iPSC

iPSC
  • 文章类型: Journal Article
    初级纤毛是指状感觉器官,从大多数细胞类型的身体延伸出来,并具有与质膜不同的脂质和蛋白质组成。这种分配是由限制非纤毛蛋白进入的扩散屏障维持的,并允许选择性进入具有睫状靶向序列(CTS)的蛋白质。然而,CTSs不是定型的,并且先前报道的序列不足以驱动跨不同细胞类型的有效纤毛定位。这里,我们描述了一个短的肽序列,有效地将跨膜蛋白靶向所有测试细胞类型的初级纤毛,包括人类神经元。我们产生了稳定表达跨膜构建体的人诱导多能干细胞(hiPSC)系,该构建体带有细胞外HaloTag和胞内荧光蛋白,这使得光明,神经元和其他细胞类型中初级纤毛的特异性标记,以促进纤毛在健康和疾病中的研究。我们通过开发用于自动测量初级纤毛的图像分析管道来检测与信号传导或疾病状态改变相关的长度变化,从而证明了该资源的实用性。
    Primary cilia are finger-like sensory organelles that extend from the bodies of most cell types and have a distinct lipid and protein composition from the plasma membrane. This partitioning is maintained by a diffusion barrier that restricts the entry of non-ciliary proteins, and allows the selective entry of proteins harboring a ciliary targeting sequence (CTS). However, CTSs are not stereotyped and previously reported sequences are insufficient to drive efficient ciliary localisation across diverse cell types. Here, we describe a short peptide sequence that efficiently targets transmembrane proteins to primary cilia in all tested cell types, including human neurons. We generate human-induced pluripotent stem cell (hiPSC) lines stably expressing a transmembrane construct bearing an extracellular HaloTag and intracellular fluorescent protein, which enables the bright, specific labeling of primary cilia in neurons and other cell types to facilitate studies of cilia in health and disease. We demonstrate the utility of this resource by developing an image analysis pipeline for the automated measurement of primary cilia to detect changes in their length associated with altered signaling or disease state.
    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
    过度的炎症反应和氧化应激是自闭症中公认的分子发现,这些过程可能会影响表观遗传景观或受其影响。尽管如此,没有足够的治疗方法,因为用于个体化治疗的患者特异性脑分子标志物仍然具有挑战性。
    方法:我们使用了自闭症患者的iPSC来源的神经元和星形胶质细胞对照组(5/组),以检查他们是否复制了自闭症的死后脑表达/表观遗传学改变。此外,分析10个死后脑样品(5个/组)的DNA甲基化的PSC衍生细胞中受影响的基因。
    结果:我们发现TGFB1,TGFB2,IL6和IFI16的过度表达和HAP1,SIRT1,NURR1,RELN,孤独症患者星形胶质细胞中的GPX1,EN2,SLC1A2和SLC1A3,随着TGFB2,IL6,TNFA和EN2基因启动子的DNA低甲基化以及自闭症患者星形胶质细胞中HAP1启动子5-羟甲基化的减少。在神经元中,HAP1和IL6表达趋势相似。虽然HAP1启动子在神经元中高度甲基化,IFI16和SLC1A3启动子被低甲基化,并且TGFB2表现出增加的启动子5-羟基甲基化。我们还发现神经元乔化减少,脊柱尺寸,增长率,和移民,但是自闭症患者的星形胶质细胞大小增加,生长速度降低。在死后的大脑样本中,我们发现TGFB2和IFI16启动子区的DNA低甲基化,但自闭症中HAP1和SLC1A2启动子的DNA甲基化。
    结论:iPSC来源的细胞中自闭症相关的表达/表观遗传学改变复制了文献中报道的那些,使它们成为研究疾病发病机理或患者特异性疗法的适当替代品。
    Excessive inflammatory reactions and oxidative stress are well-recognized molecular findings in autism and these processes can affect or be affected by the epigenetic landscape. Nonetheless, adequate therapeutics are unavailable, as patient-specific brain molecular markers for individualized therapies remain challenging.
