disease modeling

疾病建模
  • 文章类型: Journal Article
    背景:心房颤动的患病率估计为1.5-2%,使其成为最常见的心律失常.导致和维持疾病的过程仍未完全了解。心房颤动和全身性,以及当地的,已经报道了炎症过程。然而,这种关联的确切机制尚未建立。虽然人们理解炎性巨噬细胞可以影响心脏电生理,一个直接的,与心房颤动的因果关系尚未被描述.这项研究调查了激活的M1巨噬细胞对人诱导多能干细胞(hiPSC)衍生的心房心肌细胞的心律失常作用,提出炎症和心房颤动之间的机械联系。
    方法:将来自健康个体的两个hiPSC系分化为心房心肌细胞和M1巨噬细胞,无起搏,心房颤动样共培养模型。分析了共培养物的电生理特征的搏动率不规则性,使用多电极阵列的电描记图幅度和传导速度。使用糖皮质激素额外治疗共培养以抑制M1炎症。对共培养分离的心房心肌细胞进行大量RNA测序,并与心房颤动患者转录组的荟萃分析进行比较。
    结果:多电极阵列记录显示M1引起不规则搏动和电描记图幅度降低。传导分析进一步显示M1共培养物中的传导均匀性显著降低。转录组测序显示关键心脏基因如SCN5A的表达减少,心房心肌细胞中的KCNA5、ATP1A1和GJA5。房颤患者转录组的Meta分析显示与体外模型高度相关。与糖皮质激素共培养的治疗显示表型逆转,包括减少节拍不规则性,改善传导,和逆转的RNA表达谱。
    结论:这项研究建立了M1激活与随后的房性心律失常发展之间的因果关系,记录为与活化巨噬细胞共培养的心房心肌细胞自发电激活的不规则性。Further,使用糖皮质激素可以减轻心率不规则。总的来说,这些结果表明巨噬细胞介导的炎症是一种潜在的AF诱导机制,并为治疗开发提供了新的靶点.这些发现强烈支持了所提出的hiPSC衍生共培养模型的相关性,并将其作为同类疾病模型中的第一个。
    BACKGROUND: Atrial fibrillation has an estimated prevalence of 1.5-2%, making it the most common cardiac arrhythmia. The processes that cause and sustain the disease are still not completely understood. An association between atrial fibrillation and systemic, as well as local, inflammatory processes has been reported. However, the exact mechanisms underlying this association have not been established. While it is understood that inflammatory macrophages can influence cardiac electrophysiology, a direct, causative relationship to atrial fibrillation has not been described. This study investigated the pro-arrhythmic effects of activated M1 macrophages on human induced pluripotent stem cell (hiPSC)-derived atrial cardiomyocytes, to propose a mechanistic link between inflammation and atrial fibrillation.
    METHODS: Two hiPSC lines from healthy individuals were differentiated to atrial cardiomyocytes and M1 macrophages and integrated in an isogenic, pacing-free, atrial fibrillation-like coculture model. Electrophysiology characteristics of cocultures were analysed for beat rate irregularity, electrogram amplitude and conduction velocity using multi electrode arrays. Cocultures were additionally treated using glucocorticoids to suppress M1 inflammation. Bulk RNA sequencing was performed on coculture-isolated atrial cardiomyocytes and compared to meta-analyses of atrial fibrillation patient transcriptomes.
    RESULTS: Multi electrode array recordings revealed M1 to cause irregular beating and reduced electrogram amplitude. Conduction analysis further showed significantly lowered conduction homogeneity in M1 cocultures. Transcriptome sequencing revealed reduced expression of key cardiac genes such as SCN5A, KCNA5, ATP1A1, and GJA5 in the atrial cardiomyocytes. Meta-analysis of atrial fibrillation patient transcriptomes showed high correlation to the in vitro model. Treatment of the coculture with glucocorticoids showed reversal of phenotypes, including reduced beat irregularity, improved conduction, and reversed RNA expression profiles.
    CONCLUSIONS: This study establishes a causal relationship between M1 activation and the development of subsequent atrial arrhythmia, documented as irregularity in spontaneous electrical activation in atrial cardiomyocytes cocultured with activated macrophages. Further, beat rate irregularity could be alleviated using glucocorticoids. Overall, these results point at macrophage-mediated inflammation as a potential AF induction mechanism and offer new targets for therapeutic development. The findings strongly support the relevance of the proposed hiPSC-derived coculture model and present it as a first of its kind disease model.
