neuron differentiation

神经元分化
  • 文章类型: Journal Article
    ch12q13基因座是全基因组关联研究中确定的最重要的儿童肥胖基因座之一。该基因座位于FAIM2内的非编码区;因此,潜在的因果变异可能通过顺式调节影响疾病易感性。通过利用我们的内部3D基因组数据和公共领域数据集,我们将rs7132908暗示为推定的因果变体。使用荧光素酶报告基因测定法,我们观察到包含rs7132908的直接区域的等位基因特异性顺式调节活性。我们产生了rs7132908等位基因纯合的等基因人类胚胎干细胞系,以评估整个下丘脑神经元分化过程中基因表达和染色质可及性的变化。一种已知调节摄食行为的关键细胞类型。rs7132908肥胖风险等位基因影响了FAIM2和其他基因的表达,并降低了分化产生的神经元比例。我们已经在功能上验证了rs7132908是一种因果肥胖变体,它在时间上调节附近的效应基因并影响神经发育和存活。
    The ch12q13 locus is among the most significant childhood obesity loci identified in genome-wide association studies. This locus resides in a non-coding region within FAIM2; thus, the underlying causal variant(s) presumably influence disease susceptibility via cis-regulation. We implicated rs7132908 as a putative causal variant by leveraging our in-house 3D genomic data and public domain datasets. Using a luciferase reporter assay, we observed allele-specific cis-regulatory activity of the immediate region harboring rs7132908. We generated isogenic human embryonic stem cell lines homozygous for either rs7132908 allele to assess changes in gene expression and chromatin accessibility throughout a differentiation to hypothalamic neurons, a key cell type known to regulate feeding behavior. The rs7132908 obesity risk allele influenced expression of FAIM2 and other genes and decreased the proportion of neurons produced by differentiation. We have functionally validated rs7132908 as a causal obesity variant that temporally regulates nearby effector genes and influences neurodevelopment and survival.
    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
    脆性X综合征(FXS)是由FMR1基因的表观遗传沉默引起的神经系统疾病。FMR1的再激活是FXS的潜在治疗方法,可以纠正疾病的根本原因。这里,使用基于候选的shRNA筛选,我们确定了9种促进FXS细胞中FMR1沉默的表观遗传抑制因子(称为FMR1沉默因子,或FMR1-SF)。用shRNA或小分子抑制FMR1-SF在培养的未分化诱导多能干细胞中重新激活FMR1,来自FXS患者的神经祖细胞(NPC)和有丝分裂后神经元。FMR1-SF之一是组蛋白甲基转移酶EZH2,这是FDA批准的小分子抑制剂,EPZ6438(也称为tazemetostat),是可用的。我们表明EPZ6438基本上纠正了培养的FXS神经元的特征性分子和电生理异常。不幸的是,EZH2抑制剂不能有效地穿过血脑屏障,限制其对FXS的治疗用途。最近,基于反义寡核苷酸(ASO)的方法已被开发为某些中枢神经系统疾病的有效治疗选择。因此,我们获得了靶向EZH2的有效ASO,并证明它们重新激活FMR1表达并纠正培养的FXS神经元的分子和电生理异常,并重新激活移植在小鼠脑内的人FXSNPC中的FMR1表达。总的来说,我们的结果建立了EZH2抑制一般,特别是EZH2ASO,作为FXS的治疗方法。
    Fragile X Syndrome (FXS) is a neurological disorder caused by epigenetic silencing of the FMR1 gene. Reactivation of FMR1 is a potential therapeutic approach for FXS that would correct the root cause of the disease. Here, using a candidate-based shRNA screen, we identify nine epigenetic repressors that promote silencing of FMR1 in FXS cells (called FMR1 Silencing Factors, or FMR1- SFs). Inhibition of FMR1-SFs with shRNAs or small molecules reactivates FMR1 in cultured undifferentiated induced pluripotent stem cells, neural progenitor cells (NPCs) and post-mitotic neurons derived from FXS patients. One of the FMR1-SFs is the histone methyltransferase EZH2, for which an FDA-approved small molecule inhibitor, EPZ6438 (also known as tazemetostat), is available. We show that EPZ6438 substantially corrects the characteristic molecular and electrophysiological abnormalities of cultured FXS neurons. Unfortunately, EZH2 inhibitors do not efficiently cross the blood-brain barrier, limiting their therapeutic use for FXS. Recently, antisense oligonucleotide (ASO)-based approaches have been developed as effective treatment options for certain central nervous system disorders. We therefore derived efficacious ASOs targeting EZH2 and demonstrate that they reactivate FMR1 expression and correct molecular and electrophysiological abnormalities in cultured FXS neurons, and reactivate FMR1 expression in human FXS NPCs engrafted within the brains of mice. Collectively, our results establish EZH2 inhibition in general, and EZH2 ASOs in particular, as a therapeutic approach for FXS.