    METHODS: We used iPSC-derived neurons and astrocytes of patients with autism vs. controls (5/group) to examine whether they replicate the postmortem brain expression/epigenetic alterations of autism. Additionally, DNA methylation of 10 postmortem brain samples (5/group) was analyzed for genes affected in PSC-derived cells.
    RESULTS: We found hyperexpression of TGFB1, TGFB2, IL6 and IFI16 and decreased expression of HAP1, SIRT1, NURR1, RELN, GPX1, EN2, SLC1A2 and SLC1A3 in the astrocytes of patients with autism, along with DNA hypomethylation of TGFB2, IL6, TNFA and EN2 gene promoters and a decrease in HAP1 promoter 5-hydroxymethylation in the astrocytes of patients with autism. In neurons, HAP1 and IL6 expression trended alike. While HAP1 promoter was hypermethylated in neurons, IFI16 and SLC1A3 promoters were hypomethylated and TGFB2 exhibited increased promoter 5-hydroxymethlation. We also found a reduction in neuronal arborization, spine size, growth rate, and migration, but increased astrocyte size and a reduced growth rate in autism. In postmortem brain samples, we found DNA hypomethylation of TGFB2 and IFI16 promoter regions, but DNA hypermethylation of HAP1 and SLC1A2 promoters in autism.
    CONCLUSIONS: Autism-associated expression/epigenetic alterations in iPSC-derived cells replicated those reported in the literature, making them appropriate surrogates to study disease pathogenesis or patient-specific therapeutics.
    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
    称为生物素-硫胺素反应性基底神经节病(BTBGD)的神经代谢紊乱是一种罕见的常染色体隐性遗传病,与SLC19A3基因的双等位基因致病突变有关。BTBGD的特点是进行性脑病,混乱,癫痫发作,构音障碍,肌张力障碍,严重残疾。由于该病的罕见性质和不同的临床特征,诊断困难。此时BTBGD的主要治疗方法是补充硫胺素和生物素,而其长期有效性仍在调查中。在这项研究中,我们已经从一名10岁的女性BTBGD患者中产生了两个诱导多能干细胞(iPSCs)克隆,该患者携带SLC19A3基因外显子5的致病变异的纯合突变,c.1264A>G(p。Thr422Ala)。我们已经证实了所产生的iPS系的多能性,并成功地将它们分化为神经祖细胞。因为我们对BTBGD中基因型-表型相关性的理解是有限的,具有纯合SLC19A3突变的BTBGD-iPSC系的建立为探索SLC19A3相关细胞功能障碍的分子机制提供了有价值的细胞模型.该模型具有促进新型治疗策略发展的潜力。
    The neurometabolic disorder known as biotin-thiamine-responsive basal ganglia disease (BTBGD) is a rare autosomal recessive condition linked to bi-allelic pathogenic mutations in the SLC19A3 gene. BTBGD is characterized by progressive encephalopathy, confusion, seizures, dysarthria, dystonia, and severe disabilities. Diagnosis is difficult due to the disease\'s rare nature and diverse clinical characteristics. The primary treatment for BTBGD at this time is thiamine and biotin supplementation, while its long-term effectiveness is still being investigated. In this study, we have generated two clones of induced pluripotent stem cells (iPSCs) from a 10-year-old female BTBGD patient carrying a homozygous mutation for the pathogenic variant in exon 5 of the SLC19A3 gene, c.1264A > G (p.Thr422Ala). We have confirmed the pluripotency of the generated iPS lines and successfully differentiated them to neural progenitors. Because our understanding of genotype-phenotype correlations in BTBGD is limited, the establishment of BTBGD-iPSC lines with a homozygous SLC19A3 mutation provides a valuable cellular model to explore the molecular mechanisms underlying SLC19A3-associated cellular dysfunction. This model holds potential for advancing the development of novel therapeutic strategies.