    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
    动脉粥样硬化,心血管疾病的主要原因,需要先进和创新的建模技术来更好地理解和预测斑块动态。本工作提出了受不同研究领域启发的两个不同的假设模型:混沌理论的逻辑图和随机过程的马尔可夫模型。逻辑图有效地模拟了斑块稳定性的非线性进展和突然变化,反映了动脉粥样硬化事件的混沌性质。相比之下,马尔可夫模型,包括传统的马尔可夫链,空间马尔可夫模型,和马尔可夫随机场,提供了一个概率框架来评估斑块的稳定性和过渡。空间马尔可夫模型,通过热图可视化,突出过渡概率的空间分布,强调本地互动和依赖性。马尔可夫随机场包含复杂的空间相互作用,受到物理学和计算生物学进步的启发,但在参数估计和计算复杂性方面存在挑战。虽然这些假设模型提供了有希望的见解,他们需要严格验证与现实世界的数据,以确认其准确性和适用性。这项研究强调了跨学科方法在开发动脉粥样硬化斑块理论模型中的重要性。
    Atherosclerosis, a leading cause of cardiovascular disease, necessitates advanced and innovative modeling techniques to better understand and predict plaque dynamics. The present work presents two distinct hypothetical models inspired by different research fields: the logistic map from chaos theory and Markov models from stochastic processes. The logistic map effectively models the nonlinear progression and sudden changes in plaque stability, reflecting the chaotic nature of atherosclerotic events. In contrast, Markov models, including traditional Markov chains, spatial Markov models, and Markov random fields, provide a probabilistic framework to assess plaque stability and transitions. Spatial Markov models, visualized through heatmaps, highlight the spatial distribution of transition probabilities, emphasizing local interactions and dependencies. Markov random fields incorporate complex spatial interactions, inspired by advances in physics and computational biology, but present challenges in parameter estimation and computational complexity. While these hypothetical models offer promising insights, they require rigorous validation with real-world data to confirm their accuracy and applicability. This study underscores the importance of interdisciplinary approaches in developing theoretical models for atherosclerotic plaques.
    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
    Cockayne综合征B(CSB)是一种遗传性多器官综合征,通过很大程度上未知的机制,可以影响临床上表现为小头畸形的大脑,智力残疾和脱髓鞘。使用从CSB患者来源和等基因对照系产生的人诱导多能干细胞(hiPSC)来源的神经3D模型,我们在这里为这三种主要的神经病理学表型提供解释。在我们的模型中,CSB缺乏与(i)由于自噬缺陷导致的细胞迁移受损相关,作为临床小头畸形的解释;(ii)改变的神经元网络功能和神经递质GABA水平,这提示干扰的GABA开关可能损害脑回路形成并最终导致智力障碍;和(iii)受损的少突胶质细胞成熟是CSB儿童中观察到的脱髓鞘的可能原因。值得注意的是,受损的迁移和少突胶质细胞成熟都可以通过药理学HDAC抑制部分挽救。
    Cockayne Syndrome B (CSB) is a hereditary multiorgan syndrome which-through largely unknown mechanisms-can affect the brain where it clinically presents with microcephaly, intellectual disability and demyelination. Using human induced pluripotent stem cell (hiPSC)-derived neural 3D models generated from CSB patient-derived and isogenic control lines, we here provide explanations for these three major neuropathological phenotypes. In our models, CSB deficiency is associated with (i) impaired cellular migration due to defective autophagy as an explanation for clinical microcephaly; (ii) altered neuronal network functionality and neurotransmitter GABA levels, which is suggestive of a disturbed GABA switch that likely impairs brain circuit formation and ultimately causes intellectual disability; and (iii) impaired oligodendrocyte maturation as a possible cause of the demyelination observed in children with CSB. Of note, the impaired migration and oligodendrocyte maturation could both be partially rescued by pharmacological HDAC inhibition.
