disease modeling

疾病建模
  • 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
    背景:膀胱癌(BC)是一种常见的恶性肿瘤,复发率高。患者来源的膀胱癌类器官(BCO)在这两个方面都是一种有希望的方法,疾病建模和个体化治疗筛选。这项研究的目的是研究BCO中转录组的可塑性与培养时间的关系,以定义预期应用的理想时间段。
    方法:本研究包括3例经病理证实的非肌肉浸润性和肌肉浸润性膀胱癌患者的肿瘤样本,并扩展为BCO。使用用于总体转录物表达的差异基因表达和用于病理学相关标记的定量实时PCR(qRT-PCR),在培养的细胞的不同时间段研究RNA表达。
    结果:分别在第1-4代、第5-9代和第9代以上研究了BCO品系的差异基因表达。对各个BCO系的整个转录组的分析揭示了在整个培养和扩增程序中没有显著改变的一致的概况。值得注意的是,关键转录本,如TP53,PIK3CA,BRCA1,其中包括在培养期间的定量RNA分析中表现出稳定的表达水平。
    结论:BCO培养过程中强大的转录组主张在个性化医疗中使用早期的BCOs,提供了一种时间有效的药物筛选选择,以加快患者治疗方案的咨询。较高的BCOs通道在需要扩大细胞团的主题中仍然具有潜力,例如医疗设备开发等。
    BACKGROUND: Bladder cancer (BC) is a prevalent malignancy with high recurrence rates. Patient-derived bladder cancer organoids (BCO) pose as a promising approach in both, disease modeling and individualized treatment screening. The aim of this study was to investigate the transcriptomic plasticity in BCOs as a function of cultivation times to define ideal time periods for the applications envisioned.
    METHODS: Tumor samples of three patients with pathologically confirmed non-muscle invasive and muscle-invasive bladder cancer were included in this study and expanded as BCOs. RNA expression was investigated at different time periods of cells in culture using differential gene expression for overall transcript expression and quantitative real-time PCR (qRT-PCR) for pathological relevant markers.
    RESULTS: Differential gene expression of the BCO lines was investigated across passages 1-4, in passages 5-9 and above 9, respectively. Analysis of the entire transcriptome of the respective BCO lines revealed consistent profiles without significant alterations throughout the cultivation and expansion procedure. Notably, key transcripts like TP53, PIK3CA, BRCA1, among others, exhibited stable expression levels in the quantitative RNA analysis during the cultivation period.
    CONCLUSIONS: The robust transcriptome during BCO cultivation advocates for the use of earlier passages of BCOs in personalized medicine providing a time-efficient drug screening option to accelerate the counseling of patients\' treatment options. Higher passages of BCOs still hold the potential in topics demanding for expanded cell masses such as medical device development and others.
    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
    Leigh综合征(LS),复杂的多系统疾病,由于其复杂的发病机制和广泛的临床表现,在遗传医学中提出了重大挑战。值得注意的是,这些来自核遗传DNA或线粒体DNA的突变,影响ATP的产生并导致不同的临床结果。这种疾病的不可预测的轨迹,从严重的发育迟缓到早期死亡,强调需要改进的治疗解决方案。这项研究转向了诱导多能干细胞(iPSC)的新用途,作为了解疾病机制和引领患者特异性药物发现的有希望的平台。鉴于iPSCs过去在描述器官特异性疾病方面的成功,以及FDA最近对人类iPSC衍生心肌细胞(CMs)进行药物评估的认可,我们的工作旨在将这种创新与Leigh综合征研究联系起来。WedetailamethodologyapproachtogenerateiPSCsfromLSpatientsanddifferentialthemintoiPSCs-CM.Usingmulti-electrodearray(MEA)analysis,我们评估这些细胞的场电位,突出hiPSC-CM在药物验证和疾病建模中的潜力。这种开创性的方法为Leigh/Leigh样综合征以患者为中心的治疗干预措施的未来提供了一瞥。
    Leigh syndrome (LS), a complex multisystemic disorder, poses significant challenges in genetic medicine due to its intricate pathogenesis and wide-ranging clinical manifestations. Notably, these arise from mutations in either nuclear genetic DNA or mitochondrial DNA, affecting ATP production and resulting in diverse clinical outcomes. The unpredictable trajectory of this disease, ranging from severe developmental delays to early mortality, underscores the need for improved therapeutic solutions. This research pivots toward the novel use of induced pluripotent stem cells (iPSCs) as a promising platform for understanding disease mechanisms and spearheading patient-specific drug discoveries. Given the past successes of iPSCs in delineating organ-specific disorders and the recent endorsement of human iPSC-derived cardiomyocytes (CMs) by the FDA for drug evaluation, our work seeks to bridge this innovation to Leigh syndrome research. We detail a methodological approach to generate iPSCs from LS patients and differentiate them into iPSCs-CMs. Using multi-electrode array (MEA) analyses, we evaluate the field potential of these cells, spotlighting the potential of hiPSC-CM in drug validation and disease modeling. This pioneering approach offers a glimpse into the future of patient-centric therapeutic interventions for Leigh/Leigh-like syndrome.