    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
    人诱导性多能干细胞(iPSCs)可以分化为神经元,提供活的人类神经元来模拟大脑疾病。然而,目前尚不清楚不同类型的分子如何在健康和疾病状态下共同调节干细胞和神经元生物学。在这项研究中,我们进行了综合蛋白质组学,脂质组学,和具有自信识别的代谢组学分析,精确量化,和可重复测量,以比较人类iPSC和iPSC衍生神经元的分子谱。蛋白质,脂质,和与有丝分裂有关的代谢物,DNA复制,多能性,鞘糖脂,iPSCs中能量代谢高度富集,而突触蛋白,神经递质,多不饱和脂肪酸,心磷脂,和轴突引导通路在神经元中高度富集。GRN基因的突变导致颗粒蛋白前体(PGRN)蛋白的缺乏,与各种神经退行性疾病相关。使用这个多组学平台,我们从全细胞水平评价了PGRN缺乏对iPSCs和神经元的影响.蛋白质组学,脂质组学,和代谢组学分析暗示PGRN在神经炎症中的作用,嘌呤代谢,和神经突生长,揭示与神经元投射相关的常见改变的通路,突触功能障碍,和大脑新陈代谢。多组学数据集也指出了相同的假设,即与iPSCs相比,神经元似乎更容易受到PGRN丢失的影响。与携带遗传GRN突变的患者的神经症状和认知障碍一致。
    Human induced pluripotent stem cells (iPSCs) can be differentiated into neurons, providing living human neurons to model brain diseases. However, it is unclear how different types of molecules work together to regulate stem cell and neuron biology in healthy and disease states. In this study, we conducted integrated proteomics, lipidomics, and metabolomics analyses with confident identification, accurate quantification, and reproducible measurements to compare the molecular profiles of human iPSCs and iPSC-derived neurons. Proteins, lipids, and metabolites related to mitosis, DNA replication, pluripotency, glycosphingolipids, and energy metabolism were highly enriched in iPSCs, whereas synaptic proteins, neurotransmitters, polyunsaturated fatty acids, cardiolipins, and axon guidance pathways were highly enriched in neurons. Mutations in the GRN gene lead to the deficiency of the progranulin (PGRN) protein, which has been associated with various neurodegenerative diseases. Using this multiomics platform, we evaluated the impact of PGRN deficiency on iPSCs and neurons at the whole-cell level. Proteomics, lipidomics, and metabolomics analyses implicated PGRN\'s roles in neuroinflammation, purine metabolism, and neurite outgrowth, revealing commonly altered pathways related to neuron projection, synaptic dysfunction, and brain metabolism. Multiomics data sets also pointed toward the same hypothesis that neurons seem to be more susceptible to PGRN loss compared to iPSCs, consistent with the neurological symptoms and cognitive impairment from patients carrying inherited GRN mutations.
    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:初级纤毛来自大多数人类细胞类型,包括神经元。纤毛对于与细胞的直接环境的通信是重要的:信号接收和向/从纤毛细胞的转导。纤毛信号的失调可导致纤毛病和某些神经发育障碍。在发育中的大脑纤毛在神经祖细胞池的扩增中起着有据可查的作用,而有关纤毛在有丝分裂后神经元分化和成熟过程中的作用的信息很少。
    结果:我们在时程实验中采用纤毛隆德人中脑(LUHMES)细胞来评估纤毛信号对神经元分化的影响。通过比较野生型和RFX2-/-突变神经元的纤毛和非纤毛神经元前体细胞和神经元,我们发现了一个早期分化的“纤毛时间窗”,在此期间,短暂的纤毛促进轴突生长,分枝和树根化。在具有IFT88和IFT172纤毛基因敲除的神经元中进行的实验,导致较短的纤毛,确认这些结果。纤毛通过将WNT信号传导向非规范途径促进神经元分化,进而激活WNT途径输出与细胞结构变化有关的基因。
    结论:我们提供了纤毛信号何时以及如何协调的机械切入点,促进和转化为解剖学的变化。我们假设纤毛改变导致神经元分化缺陷可能导致大脑发育的“轻度”损害,可能是神经发育障碍某些方面的基础。
    BACKGROUND: Primary cilia emanate from most human cell types, including neurons. Cilia are important for communicating with the cell\'s immediate environment: signal reception and transduction to/from the ciliated cell. Deregulation of ciliary signaling can lead to ciliopathies and certain neurodevelopmental disorders. In the developing brain cilia play well-documented roles for the expansion of the neural progenitor cell pool, while information about the roles of cilia during post-mitotic neuron differentiation and maturation is scarce.