    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
    这项研究试图记录和理解促进者和生产障碍,翻译,在布基纳法索的决策中使用建模证据,尼日利亚,印度,肯尼亚。我们探索了研究人员-决策者参与机制作为证据使用的关键促进者,专注于知识经纪人和边界组织。
    该研究使用了混合方法,该方法借鉴了关键线人访谈和调查的分析,辅之以对地图建模活动和参与者的快速案头审查。这项调查是在网上进行的,而定性研究需要与建模者进行深入访谈,知识经纪人,以及在各种具有代表性的卫生领域工作的决策者,组织,和各级政府。这项研究得到了美国健康媒体实验室IRB(机构审查委员会)和每个研究国家的当地IRB的批准,并于2021年9月至2022年6月进行。
    接受采访的调查人员描述了一系列因素,这些因素促进和抑制了个人在公共卫生决策中使用建模证据,组织,和环境水平。关键主题包括生产能力,翻译,并使用建模证据;建模输出的时间和相关性;建模者和决策者之间存在沟通渠道;基础数据系统的强度;持续资金的作用;以及全球危机的影响。
    这项研究强调了采取生态系统方法来支持建模活动的重要性,考虑到个人,组织,和环境因素,以及不同的参与者如何相互作用来告知生产,翻译,和使用模型证据。促进对话的结构化互动,辩论,证据的生产者和使用者之间的共同意义对于告知和影响决策中证据的使用至关重要。
    UNASSIGNED: This study sought to document and understand facilitators and barriers to producing, translating, and using modeled evidence in decision-making in Burkina Faso, Nigeria, India, and Kenya. We explored researcher-decision-maker engagement mechanisms as key facilitators of evidence use, with a focus on knowledge brokers and boundary organizations.
    UNASSIGNED: The study used mixed methods drawing on analysis from key informant interviews and surveys, complemented by a rapid desk review to map modeling activities and actors. The survey was conducted online while the qualitative research entailed in-depth interviews with modelers, knowledge brokers, and decision-makers working in a representative variety of health fields, organizations, and levels of government. This study was approved by Health Media Lab IRB (Institutional Review Board) in the United States and a local IRB in each study country and conducted between September 2021 and June 2022.
    UNASSIGNED: Informants interviewed for this study described a range of factors that facilitate and inhibit the use of modeled evidence in public health decision-making at the individual, organizational, and environmental levels. Key themes included the capacity to produce, translate, and use modeled evidence; the timing and relevance of modeling outputs; the existence of communications channels between modelers and decision-makers; the strength of underlying data systems; the role of sustained funding; and the impact of global crises.
    UNASSIGNED: This study highlights the importance of taking an ecosystem approach to supporting modeling activities, considering individual, organizational, and environmental factors and how different actors and interact to inform the production, translation, and use of modeled evidence. Structured interaction that promotes dialogue, debate, and joint sense making between the producers and users of evidence is critical to informing and influencing the use of evidence in decision-making.
    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
    人类多能干细胞(hPSC)技术的最新进展促使人类神经元和脑类器官的新研究领域和应用的出现。脑类器官作为一种概括高级结构的体外模型系统而受到关注,细胞多样性和大脑的功能来探索大脑发育,疾病建模,药物筛选,和再生医学。脑类器官技术与各种研究领域的丰富相互作用加速了这一进展。与人脑类器官技术的跨学科方法为更准确地理解人脑提供了更有序的进步。在这次审查中,我们总结了hPSCs在二维和三维培养系统中的神经诱导状态以及使用脑类器官对神经退行性疾病的建模。我们还强调了用于组装空间高阶神经组织的最新生物工程技术,以及脑类器官技术对理解人脑潜力和能力的前景。
    Recent advances in human pluripotent stem cell (hPSC) technologies have prompted the emergence of new research fields and applications for human neurons and brain organoids. Brain organoids have gained attention as an in vitro model system that recapitulates the higher structure, cellular diversity and function of the brain to explore brain development, disease modeling, drug screening, and regenerative medicine. This progress has been accelerated by abundant interactions of brain organoid technology with various research fields. A cross-disciplinary approach with human brain organoid technology offers a higher-ordered advance for more accurately understanding the human brain. In this review, we summarize the status of neural induction in two- and three-dimensional culture systems from hPSCs and the modeling of neurodegenerative diseases using brain organoids. We also highlight the latest bioengineered technologies for the assembly of spatially higher-ordered neural tissues and prospects of brain organoid technology toward the understanding of the potential and abilities of the human brain.
    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
    尽管患者预后有所改善,儿科癌症仍然是儿童非意外死亡的主要原因.最近对儿科癌症患者的遗传分析表明,种系遗传易感性和癌症特异性体细胞驱动突变都具有重要作用。越来越多,有证据表明,起源癌细胞转化的发育时间点对于肿瘤身份和治疗反应至关重要.因此,未来的治疗发展将通过使用忠实地概括遗传背景的疾病模型来支持,细胞起源,和儿童癌症的脆弱性发展窗口。人类干细胞有可能将所有这些特征整合到儿科癌症模型中,同时作为快速遗传和药理测试的平台。在这次审查中,我们描述了人类干细胞是如何用于儿科癌症模型的,以及这些模型与其他儿科癌症模型模式的比较.