    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
    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
    背景:最近针对β淀粉样蛋白(Aβ)靶向治疗阿尔茨海默病(AD)的临床试验已证明其临床获益超过18个月,但它们对疾病轨迹的长期影响尚不清楚。我们提出了一个评估现实长期情景的框架。
    方法:将Aβ靶向抗体的最近3期试验的结果与临床痴呆评分-箱和(CDR-SB)评分的长期患者水平自然史轨迹的估计相结合,以探索现实的长期疗效方案。
    结果:检查了三种不同的长期疗效方案,从保守到乐观。这些阳性3期试验的推断表明,治疗可将严重痴呆的发作延迟0.3至0.6年(保守)。1.1至1.9年(中间),2.0到4.2年(乐观)。
    结论:我们的研究为疾病改善治疗的长期影响提供了一种通用语言。我们的工作需要更长时间的随访研究和早期干预试验的结果,以全面评估这些疗法的真正长期影响。
    结论:我们提出了AD治疗疗效的长期方案。在这个框架中,场景是相对于AD的自然史定义的。可以比较不同乐观程度的长期预测。它为表达对长期疗效的信念提供了一种通用语言。
    BACKGROUND: Recent clinical trials of amyloid beta (Aβ)-targeting therapies in Alzheimer\'s disease (AD) have demonstrated a clinical benefit over 18 months, but their long-term impact on disease trajectory is not yet understood. We propose a framework for evaluating realistic long-term scenarios.
    METHODS: Results from recent phase 3 trials of Aβ-targeting antibodies were integrated with an estimate of the long-term patient-level natural history trajectory of the Clinical Dementia Rating-Sum of Boxes (CDR-SB) score to explore realistic long-term efficacy scenarios.
    RESULTS: Three distinct long-term efficacy scenarios were examined, ranging from conservative to optimistic. These extrapolations of positive phase 3 trials suggested treatments delayed onset of severe dementia by 0.3 to 0.6 years (conservative), 1.1 to 1.9 years (intermediate), and 2.0 to 4.2 years (optimistic).
    CONCLUSIONS: Our study provides a common language for long-term impact of disease-modifying treatments. Our work calls for studies with longer follow-up and results from early intervention trials to provide a comprehensive assessment of these therapies\' true long-term impact.
    CONCLUSIONS: We present long-term scenarios of the efficacy of AD therapies. In this framework, scenarios are defined relative to the natural history of AD. Long-term projections with different levels of optimism can be compared. It provides a common language for expressing beliefs about long-term efficacy.
    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
    背景:帕金森病(PD)是第二常见的神经退行性疾病,在阿尔茨海默氏症之后。其特征在于α-突触核蛋白在脑中聚集成路易体和路易神经突。小胶质细胞驱动的神经炎症可能导致PD中的神经元死亡,然而,小胶质细胞的确切作用尚不清楚,且研究不足.编码α-突触核蛋白的基因中的A53T突变与早发性PD有关,暴露于A53T突变的人α-突触核蛋白会增加鼠类小胶质细胞炎症的可能性。迄今为止,尚未在人类小胶质细胞中研究其作用。
    方法:这里,我们使用人iPSC来源的小胶质细胞的二维培养物,并将这些细胞移植到小鼠脑中,以评估A53T突变对人小胶质细胞的细胞自主效应.