    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:哺乳动物脑中神经干细胞(NSC)的增殖和分化在出生后降低到最低水平。然而,在啮齿动物的成年皮质和海马中存在神经源性生态位,灵长类动物和人类,成人NSC分化与发展共享关键的监管机制。成人神经发生损伤与阿尔茨海默病(AD)病理有关。通过使用神经营养因子来解决这些损伤是基于神经发生的治疗干预的有希望的新途径。然而,使用体内模型进行筛查的技术困难阻碍了这种可能性,包括使用成人大脑中稀缺的神经干细胞以及人类和小鼠模型之间的差异或伦理限制。
    方法:这里,我们使用小鼠和人类干细胞模型的组合,对一种新的神经源性化合物进行全面的体外表征,关注脑源性神经营养因子(BDNF)通路。ENT-A011,一种甾体脱氢表雄酮衍生物,通过蛋白质印迹在NIH-3T3细胞中测试酪氨酸受体激酶B(TrkB)受体的激活,并通过增殖评估其神经源性和神经保护作用,小鼠原代成年海马神经干细胞的细胞死亡和淀粉样β(Aβ)毒性测定,小鼠胚胎皮质神经干细胞和神经祖细胞(NPC)从三个人诱导多能干细胞系从健康和AD供体分化。RNA-seq谱分析用于评估化合物是否通过与人NPC中的BDNF相同的基因网络起作用。
    结果:ENT-A011能够增加小鼠原代成年海马神经干细胞和胚胎皮质神经干细胞的增殖,在没有EGF/FGF的情况下,同时减少Aβ诱导的细胞死亡,有选择地通过TrkB激活。该化合物能够增加参与NSC维持的星形细胞基因标记,保护海马神经元免受Αβ毒性并防止Aβ治疗后的突触丢失。ENT-A011成功诱导增殖并防止人NPC中Aβ毒性后的细胞死亡,通过与BDNF共享的核心基因网络起作用,如通过RNA-seq所示。
    结论:我们的工作表征了一种新型BDNF模拟物,通过基于干细胞的筛选,在阿尔茨海默病中具有较好的药理特性和神经源性和神经保护作用。证明干细胞系统有望入围竞争候选人进行进一步测试。
    BACKGROUND: Neural stem cell (NSC) proliferation and differentiation in the mammalian brain decreases to minimal levels postnatally. Nevertheless, neurogenic niches persist in the adult cortex and hippocampus in rodents, primates and humans, with adult NSC differentiation sharing key regulatory mechanisms with development. Adult neurogenesis impairments have been linked to Alzheimer\'s disease (AD) pathology. Addressing these impairments by using neurotrophic factors is a promising new avenue for therapeutic intervention based on neurogenesis. However, this possibility has been hindered by technical difficulties of using in-vivo models to conduct screens, including working with scarce NSCs in the adult brain and differences between human and mouse models or ethical limitations.
    METHODS: Here, we use a combination of mouse and human stem cell models for comprehensive in-vitro characterization of a novel neurogenic compound, focusing on the brain-derived neurotrophic factor (BDNF) pathway. The ability of ENT-A011, a steroidal dehydroepiandrosterone derivative, to activate the tyrosine receptor kinase B (TrkB) receptor was tested through western blotting in NIH-3T3 cells and its neurogenic and neuroprotective action were assessed through proliferation, cell death and Amyloid-β (Aβ) toxicity assays in mouse primary adult hippocampal NSCs, mouse embryonic cortical NSCs and neural progenitor cells (NPCs) differentiated from three human induced pluripotent stem cell lines from healthy and AD donors. RNA-seq profiling was used to assess if the compound acts through the same gene network as BDNF in human NPCs.
    RESULTS: ENT-A011 was able to increase proliferation of mouse primary adult hippocampal NSCs and embryonic cortical NSCs, in the absence of EGF/FGF, while reducing Aβ-induced cell death, acting selectively through TrkB activation. The compound was able to increase astrocytic gene markers involved in NSC maintenance, protect hippocampal neurons from Αβ toxicity and prevent synapse loss after Aβ treatment. ENT-A011 successfully induces proliferation and prevents cell death after Aβ toxicity in human NPCs, acting through a core gene network shared with BDNF as shown through RNA-seq.
