VSX2

VSX2
  • 文章类型: Journal Article
    视觉系统Homeobox2(Vsx2)是在发育中的视网膜中表达的转录因子,调节组织身份,增长,和命运的决心。Vsx2基因在小鼠中存在几种突变,包括最初在人类中发现的自发无义突变和两个有针对性的错义突变。这里,我们扩展了遗传库,以包括设计用于表达β-半乳糖苷酶(b-GAL)并同时破坏Vsx2功能(敲入/敲除)的LacZ报道等位基因(Vsx2LacZ)。b-GAL的视网膜表达模式与VSX2一致,突变等位基因是隐性的。Vsx2LacZ纯合子小鼠患有先天性双侧小眼,伴有视网膜发育缺陷,包括非视网膜基因的异位表达,减少扩散,延迟的神经发生,组织形态异常,和缺乏双极中间神经元-Vsx2功能丧失的所有标志。出乎意料的是,突变体VSX2蛋白稳定表达,与空突变体相比,眼睛大小和早期视网膜神经发生有细微的差异,眼发育迟滞J.我们建议来自Vsx2LacZ等位基因的b-GAL表达是VSX2表达的可靠报道者,并且该等位基因表现出功能丧失特征。然而,突变型VSX2蛋白的持久性加上与无效表型的细微偏差,使Vsx2LacZ等位基因没有完全敲除的可能性。Vsx2LacZ等位基因添加到用于理解Vsx2功能的遗传工具包中。
    Visual System Homeobox 2 (Vsx2) is a transcription factor expressed in the developing retina that regulates tissue identity, growth, and fate determination. Several mutations in the Vsx2 gene exist in mice, including a spontaneous nonsense mutation and two targeted missense mutations originally identified in humans. Here, we expand the genetic repertoire to include a LacZ reporter allele (Vsx2 LacZ ) designed to express beta-Galactosidase (b-GAL) and simultaneously disrupt Vsx2 function (knock-in/knock-out). The retinal expression pattern of b-GAL is concordant with VSX2, and the mutant allele is recessive. Vsx2 LacZ homozygous mice have congenital bilateral microphthalmia accompanied by defects in retinal development including ectopic expression of non-retinal genes, reduced proliferation, delayed neurogenesis, aberrant tissue morphology, and an absence of bipolar interneurons - all hallmarks of Vsx2 loss-of-function. Unexpectedly, the mutant VSX2 protein is stably expressed, and there are subtle differences in eye size and early retinal neurogenesis when compared to the null mutant, ocular retardation J. We propose that b-GAL expression from the Vsx2 LacZ allele is a reliable reporter of VSX2 expression and that the allele exhibits loss-of-function characteristics. However, the perdurance of the mutant VSX2 protein combined with subtle deviations from the null phenotype leaves open the possibility that Vsx2 LacZ allele is not a complete knock-out. The Vsx2 LacZ allele adds to the genetic toolkit for understanding Vsx2 function.
    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
    视网膜发生涉及早期脊椎动物发育过程中视网膜细胞类型的规范。虽然模型生物对于确定动态染色质和细胞类型特异性转录网络在此过程中的作用至关重要,由于对人类胎儿组织的需求,对发育中的人类视网膜的理解更加难以捉摸。多能干细胞(PSC)衍生的视网膜类器官为研究发育中的人类视网膜提供了实验上可获得的解决方案。为了研究发育早期视网膜类器官的细胞和分子变化,我们开发了SIX6-GFP和VSX2-tdTomato(或VSX2-h2b-mRuby3)双荧光报告基因。当分化为3D类器官时,这些分别在第15天表达GFP和在第25天表达tdTomato(或mRuby3)。这使我们能够使用RNA-seq和ATAC-seq从多能性到早期视网膜规格探索转录和染色质相关的变化。发育中的类器官的通路分析显示多能性的逐步丧失,而视神经囊泡和视网膜通路变得越来越普遍。相关的基因转录与染色质可及性在早期的眼场发育表明,视网膜细胞经历了一个明显的变化,在染色质景观,以及基因表达谱。虽然每个数据集单独提供了有价值的信息,同时考虑这两者提供了对分子性质眼睛发展的有益了解。
    Retinogenesis involves the specification of retinal cell types during early vertebrate development. While model organisms have been critical for determining the role of dynamic chromatin and cell-type specific transcriptional networks during this process, an enhanced understanding of the developing human retina has been more elusive due to the requirement for human fetal tissue. Pluripotent stem cell (PSC) derived retinal organoids offer an experimentally accessible solution for investigating the developing human retina. To investigate cellular and molecular changes in developing early retinal organoids, we developed SIX6-GFP and VSX2-tdTomato (or VSX2-h2b-mRuby3) dual fluorescent reporters. When differentiated as 3D organoids these expressed GFP at day 15 and tdTomato (or mRuby3) at day 25, respectively. This enabled us to explore transcriptional and chromatin related changes using RNA-seq and ATAC-seq from pluripotency through early retina specification. Pathway analysis of developing organoids revealed a stepwise loss of pluripotency, while optic vesicle and retina pathways became progressively more prevalent. Correlating gene transcription with chromatin accessibility in early eye field development showed that retinal cells underwent a clear change in chromatin landscape, as well as gene expression profiles. While each dataset alone provided valuable information, considering both in parallel provided an informative glimpse into the molecular nature eye development.