    结果:我们发现A53T突变的人类小胶质细胞在炎症刺激后具有内在增加的促炎激活倾向。此外,移植的A53T突变小胶质细胞在非炎症条件下显示过氧化氢酶表达的强烈下降,和增加的氧化应激。
    结论:我们的结果表明,A53T突变人小胶质细胞表现出细胞自主表型,可能会在早发性PD中加重神经元损伤。
    BACKGROUND: Parkinson\'s disease (PD) is the second most common neurodegenerative disease, following Alzheimer\'s. It is characterized by the aggregation of α-synuclein into Lewy bodies and Lewy neurites in the brain. Microglia-driven neuroinflammation may contribute to neuronal death in PD, however the exact role of microglia remains unclear and has been understudied. The A53T mutation in the gene coding for α-synuclein has been linked to early-onset PD, and exposure to A53T-mutant human α-synuclein increases the potential for inflammation of murine microglia. To date, its effect has not been studied in human microglia.
    METHODS: Here, we used 2-dimensional cultures of human iPSC-derived microglia and transplantation of these cells into the mouse brain to assess the cell-autonomous effects of the A53T mutation on human microglia.
    RESULTS: We found that A53T-mutant human microglia had an intrinsically increased propensity towards pro-inflammatory activation upon inflammatory stimulus. Additionally, transplanted A53T mutant microglia showed a strong decrease in catalase expression in non-inflammatory conditions, and increased oxidative stress.
    CONCLUSIONS: Our results indicate that A53T mutant human microglia display cell-autonomous phenotypes that may worsen neuronal damage in early-onset PD.
    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
    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
    在LZTR1中具有致病变异的Noonan综合征患者尤其有发展为严重和早发性肥厚型心肌病的风险。在这项研究中,我们通过使用患者特异性和CRISPR-Cas9校正的诱导多能干细胞(iPSC)心肌细胞,研究了纯合变体LZTR1L580P的机制后果.分子,细胞,功能表型与计算机预测相结合,可确定引起心脏肥大的LZTR1L580P特异性疾病机制。预测变体改变二聚化结构域的结合亲和力,促进线性LZTR1聚合物的形成。LZTR1复杂功能障碍导致RASGTPases的积累,从而引起蛋白质组景观的整体病理变化,最终导致细胞肥大。此外,我们的数据表明,心肌细胞特异性MRAS降解是由LZTR1通过非蛋白酶体途径介导的,而RIT1降解是由LZTR1依赖性和LZTR1非依赖性途径介导的。LZTR1L580P错义变异的单等位基因或双等位基因基因校正挽救了分子和细胞疾病表型,为基于CRISPR的疗法提供概念证明。
    Noonan syndrome patients harboring causative variants in LZTR1 are particularly at risk to develop severe and early-onset hypertrophic cardiomyopathy. In this study, we investigate the mechanistic consequences of a homozygous variant LZTR1L580P by using patient-specific and CRISPR-Cas9-corrected induced pluripotent stem cell (iPSC) cardiomyocytes. Molecular, cellular, and functional phenotyping in combination with in silico prediction identify an LZTR1L580P-specific disease mechanism provoking cardiac hypertrophy. The variant is predicted to alter the binding affinity of the dimerization domains facilitating the formation of linear LZTR1 polymers. LZTR1 complex dysfunction results in the accumulation of RAS GTPases, thereby provoking global pathological changes of the proteomic landscape ultimately leading to cellular hypertrophy. Furthermore, our data show that cardiomyocyte-specific MRAS degradation is mediated by LZTR1 via non-proteasomal pathways, whereas RIT1 degradation is mediated by both LZTR1-dependent and LZTR1-independent pathways. Uni- or biallelic genetic correction of the LZTR1L580P missense variant rescues the molecular and cellular disease phenotype, providing proof of concept for CRISPR-based therapies.
    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
    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