    CONCLUSIONS: Our work characterizes a novel BDNF mimetic with preferable pharmacological properties and neurogenic and neuroprotective actions in Alzheimer\'s disease via stem cell-based screening, demonstrating the promise of stem cell systems for short-listing competitive candidates for further testing.
    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:遗传性视网膜营养不良(IRD)是全球范围内无法治愈的失明的主要原因之一。IRD是由编码视网膜必需蛋白的基因突变引起的,导致光感受器退化和视觉功能丧失。由于缺乏对其病理生理学的重要部分的了解,IRD产生了巨大的全球财务负担,分子诊断,以及几乎没有非姑息治疗方案。用于IRD的患者来源的诱导多能干细胞(iPSC)似乎是解决这些问题的绝佳选择,作为IRD病理生理学深入研究和测试新治疗方法的特殊工具。
    方法:从8名与PROM1相关的IRD患者的队列中,我们确定了3名患者携带相同的变体(c.1354dupT),但表达三种不同的IRD表型:锥形和杆状营养不良(CORD),色素性视网膜炎(RP),和Stargardt病4型(STGD4)。这三个目标患者,每个人都有一个健康的亲戚,接受了全面的眼科检查,并通过临床外显子组测序(CES)扩展了他们的遗传小组研究。随后,产生非整合性患者来源的iPSC,并对其进行充分表征.使用CRISPR/Cas9进行c.1354dupT突变的校正,并且在患者来源的iPSC系中通过流式细胞术和蛋白质印迹确认PROM1基因的遗传恢复。
    结果:CES显示,2名具有c.1354dupT突变的目标患者在与补体系统或光感受器分化和过氧化物酶体生物发生障碍相关的基因中呈现单等位基因变异,分别。证实了患者来源的iPSC细胞系的多能性和功能性,目标突变的校正完全恢复了基因修复的患者来源的iPSC系中编码Prominin-1(CD133)的能力。
    结论:PROM1基因的c.1354dupT突变与IRD的三种不同的AR表型相关。这种多向效应可能与视网膜营养不良相关的其他基因中单等位基因变体的影响有关。然而,需要提供进一步的证据。未来的实验应该包括基因编辑的患者来源的iPSC,因为它有潜力作为疾病建模工具来阐明这一问题。
    BACKGROUND: Inherited retinal dystrophies (IRD) are one of the main causes of incurable blindness worldwide. IRD are caused by mutations in genes that encode essential proteins for the retina, leading to photoreceptor degeneration and loss of visual function. IRD generates an enormous global financial burden due to the lack of understanding of a significant part of its pathophysiology, molecular diagnosis, and the near absence of non-palliative treatment options. Patient-derived induced pluripotent stem cells (iPSC) for IRD seem to be an excellent option for addressing these questions, serving as exceptional tools for in-depth studies of IRD pathophysiology and testing new therapeutic approaches.
    METHODS: From a cohort of 8 patients with PROM1-related IRD, we identified 3 patients carrying the same variant (c.1354dupT) but expressing three different IRD phenotypes: Cone and rod dystrophy (CORD), Retinitis pigmentosa (RP), and Stargardt disease type 4 (STGD4). These three target patients, along with one healthy relative from each, underwent comprehensive ophthalmic examinations and their genetic panel study was expanded through clinical exome sequencing (CES). Subsequently, non-integrative patient-derived iPSC were generated and fully characterized. Correction of the c.1354dupT mutation was performed using CRISPR/Cas9, and the genetic restoration of the PROM1 gene was confirmed through flow cytometry and western blotting in the patient-derived iPSC lines.
    RESULTS: CES revealed that 2 target patients with the c.1354dupT mutation presented monoallelic variants in genes associated with the complement system or photoreceptor differentiation and peroxisome biogenesis disorders, respectively. The pluripotency and functionality of the patient-derived iPSC lines were confirmed, and the correction of the target mutation fully restored the capability of encoding Prominin-1 (CD133) in the genetically repaired patient-derived iPSC lines.