    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
    Vsx2是视网膜增殖和双极细胞分化所必需的转录因子,但其发育作用的分子机制尚不清楚。这里,我们对小鼠视网膜发生过程中的VSX2基因组占有率进行了分析,揭示了在发育过程中与VSX2相关的广泛的视网膜遗传程序。VSX2结合并反式激活其与转录因子PAX6相关的增强子。在Vsx2调节景观中带有缺失的小鼠在视网膜增殖和双极细胞分化中表现出特定的异常。在其中一个删除中,双极细胞的完全丧失与光感受器产生的偏向有关。VSX2在成年小鼠和人类视网膜中占据与光感受器分化和稳态相关的基因附近的顺式调节元件,包括Prdm1附近的保守区域,这与视杆光感受器的规格和双极细胞命运的抑制有关。VSX2与转录因子OTX2相互作用,可以抑制Prdm1增强子的OTX2依赖性增强子反式激活。一起来看,我们的分析表明,Vsx2表达可以在增强子水平上在时间和空间上解耦,他们阐明了VSX2如何参与视网膜增殖和细胞命运获取所必需的基因调控网络的重要机制见解。
    Vsx2 is a transcription factor essential for retinal proliferation and bipolar cell differentiation, but the molecular mechanisms underlying its developmental roles are unclear. Here, we have profiled VSX2 genomic occupancy during mouse retinogenesis, revealing extensive retinal genetic programs associated with VSX2 during development. VSX2 binds and transactivates its enhancer in association with the transcription factor PAX6. Mice harboring deletions in the Vsx2 regulatory landscape exhibit specific abnormalities in retinal proliferation and in bipolar cell differentiation. In one of those deletions, a complete loss of bipolar cells is associated with a bias towards photoreceptor production. VSX2 occupies cis-regulatory elements nearby genes associated with photoreceptor differentiation and homeostasis in the adult mouse and human retina, including a conserved region nearby Prdm1, a factor implicated in the specification of rod photoreceptors and suppression of bipolar cell fate. VSX2 interacts with the transcription factor OTX2 and can act to suppress OTX2-dependent enhancer transactivation of the Prdm1 enhancer. Taken together, our analyses indicate that Vsx2 expression can be temporally and spatially uncoupled at the enhancer level, and they illuminate important mechanistic insights into how VSX2 is engaged with gene regulatory networks that are essential for retinal proliferation and cell fate acquisition.
