更可控的筛查平台,这在儿科患者队列中无法做到。另一方面,需要更简化的模型和更高的通量容量来处理大量的癫痫候选基因和需要新的治疗方案.因此,不同建模方法的组合将有利于解决小儿癫痫患者未满足的需求.在这次审查中,我们总结了在第十六届癫痫神经生物学研讨会上对这一主题的讨论,由国际抗癫痫联盟神经生物学委员会于2022年组织。我们提供了早期发作癫痫的选定模型的概述,讨论它们的优点和缺点。异源表达模型提供了初步的功能见解,和斑马鱼,啮齿动物模型,和脑类器官提出了越来越复杂的平台,用于建模和验证癫痫相关现象。一起,这些模型为早发性癫痫提供了有价值的见解,并加速了假说的产生和治疗发现。
    Epilepsy has a peak incidence during the neonatal to early childhood period. These early onset epilepsies may be severe conditions frequently associated with comorbidities such as developmental deficits and intellectual disability and, in a significant percentage of patients, may be medication-resistant. The use of adult rodent models in the exploration of mechanisms and treatments for early life epilepsies is challenging, as it ignores significant age-specific developmental differences. More recently, models developed in immature animals, such as rodent pups, or in three-dimensional organoids may more closely model aspects of the immature brain and could result in more translatable findings. Although models are not perfect, they may offer a more controlled screening platform in studies of mechanisms and treatments, which cannot be done in pediatric patient cohorts. On the other hand, more simplified models with higher throughput capacities are required to deal with the large number of epilepsy candidate genes and the need for new treatment options. Therefore, a combination of different modeling approaches will be beneficial in addressing the unmet needs of pediatric epilepsy patients. In this review, we summarize the discussions on this topic that occurred during the XVI Workshop on Neurobiology of Epilepsy, organized in 2022 by the Neurobiology Commission of the International League Against Epilepsy. We provide an overview of selected models of early onset epilepsies, discussing their advantages and disadvantages. Heterologous expression models provide initial functional insights, and zebrafish, rodent models, and brain organoids present increasingly complex platforms for modeling and validating epilepsy-related phenomena. Together, these models offer valuable insights into early onset epilepsies and accelerate hypothesis generation and therapy discovery.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    需要一个道德和法律框架来适当地规范快速发展的人脑类器官研究领域。然而,考虑到人类大脑类器官研究中涉及的法律问题仍然不发达和分散。本文回顾了人脑类器官研究的法律问题,将它们分为以下五个广泛的主题:(1)意识,(2)法律地位,(3)同意,(4)所有权,(5)移植。每个主题中的问题都包括紧急(例如,适当的同意形式)和投机性(例如,保护有意识的人脑类器官)。因此,我们试图尽可能明确每个问题的实现时间表,并优先考虑每个问题。研究这些问题揭示了人脑类器官研究特有的法律问题以及其他领域研究中常见的问题。未来需要从法律角度进一步讨论人脑类器官研究,考虑相关领域的讨论。
    An ethical and legal framework is needed to regulate the rapidly developing human brain organoid research field properly. However, considering the legal issues involved in human brain organoid research remains underdeveloped and scattered. This article reviews the legal issues of human brain organoid research, grouping them into the following five broad themes: (1) consciousness, (2) legal status, (3) consent, (4) ownership, and (5) transplantation. The issues in each topic include both the urgent (e.g., appropriate forms of consent) and the speculative (e.g., protection of conscious human brain organoids). Therefore, we have attempted to be as explicit as possible about the timescale within which each issue will be realized and to prioritize each. Examining these issues has revealed legal issues specific to human brain organoid research and issues common to research in other fields. Further discussion of human brain organoid research from a legal perspective is needed in the future, considering discussions in related fields.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    全氟辛烷磺酸(PFOS),在各种环境隔室中检测到的一类合成化学物质,与人类中枢神经系统(CNS)的功能障碍有关。然而,由于缺乏相关的人体模型,全氟辛烷磺酸暴露的潜在神经毒理学在很大程度上没有得到充分研究.这里,我们报告了经过生物工程改造的人类中脑类器官微生理系统(hMO-MPS),以概括胎儿人脑对多种并发PFOS暴露条件的反应.每个hMO-MPS由完全3D打印的支架设备上的hMO组成,该支架设备具有可灌注的类器官粘附层,用于增强气液界面培养。利用独特的,简单制造的支架装置,hMO-MPS是可扩展的,易于使用,与常规孔板兼容,并允许轻松转移到多电极阵列(MEA)系统上,以即插即用地测量神经活动。有趣的是,hMO-MPS的神经活性最初增加,随后由于暴露于浓度范围为0、30、100至300μM的PFOS而降低。此外,全氟辛烷磺酸暴露会损害工程化hMO-MPS的神经发育并促进神经炎症。与全氟辛烷磺酸一起,我们的平台广泛适用于各种其他环境污染物的毒理学研究。
    Perfluorooctane sulfonate (PFOS), a class of synthetic chemicals detected in various environmental compartments, has been associated with dysfunctions of the human central nervous system (CNS). However, the underlying neurotoxicology of PFOS exposure is largely understudied due to the lack of relevant human models. Here, we report bioengineered human midbrain organoid microphysiological systems (hMO-MPSs) to recapitulate the response of a fetal human brain to multiple concurrent PFOS exposure conditions. Each hMO-MPS consists of an hMO on a fully 3D printed holder device with a perfusable organoid adhesion layer for enhancing air-liquid interface culturing. Leveraging the unique, simply-fabricated holder devices, hMO-MPSs are scalable, easy to use, and compatible with conventional well-plates, and allow easy transfer onto a multiple-electrode array (MEA) system for plug-and-play measurement of neural activity. Interestingly, the neural activity of hMO-MPSs initially increased and subsequently decreased by exposure to a concentration range of 0, 30, 100, to 300 μM of PFOS. Furthermore, PFOS exposure impaired neural development and promoted neuroinflammation in the engineered hMO-MPSs. Along with PFOS, our platform is broadly applicable for studies toxicology of various other environmental pollutants.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号