    CONCLUSIONS: The c.1354dupT mutation in the PROM1 gene is associated to three distinct AR phenotypes of IRD. This pleotropic effect might be related to the influence of monoallelic variants in other genes associated with retinal dystrophies. However, further evidence needs to be provided. Future experiments should include gene-edited patient-derived iPSC due to its potential as disease modelling tools to elucidate this matter in question.
    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:干细胞衍生疗法具有治疗再生临床适应症的潜力。静态培养具有有限的放大能力,因此限制了其使用。悬浮培养可用于大量生产靶细胞,但也提出了与压力和聚集稳定性相关的挑战。
    方法:在垂直轮式生物反应器中利用实验设计(DoE)方法,我们评估了具有多种特性的介质添加剂。评估的添加剂是肝素钠盐(HS),聚乙二醇(PEG),聚乙烯醇(PVA),PluronicF68和硫酸葡聚糖(DS)。选择多个响应变量来评估细胞生长,响应于添加剂输入的多能性维持和聚集体稳定性,和数学模型被生成和调整为最大预测能力。
    结果:在19种不同的培养基组合上使用100ml立式轮生物反应器测定4天扩增iPSC,从而产生可以优化多能性的模型,稳定性,和扩张。扩展优化导致了PA的组合,PVA和PEG与E8。该混合物导致比单独的E8短40%的膨胀倍增时间。多能性优化器强调了向E8培养基中添加1%PEG的重要性。使3D培养中的聚集体融合最小化的聚集体稳定性优化表明肝素和PEG两者的相互作用可以限制聚集以及增加hiPSC的维持能力和扩增。表明控制融合是扩展和维护的关键参数。在生物反应器中以40RPM的速度降低的两种细胞系上验证优化的解决方案,显示了对具有高频率的OCT4和S0X2的多能性标记物(>90%)的聚集体的一致性和延长的控制。在优化培养基中作为团块传代后,维持约1-1.4天的倍增时间。控制聚集体融合允许生物反应器速度降低,因此在大规模扩增中施加在细胞上的剪切应力降低。
    结论:本研究控制了悬浮培养物中的聚集体大小,同时告知iPSC状态的伴随状态控制。这种方法的更广泛的应用可以解决培养基优化复杂性和生物反应器放大的挑战。
    BACKGROUND: Stem cell-derived therapies hold the potential for treatment of regenerative clinical indications. Static culture has a limited ability to scale up thus restricting its use. Suspension culturing can be used to produce target cells in large quantities, but also presents challenges related to stress and aggregation stability.
    METHODS: Utilizing a design of experiments (DoE) approach in vertical wheel bioreactors, we evaluated media additives that have versatile properties. The additives evaluated are Heparin sodium salt (HS), polyethylene glycol (PEG), poly (vinyl alcohol) (PVA), Pluronic F68 and dextran sulfate (DS). Multiple response variables were chosen to assess cell growth, pluripotency maintenance and aggregate stability in response to the additive inputs, and mathematical models were generated and tuned for maximal predictive power.
    RESULTS: Expansion of iPSCs using 100 ml vertical wheel bioreactor assay for 4 days on 19 different media combinations resulted in models that can optimize pluripotency, stability, and expansion. The expansion optimization resulted in the combination of PA, PVA and PEG with E8. This mixture resulted in an expansion doubling time that was 40% shorter than that of E8 alone. Pluripotency optimizer highlighted the importance of adding 1% PEG to the E8 medium. Aggregate stability optimization that minimizes aggregate fusion in 3D culture indicated that the interaction of both Heparin and PEG can limit aggregation as well as increase the maintenance capacity and expansion of hiPSCs, suggesting that controlling fusion is a critical parameter for expansion and maintenance. Validation of optimized solution on two cell lines in bioreactors with decreased speed of 40 RPM, showed consistency and prolonged control over aggregates that have high frequency of pluripotency markers of OCT4 and SOX2 (> 90%). A doubling time of around 1-1.4 days was maintained after passaging as clumps in the optimized medium. Controlling aggregate fusion allowed for a decrease in bioreactor speed and therefore shear stress exerted on the cells in a large-scale expansion.
    CONCLUSIONS: This study resulted in a control of aggregate size within suspension cultures, while informing about concomitant state control of the iPSC state. Wider application of this approach can address media optimization complexity and bioreactor scale-up challenges.