    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
    动物模型为眼部发育和疾病提供了许多见解,但是它们对于理解人类的眼发育仍然不够理想。眼睛发育需要时空基因表达模式,人类和动物模型之间的疾病表型可能存在显着差异。在一些动物模型中报道了导致胚胎致死率的患者相关突变。人类诱导多能干细胞(hiPSC)技术的出现为通过生成三维(3D)细胞模型来解剖早期眼睛形态发生的复杂性质提供了新的资源。通过使用患者特异性hiPSCs来产生体外视神经泡样模型,我们可以加强对早期发育性眼疾的了解,并为疾病建模和治疗测试提供临床前平台。体外光学囊泡生成的主要挑战是3D培养中分化的低效率。为了解决这个问题,我们使用微孔板调整了以前发表的视网膜类器官分化方案,以改善胚状体的形成.已建立的形态学,已知的早期眼场转录因子的转录水平上调和标准视网膜祖细胞标志物的蛋白质表达证实了在培养的第20天至第50天之间的体外模型的视囊泡/假定视杯身份。这种适应的方案与寻求早期人类眼部发育和疾病的生理相关模型以取代动物模型的研究人员有关。
    Animal models have provided many insights into ocular development and disease, but they remain suboptimal for understanding human oculogenesis. Eye development requires spatiotemporal gene expression patterns and disease phenotypes can differ significantly between humans and animal models, with patient-associated mutations causing embryonic lethality reported in some animal models. The emergence of human induced pluripotent stem cell (hiPSC) technology has provided a new resource for dissecting the complex nature of early eye morphogenesis through the generation of three-dimensional (3D) cellular models. By using patient-specific hiPSCs to generate in vitro optic vesicle-like models, we can enhance the understanding of early developmental eye disorders and provide a pre-clinical platform for disease modelling and therapeutics testing. A major challenge of in vitro optic vesicle generation is the low efficiency of differentiation in 3D cultures. To address this, we adapted a previously published protocol of retinal organoid differentiation to improve embryoid body formation using a microwell plate. Established morphology, upregulated transcript levels of known early eye-field transcription factors and protein expression of standard retinal progenitor markers confirmed the optic vesicle/presumptive optic cup identity of in vitro models between day 20 and 50 of culture. This adapted protocol is relevant to researchers seeking a physiologically relevant model of early human ocular development and disease with a view to replacing animal models.
    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
    Approximately one-third of childhood blindness is attributed to developmental eye disorders, of which 80% have a genetic cause. Eye morphogenesis is tightly regulated by a highly conserved network of transcription factors when disrupted by genetic mutations can result in severe ocular malformation. Human-induced pluripotent stem cells (hiPSCs) are an attractive tool to study early eye development as they are more physiologically relevant than animal models, can be patient-specific and their use does not elicit the ethical concerns associated with human embryonic stem cells. The generation of self-organizing hiPSC-derived optic cups is a major advancement to understanding mechanisms of ocular development and disease. Their development in vitro has been found to mirror that of the human eye and these early organoids have been used to effectively model microphthalmia caused by a VSX2 variant. hiPSC-derived optic cups, retina, and cornea organoids are powerful tools for future modeling of disease phenotypes and will enable a greater understanding of the pathophysiology of many other developmental eye disorders. These models will also provide an effective platform for identifying molecular therapeutic targets and for future clinical applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    在哺乳动物眼睛发育的早期,VSX2,BRN3b,RCVRN表达标记神经视网膜祖细胞(NRP),视网膜神经节细胞,和光感受器(PR),分别。从人类诱导的多能干细胞(hiPSC)产生视网膜类器官的能力具有模拟人类视网膜发育和视网膜疾病的巨大潜力。然而,没有方法允许同时,目前存在对发育过程中多种特定视网膜细胞类型的实时监测。
    在hiPSC中CRISPR/Cas9介导的同源定向修复(HDR)促进了VSX2(祖细胞)的替换,BRN3b(神经节),和RCVRN(光感受器)终止密码子,其序列编码与Cerulean融合的病毒P2A肽,绿色荧光蛋白,和mCherry报告基因,分别,以产生称为PGP1的三重转基因报道HiPSC系。这是通过将HDR模板和sgRNA/Cas9载体共电穿孔到hiPSC中,然后进行抗生素选择来实现的。PGP1hiPSC系的功能验证包括产生视网膜类器官的能力,所有主要的视网膜细胞类型,显示与靶基因表达的起始一致的三个荧光报告基因的表达。还通过荧光激活的细胞分选分析了解聚的类器官,并测试了荧光群体的靶向基因的表达。
    从PGP1系形成的视网膜类器官表达与NRP分化一致的适当荧光蛋白,RGC,和PR。从PGP1系产生的类器官表达的转录本与所有主要视网膜细胞类型的发展一致。
    PGP1系列提供了一个强大的新工具来研究视网膜发育,视网膜重新编程,和治疗药物筛选。
    Early in mammalian eye development, VSX2, BRN3b, and RCVRN expression marks neural retinal progenitors (NRPs), retinal ganglion cells (RGCs), and photoreceptors (PRs), respectively. The ability to create retinal organoids from human induced pluripotent stem cells (hiPSC) holds great potential for modeling both human retinal development and retinal disease. However, no methods allowing the simultaneous, real-time monitoring of multiple specific retinal cell types during development currently exist.