    今天,儿童癌症是儿童非意外死亡的主要原因.为了进一步改善结果,对于研究人员和临床医生来说,认识到小儿癌症与成人癌症的区别非常重要。遗传的癌症风险可能在儿科癌症风险中发挥更大的作用,和随后的肿瘤特异性获得性驱动突变启动肿瘤形成。然而,遗传和获得性突变之间存在实质性的相互作用,这支持同时考虑两者。生物技术的最新进展,改善了早期发育细胞和儿科癌细胞之间的匹配,尽管某些儿童中枢神经系统肿瘤的细胞来源仍然难以捉摸。越来越多,证据,特别是在小儿髓母细胞瘤中,证明了癌细胞起源转化的发育时间点对于肿瘤身份和治疗反应至关重要。因此,未来的治疗发展将通过使用忠实地概括遗传背景的疾病模型来支持,细胞起源,和儿科癌症的发育窗口。人类干细胞有可能将所有这些特征整合到儿科癌症模型中,同时作为快速遗传和药理测试的平台。在这次审查中,我们描述了如何使用人类干细胞来模拟儿科癌症,这些模型与其他儿科癌症模型相比,以及未来如何改进这些模型。
    Despite improvements in patient outcomes, pediatric cancer remains a leading cause of non-accidental death in children. Recent genetic analysis of patients with pediatric cancers indicates an important role for both germline genetic predisposition and cancer-specific somatic driver mutations. Increasingly, evidence demonstrates that the developmental timepoint at which the cancer cell-of-origin transforms is critical to tumor identity and therapeutic response. Therefore, future therapeutic development would be bolstered by the use of disease models that faithfully recapitulate the genetic context, cell-of-origin, and developmental window of vulnerability in pediatric cancers. Human stem cells have the potential to incorporate all of these characteristics into a pediatric cancer model, while serving as a platform for rapid genetic and pharmacological testing. In this review, we describe how human stem cells have been used to model pediatric cancers and how these models compare to other pediatric cancer model modalities.
    Today, pediatric cancer is a leading cause of non-accidental death in children. In order to further improve outcomes, it is important for researchers and clinicians alike to recognize how pediatric cancers are distinct from adult cancers. Inherited risk of cancer may play a greater role in pediatric cancer risk, and subsequent tumor-specific acquired driver mutations initiate tumor formation. However, there is substantial interaction between inherited and acquired mutations, which supports consideration of both simultaneously. Recent advancements in biotechnology, have improved matching between early cells of development and pediatric cancer cells, although cell-of-origin for certain pediatric central nervous system tumors remain elusive. Increasingly, evidence, particularly in pediatric medulloblastoma, demonstrates that the developmental timepoint at which the cancer cell-of-origin transforms is critical to tumor identity and therapeutic response. Therefore, future therapeutic development would be bolstered by the use of disease models that faithfully recapitulate the genetic context, cell-of-origin, and developmental window of pediatric cancers. Human stem cells have the potential to incorporate all of these characteristics into a pediatric cancer model, while serving as a platform for rapid genetic and pharmacological testing. In this review, we describe how human stem cells have been used to model pediatric cancers, how human these models compare to other pediatric cancer model modalities, and how these models can be improved in the future.