    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
    听觉器官发育异常,耳蜗,大约四分之一的先天性耳聋患者被诊断出。由于对潜在基因的知识不足或无法对已确定的遗传变异做出结论性解释,大多数耳蜗畸形患者在病因上仍未被诊断。我们使用外显子组测序技术对来自无关家庭的三个先证者中与耳蜗畸形相关的听力损失进行了遗传评估。我们随后产生了单克隆诱导多能干细胞(iPSC)系,使用CRISPR/Cas9进行患者特异性敲入和敲除以评估候选变体的致病性。我们检测到FGF3(p。Arg165Gly)和GREB1L(p。Cys186Arg),在两个公认的耳聋基因中具有不确定意义的变异,和PBXIP1(p。Trp574*)中的一个候选基因。iPSCs向内耳类器官分化后,与同基因对照相比,我们在敲除品系中观察到显着的发育异常。患者特异性单核苷酸变体(SNV)显示出与敲除系相似的异常,在功能上支持它们在观察到的表型中的因果关系。因此,我们提出了人类内耳类器官作为快速验证与耳蜗畸形相关的DNA变异的致病性的工具。
    Developmental anomalies of the hearing organ, the cochlea, are diagnosed in approximately one-fourth of individuals with congenital deafness. Most patients with cochlear malformations remain etiologically undiagnosed due to insufficient knowledge about underlying genes or the inability to make conclusive interpretations of identified genetic variants. We used exome sequencing for genetic evaluation of hearing loss associated with cochlear malformations in three probands from unrelated families. We subsequently generated monoclonal induced pluripotent stem cell (iPSC) lines, bearing patient-specific knockins and knockouts using CRISPR/Cas9 to assess pathogenicity of candidate variants. We detected FGF3 (p.Arg165Gly) and GREB1L (p.Cys186Arg), variants of uncertain significance in two recognized genes for deafness, and PBXIP1(p.Trp574*) in a candidate gene. Upon differentiation of iPSCs towards inner ear organoids, we observed significant developmental aberrations in knockout lines compared to their isogenic controls. Patient-specific single nucleotide variants (SNVs) showed similar abnormalities as the knockout lines, functionally supporting their causality in the observed phenotype. Therefore, we present human inner ear organoids as a tool to rapidly validate the pathogenicity of DNA variants associated with cochlear malformations.
    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
    GGGGCC(G4C2)在C9ORF72中重复扩增是肌萎缩侧索硬化症(ALS)和额颞叶痴呆(FTD)的最常见遗传原因。这种基因突变如何导致神经变性仍然是未知的。使用CRISPR-Cas9技术,我们删除了EXOC2,它编码一个必需的外囊亚基,来自C9ORF72-ALS/FTD患者的诱导多能干细胞(iPSC)。由于截短的EXOC2的存在,这些细胞是活的,表明外囊功能被部分维持。C9ORF72iPSC衍生的运动神经元中几种疾病相关的细胞表型被拯救,令人惊讶的是,降低二肽重复(DPR)蛋白的水平和扩大的含有G4C2重复的RNA。用EXOC2反义寡核苷酸处理完全分化的C9ORF72神经元还减少了扩增的含有G4C2重复的RNA和部分挽救的疾病表型。这些结果表明,EXOC2直接或间接调节含有G4C2重复序列的RNA的水平,使其成为C9ORF72-ALS/FTD的潜在治疗靶标。
    GGGGCC (G4C2) repeat expansion in C9ORF72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). How this genetic mutation leads to neurodegeneration remains largely unknown. Using CRISPR-Cas9 technology, we deleted EXOC2, which encodes an essential exocyst subunit, in induced pluripotent stem cells (iPSCs) derived from C9ORF72-ALS/FTD patients. These cells are viable owing to the presence of truncated EXOC2, suggesting that exocyst function is partially maintained. Several disease-relevant cellular phenotypes in C9ORF72 iPSC-derived motor neurons are rescued due to, surprisingly, the decreased levels of dipeptide repeat (DPR) proteins and expanded G4C2 repeats-containing RNA. The treatment of fully differentiated C9ORF72 neurons with EXOC2 antisense oligonucleotides also decreases expanded G4C2 repeats-containing RNA and partially rescued disease phenotypes. These results indicate that EXOC2 directly or indirectly regulates the level of G4C2 repeats-containing RNA, making it a potential therapeutic target in C9ORF72-ALS/FTD.