    CRISPR/Cas9-mediated homology-directed repair (HDR) in hiPSCs facilitated the replacement of the VSX2 (Progenitor), BRN3b (Ganglion), and RCVRN (Photoreceptor) stop codons with sequences encoding a viral P2A peptide fused to Cerulean, green fluorescent protein, and mCherry reporter genes, respectively, to generate a triple transgenic reporter hiPSC line called PGP1. This was accomplished by co-electroporating HDR templates and sgRNA/Cas9 vectors into hiPSCs followed by antibiotic selection. Functional validation of the PGP1 hiPSC line included the ability to generate retinal organoids, with all major retinal cell types, displaying the expression of the three fluorescent reporters consistent with the onset of target gene expression. Disaggregated organoids were also analyzed by fluorescence-activated cell sorting and fluorescent populations were tested for the expression of the targeted gene.
    Retinal organoids formed from the PGP1 line expressed appropriate fluorescent proteins consistent with the differentiation of NRPs, RGCs, and PRs. Organoids produced from the PGP1 line expressed transcripts consistent with the development of all major retinal cell types.
    The PGP1 line offers a powerful new tool to study retinal development, retinal reprogramming, and therapeutic drug screening.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    The transcription factors Prdm1 (Blimp1) and Vsx2 (Chx10) work downstream of Otx2 to regulate photoreceptor and bipolar cell fates in the developing retina. Mice that lack Vsx2 fail to form bipolar cells while Prdm1 mutants form excess bipolars at the direct expense of photoreceptors. Excess bipolars in Prdm1 mutants appear to derive from rods, suggesting that photoreceptor fate remains mutable for some time after cells become specified. Here we tested whether bipolar cell fate is also plastic during development. To do this, we created a system to conditionally misexpress Prdm1 at different stages of bipolar cell development. We found that Prdm1 blocks bipolar cell formation if expressed before the fate choice decision occurred. When we misexpressed Prdm1 just after the decision to become a bipolar cell was made, some cells were reprogrammed into photoreceptors. In contrast, Prdm1 misexpression in mature bipolar cells did not affect cell fate. We also provide evidence that sustained misexpression of Prdm1 was selectively toxic to photoreceptors. Our data show that bipolar fate is malleable, but only for a short temporal window following fate specification. Prdm1 and Vsx2 act by stabilizing photoreceptor and bipolar fates in developing OTX2+ cells of the retina.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    转录因子OTX2是视网膜中感光细胞和双极细胞形成所必需的。它通过细胞类型特异性增强子直接激活转录因子Prdm1和Vsx2。PRDM1和VSX2是对立的,这样PRDM1促进光感受器命运和VSX2双极细胞命运。为了确定OTX2+细胞命运在小鼠中的调控,我们使用CRISPR/Cas9体内视网膜电穿孔策略同时删除了Prdm1和Vsx2或其细胞类型特异性增强子.双基因或增强子靶向有效去除PRDM1和VSX2蛋白表达。然而,双增强子靶向有利于双相命运的结果,而双基因打靶有利于光感受器的命运。两种条件都产生过量的无长突细胞。合并,这些命运变化表明,光感受器是OTX2+细胞的默认命运结果,VSX2必须存在于狭窄的时间窗口中以驱动双极细胞形成.Prdm1和Vsx2似乎也冗余地限制了OTX2+小区的能力,防止无长突细胞形成。通过采取编码序列和增强子的组合缺失方法,我们的工作为控制细胞命运选择的复杂调节机制提供了新的见解.