    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
    自2020年初以来,COVID-19大流行使世界瘫痪,在28个月内导致超过5亿的感染和600多万人死亡。关于这种疾病的知识在很大程度上仍然脱节,特别是当考虑到驱动临床表现和症状多样性的分子机制时。尽管最近有疫苗,仍然迫切需要为严重疾病病例开发有效的治疗方法,尤其是面对新的病毒变种.COVID-19作为一种影响全身独立组织和器官的复杂和多方面的全身性疾病的出现加剧了情况的复杂性。因此,有效治疗策略的开发基于对潜在疾病机制及其与观察到的临床表型多样性的潜在致病联系的综合理解。为了满足这一需求,我们利用计算技术(Dataome平台)对最重要的COVID-19临床表型建立了一个完整的临床分子视图.我们的结果提供了第一个综合,COVID-19症状学的全患者模型,该模型将SARS-CoV-2的分子生命周期与微囊泡介导的细胞间通讯以及接触激活和激肽释放酶激肽系统联系起来。该模型不仅解释了COVID-19的临床多效性,而且为药物开发/再利用和关键风险因素的识别提供了一个证据驱动的框架。相关知识以开源COVID-19Explorer(https://covid19。molecularhealth.com),使全球社会能够探索和分析系统性COVID-19的关键分子特征以及对研究重点和治疗策略的相关影响。我们的工作表明,知识建模解决方案可能在加快全球应对未来卫生紧急情况方面提供重要的效用。
    Since early 2020 the COVID-19 pandemic has paralyzed the world, resulting in more than half a billion infections and over 6 million deaths within a 28-month period. Knowledge about the disease remains largely disjointed, especially when considering the molecular mechanisms driving the diversity of clinical manifestations and symptoms. Despite the recent availability of vaccines, there remains an urgent need to develop effective treatments for cases of severe disease, especially in the face of novel virus variants. The complexity of the situation is exacerbated by the emergence of COVID-19 as a complex and multifaceted systemic disease affecting independent tissues and organs throughout the body. The development of effective treatment strategies is therefore predicated on an integrated understanding of the underlying disease mechanisms and their potentially causative link to the diversity of observed clinical phenotypes. To address this need, we utilized a computational technology (the Dataome platform) to build an integrated clinico-molecular view on the most important COVID-19 clinical phenotypes. Our results provide the first integrated, whole-patient model of COVID-19 symptomatology that connects the molecular lifecycle of SARS-CoV-2 with microvesicle-mediated intercellular communication and the contact activation and kallikrein-kinin systems. The model not only explains the clinical pleiotropy of COVID-19, but also provides an evidence-driven framework for drug development/repurposing and the identification of critical risk factors. The associated knowledge is provided in the form of the open source COVID-19 Explorer (https://covid19.molecularhealth.com), enabling the global community to explore and analyze the key molecular features of systemic COVID-19 and associated implications for research priorities and therapeutic strategies. Our work suggests that knowledge modeling solutions may offer important utility in expediting the global response to future health emergencies.
    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
    离体心脏灌注模型,心血管研究的基本工具,自19世纪末成立以来,已经发生了重大变化。这篇综述追溯了孤立心脏模型的发展,从Langendorff和Starling等先驱的早期改编到Morgan和Neely等研究人员的现代进步。我们讨论了该模型在药理测试中的各种应用,疾病建模,和教育环境,强调其在了解心脏功能和疾病机制方面的关键作用。最近的技术改进,包括高分辨率成像,与生物工程的整合,以及先进的基因组和蛋白质组学分析,大大扩展了这些模型的功能。展望未来,我们探索潜在的未来发展,例如精准医学的整合,干细胞研究,和人工智能,这有望彻底改变孤立心脏灌注模型的使用。这篇综述强调了该模型在桥接实验研究和临床应用中的关键作用。
    The isolated heart perfusion model, a fundamental tool in cardiovascular research, has evolved significantly since its inception in the late 19th century. This review traces the development of the isolated heart model, from its early adaptations by pioneers such as Langendorff and Starling to modern advancements by researchers like Morgan and Neely. We discuss the various applications of the model in pharmacological testing, disease modeling, and educational settings, emphasizing its crucial role in understanding cardiac function and disease mechanisms. Recent technological enhancements, including high-resolution imaging, integration with bioengineering, and advanced genomic and proteomic analyses, have significantly broadened the capabilities of these models. Looking forward, we explore potential future developments such as the integration of precision medicine, stem cell research, and artificial intelligence, which promise to revolutionize the use of isolated heart perfusion models. This review highlights the model\'s crucial role in bridging experimental research and clinical applications.