    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
    罕见和未诊断的疾病大大降低了患者的生活质量,并日益成为医疗保健系统的沉重负担。由于在致病基因鉴定和机制阐明方面的挑战,患者通常面临难以获得精确的诊断和治疗。由于测序和多组学分析方法与患者来源的iPSC模型和基因编辑平台相结合的进步,在罕见和未诊断疾病的诊断和治疗方面取得了实质性进展。上述技术也为未来的精准医学研究提供了操作基础。在这次审查中,我们总结了基于GWAS/WES/WGS指导的多组学分析方法鉴定致病基因的最新进展.此外,我们用最先进的iPSC和类器官模型讨论疾病致病机制和治疗的最新进展,通过细胞成熟水平和基因编辑技术提高。上述综合策略将产生一种新的疾病分类范式,将显著提高罕见和未诊断疾病的诊断和治疗的精度和效率。
    Rare and undiagnosed diseases substantially decrease patient quality of life and have increasingly become a heavy burden on healthcare systems. Because of the challenges in disease-causing gene identification and mechanism elucidation, patients are often confronted with difficulty obtaining a precise diagnosis and treatment. Due to advances in sequencing and multiomics analysis approaches combined with patient-derived iPSC models and gene-editing platforms, substantial progress has been made in the diagnosis and treatment of rare and undiagnosed diseases. The aforementioned techniques also provide an operational basis for future precision medicine studies. In this review, we summarize recent progress in identifying disease-causing genes based on GWAS/WES/WGS-guided multiomics analysis approaches. In addition, we discuss recent advances in the elucidation of pathogenic mechanisms and treatment of diseases with state-of-the-art iPSC and organoid models, which are improved by cell maturation level and gene editing technology. The comprehensive strategies described above will generate a new paradigm of disease classification that will significantly promote the precision and efficiency of diagnosis and treatment for rare and undiagnosed diseases.
    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
    基于细胞的疗法对中风后的大脑修复大有希望。虽然越来越多的证据证实了细胞疗法的临床前和临床益处,它们促进大脑修复的潜在机制仍不清楚。这里,我们简要回顾了缺血性卒中后脑修复的内源性机制,然后重点介绍了不同干细胞和祖细胞来源如何促进脑修复。具体来说,我们研究了移植的细胞移植物如何通过直接细胞置换或刺激内源性修复途径促进功能恢复.此外,我们讨论了最近实施的临床前改进方法,如预处理,微载体,遗传安全开关,和普遍(免疫规避)细胞移植,以及这些药物和基因操作的治疗潜力,以进一步提高细胞疗法的疗效和安全性。通过对缺血后修复机制的深入了解,前瞻性临床试验可能会进一步完善,以将卒中后细胞治疗推向临床.
    Cell-based therapies hold great promise for brain repair after stroke. While accumulating evidence confirms the preclinical and clinical benefits of cell therapies, the underlying mechanisms by which they promote brain repair remain unclear. Here, we briefly review endogenous mechanisms of brain repair after ischemic stroke and then focus on how different stem and progenitor cell sources can promote brain repair. Specifically, we examine how transplanted cell grafts contribute to improved functional recovery either through direct cell replacement or by stimulating endogenous repair pathways. Additionally, we discuss recently implemented preclinical refinement methods, such as preconditioning, microcarriers, genetic safety switches, and universal (immune evasive) cell transplants, as well as the therapeutic potential of these pharmacologic and genetic manipulations to further enhance the efficacy and safety of cell therapies. By gaining a deeper understanding of post-ischemic repair mechanisms, prospective clinical trials may be further refined to advance post-stroke cell therapy to the clinic.
    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