    The transcription factor OTX2 is required for photoreceptor and bipolar cell formation in the retina. It directly activates the transcription factors Prdm1 and Vsx2 through cell type-specific enhancers. PRDM1 and VSX2 work in opposition, such that PRDM1 promotes photoreceptor fate and VSX2 bipolar cell fate. To determine how OTX2+ cell fates are regulated in mice, we deleted Prdm1 and Vsx2 or their cell type-specific enhancers simultaneously using a CRISPR/Cas9 in vivo retina electroporation strategy. Double gene or enhancer targeting effectively removed PRDM1 and VSX2 protein expression. However, double enhancer targeting favored bipolar fate outcomes, whereas double gene targeting favored photoreceptor fate. Both conditions generated excess amacrine cells. Combined, these fate changes suggest that photoreceptors are a default fate outcome in OTX2+ cells and that VSX2 must be present in a narrow temporal window to drive bipolar cell formation. Prdm1 and Vsx2 also appear to redundantly restrict the competence of OTX2+ cells, preventing amacrine cell formation. By taking a combinatorial deletion approach of both coding sequences and enhancers, our work provides new insights into the complex regulatory mechanisms that control cell fate choice.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Anophthalmia and microphthalmia (A/M) are rare distinct phenotypes that represent a continuum of structural developmental eye defects. Here, we describe three probands from an Egyptian population with various forms of A/M: two patients with bilateral anophthalmia and one with bilateral microphthalmia that were investigated using whole exome sequencing (WES). We identified three causative mutations in three different genes. A new homozygous frameshift mutation c.[422delA];[422delA], p.[N141Ifs∗19];[N141Ifs∗19] in VSX2 was identified in a patient showing bilateral anophthalmia. A previously reported SOX2 deletion c.[70_89del20] p.[N24Rfs∗65];[=] was found in one subject with bilateral anophthalmia. A novel homozygous in-frame mutation c.[431_433delACT];[431_433delACT], p.[Y144del]; [Y144del]) in FOXE3 was identified in a patient with severe bilateral microphthalmia and anterior segment dysgenesis. This study shows that whole exome sequencing (WES) is a reliable and effective strategy for the molecular diagnosis of A/M. Our results expand its allelic heterogeneity and highlight the need for the testing of patient with this developmental anomaly.
    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
    超过8,000个基因被打开或关闭,因为祖细胞在发育过程中产生7类视网膜细胞类型。成千上万的增强子也活跃在发育中的视网膜中,许多具有细胞和发育阶段特定活动的特征。我们通过对鼠视网膜进行超深度原位Hi-C分析,研究了3D染色质景观的动态变化,这对于视网膜发育过程中基因表达的精确编排变化很重要。我们确定了染色质区室和增强子-启动子相互作用的发育阶段特异性变化。我们开发了一种基于机器学习的算法来映射全基因组的常染色质和异染色质域,并用Hi-C识别的染色质区室覆盖它。将单细胞ATAC-seq和RNA-seq与我们的Hi-C和先前的ChIP-seq数据整合,以鉴定细胞和发育阶段特异性超增强子(SE)。我们确定了Vsx2上游的双极神经元特异性核心调节回路SE,其在小鼠中的缺失导致双极神经元的丢失。
    More than 8,000 genes are turned on or off as progenitor cells produce the 7 classes of retinal cell types during development. Thousands of enhancers are also active in the developing retinae, many having features of cell- and developmental stage-specific activity. We studied dynamic changes in the 3D chromatin landscape important for precisely orchestrated changes in gene expression during retinal development by ultra-deep in situ Hi-C analysis on murine retinae. We identified developmental-stage-specific changes in chromatin compartments and enhancer-promoter interactions. We developed a machine learning-based algorithm to map euchromatin and heterochromatin domains genome-wide and overlaid it with chromatin compartments identified by Hi-C. Single-cell ATAC-seq and RNA-seq were integrated with our Hi-C and previous ChIP-seq data to identify cell- and developmental-stage-specific super-enhancers (SEs). We identified a bipolar neuron-specific core regulatory circuit SE upstream of Vsx2, whose deletion in mice led to the loss of bipolar neurons.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号