    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
    基因组编辑是一种在细胞或生物体的DNA中进行特定改变的技术。它极大地改变了生命科学的格局,促进建立极其定制的遗传修饰。在各种基因组编辑技术中,CRISPR/Cas9系统,特定的核酸内切酶诱导双链DNA断裂,并能够对基因组进行修饰,已经成为一种强大而适应性强的工具。它的重要性怎么强调都不为过,因为它不仅允许在模型生物中操纵基因组,而且在医学上具有革命性进步的巨大潜力,特别是在治疗遗传疾病方面。这篇综述论文探讨了CRISPR/Cas9的非凡旅程,它的自然功能,机制,以及对基因组编辑的变革性影响,最后是人工智能和其他智能制造工具的使用。引言提供了基因组编辑的背景,强调CRISPR/Cas9的出现和意义。随后的章节全面阐述了它的自然功能,疾病建模,农业,和生物技术,解决治疗应用,和正在进行的临床试验,同时还讨论前景和伦理影响。我们总结了主要发现,这表明CRISPR/Cas9增强了疾病特异性动物模型的创建能力。这为致病机制提供了宝贵的见解,并为药物发现开辟了新途径,重申CRISPR/Cas9对基因组编辑的变革性影响。最后,我们讨论了继续研究和合作的重要性,以综合利用这种分子精密工具在塑造即将到来的进步中的固有能力。
    Genome editing is a technology to make specific changes in the DNA of a cell or an organism. It has significantly altered the landscape of life sciences, facilitating the establishment of exceedingly customized genetic modifications. Among various genome editing technologies, the CRISPR/Cas9 system, a specific endonuclease induces a double stranded DNA break and enabling modifications to the genome, has surfaced as a formidable and adaptable instrument. Its significance cannot be overstated, as it not only allows for the manipulation of genomes in model organisms but also holds great potential for revolutionary advances in medicine, particularly in treating genetic diseases. This review paper explores the remarkable journey of CRISPR/Cas9, its natural function, mechanisms, and transformative impact on genome editing and finally the use of artificial intelligence and other intelligent manufacturing tools used. The introduction provides the background on genome editing, emphasizing the emergence and significance of CRISPR/Cas9. Subsequent sections comprehensively elucidate its natural function, disease modeling, agriculture, and biotechnology, address therapeutic applications, and ongoing clinical trials while also discussing prospects and ethical implications. We summarized the key findings, indicating that CRISPR/Cas9 has empowered the creation of disease-specific animal models. This provides invaluable insights into pathogenic mechanisms and opens new avenues for drug discovery, reaffirming the transformative impact of CRISPR/Cas9 on genome editing. Finally we discussed the importance of continued research and collaboration for comprehensive utilization of the inherent capabilities of this molecular precision tool in shaping forthcoming advancements.
    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
    常染色体显性视神经萎缩(ADOA)是一种罕见的进行性疾病,主要由OPA1突变引起,OPA1是一种编码线粒体蛋白的核基因,在线粒体动力学中起着至关重要的作用。细胞存活,氧化磷酸化,和mtDNA维护。ADOA的特征在于视网膜神经节细胞(RGCs)的变性。这会导致视力丧失,这在许多情况下可能导致法律失明。如今,ADOA没有有效的治疗方法。在这篇文章中,我们使用iPSC技术和基因组编辑工具CRISPR/Cas9从先前生成的具有致病性变异NM_015560.3:c.1861C>T的ADOA加患者的iPSC细胞系中建立了ADOA的等基因人类RGC模型(p.Gln621Ter)在OPA1中的杂合。为此,已经采用了基于向iPSC培养基补充几种小分子和试图模拟胚胎发育的定义因子的方案。随后,创建的模型经过验证,确认基因组间通信缺陷的存在,线粒体呼吸受损,以及细胞凋亡和ROS产生的增加。最后,我们建议通过qPCR分析OPA1表达作为一种简单的读出方法,用于使用创建的RGC模型进行未来的药物筛选研究。总之,该模型为进一步研究ADOAplus的潜在病理生理机制以及测试具有潜在药理作用的化合物提供了有用的平台.
    Autosomal dominant optic atrophy (ADOA) is a rare progressive disease mainly caused by mutations in OPA1, a nuclear gene encoding for a mitochondrial protein that plays an essential role in mitochondrial dynamics, cell survival, oxidative phosphorylation, and mtDNA maintenance. ADOA is characterized by the degeneration of retinal ganglion cells (RGCs). This causes visual loss, which can lead to legal blindness in many cases. Nowadays, there is no effective treatment for ADOA. In this article, we have established an isogenic human RGC model for ADOA using iPSC technology and the genome editing tool CRISPR/Cas9 from a previously generated iPSC line of an ADOA plus patient harboring the pathogenic variant NM_015560.3: c.1861C>T (p.Gln621Ter) in heterozygosis in OPA1. To this end, a protocol based on supplementing the iPSC culture media with several small molecules and defined factors trying to mimic embryonic development has been employed. Subsequently, the created model was validated, confirming the presence of a defect of intergenomic communication, impaired mitochondrial respiration, and an increase in apoptosis and ROS generation. Finally, we propose the analysis of OPA1 expression by qPCR as an easy read-out method to carry out future drug screening studies using the created RGC model. In summary, this model provides a useful platform for further investigation of the underlying pathophysiological mechanisms of ADOA plus and for testing compounds with potential pharmacological action.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号