    RESULTS: We employed ciliated Lund Human Mesencephalic (LUHMES) cells in time course experiments to assess the impact of ciliary signaling on neuron differentiation. By comparing ciliated and non-ciliated neuronal precursor cells and neurons in wild type and in RFX2 -/- mutant neurons with altered cilia, we discovered an early-differentiation \"ciliary time window\" during which transient cilia promote axon outgrowth, branching and arborization. Experiments in neurons with IFT88 and IFT172 ciliary gene knockdowns, leading to shorter cilia, confirm these results. Cilia promote neuron differentiation by tipping WNT signaling toward the non-canonical pathway, in turn activating WNT pathway output genes implicated in cyto-architectural changes.
    CONCLUSIONS: We provide a mechanistic entry point into when and how ciliary signaling coordinates, promotes and translates into anatomical changes. We hypothesize that ciliary alterations causing neuron differentiation defects may result in \"mild\" impairments of brain development, possibly underpinning certain aspects of neurodevelopmental disorders.
    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
    印迹基因簇是包含具有亲本来源依赖性转录活性的基因的受限基因组区域。在本期《基因与发育》中,Loftus及其同事(pp.XXX-XXX)使用了富有洞察力的描述性方法组合,遗传操作,和表观基因组编辑方法表明,核拓扑结构的差异先于Peg13-Kcnk9基因座印迹表达的开始。此外,研究者提供的数据与一个模型一致,该模型提示亲代来源特异性拓扑差异可能是亲代来源特异性增强子活性和印迹表达的原因.
    Imprinted gene clusters are confined genomic regions containing genes with parent-of-origin-dependent transcriptional activity. In this issue of Genes & Development, Loftus and colleagues (pp. 829-843) made use of an insightful combination of descriptive approaches, genetic manipulations, and epigenome-editing approaches to show that differences in nuclear topology precede the onset of imprinted expression at the Peg13-Kcnk9 locus. Furthermore, the investigators provide data in line with a model suggesting that parent-of-origin-specific topological differences could be responsible for parent-of-origin-specific enhancer activity and thus imprinted expression.
    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
    从亲本配子遗传的染色质状态的差异影响后代中母本和父本等位基因的调节。这种现象,被称为基因组印记,结果基因优先从一个亲本等位基因转录。虽然已知DNA甲基化等局部表观遗传因素对于建立印迹基因表达很重要,关于差异甲基化区域(DMRs)导致跨广泛染色质的等位基因表达差异的机制知之甚少。在多个印迹基因座上观察到等位基因特异性的高阶染色质结构,与在多个DMRs上观察到染色质组织因子CTCF的等位基因结合一致。然而,等位基因染色质结构是否影响等位基因基因的表达对于大多数印迹位点是未知的。在这里,我们描述了Peg13-Kcnk9基因座的脑特异性印迹表达的潜在机制,与智力残疾相关的印记区域。我们从相互杂交杂交中对小鼠大脑进行了区域捕获Hi-C,并发现了由CTCF与Peg13DMR的等位基因结合引起的印迹高阶染色质结构。使用体外神经元分化系统,我们表明印迹染色质结构先于印迹表达。此外,远端增强子的激活以等位基因染色质结构依赖性方式诱导Kcnk9的印迹表达。这项工作提供了印迹染色质结构的高分辨率图谱,并证明了在早期发育中建立的染色质状态可以促进分化后的印迹表达。
    Differences in chromatin state inherited from the parental gametes influence the regulation of maternal and paternal alleles in offspring. This phenomenon, known as genomic imprinting, results in genes preferentially transcribed from one parental allele. While local epigenetic factors such as DNA methylation are known to be important for the establishment of imprinted gene expression, less is known about the mechanisms by which differentially methylated regions (DMRs) lead to differences in allelic expression across broad stretches of chromatin. Allele-specific higher-order chromatin structure has been observed at multiple imprinted loci, consistent with the observation of allelic binding of the chromatin-organizing factor CTCF at multiple DMRs. However, whether allelic chromatin structure impacts allelic gene expression is not known for most imprinted loci. Here we characterize the mechanisms underlying brain-specific imprinted expression of the Peg13-Kcnk9 locus, an imprinted region associated with intellectual disability. We performed region capture Hi-C on mouse brains from reciprocal hybrid crosses and found imprinted higher-order chromatin structure caused by the allelic binding of CTCF to the Peg13 DMR. Using an in vitro neuron differentiation system, we showed that imprinted chromatin structure precedes imprinted expression at the locus. Additionally, activation of a distal enhancer induced imprinted expression of Kcnk9 in an allelic chromatin structure-dependent manner. This work provides a high-resolution map of imprinted chromatin structure and demonstrates that chromatin state established in early development can promote imprinted expression upon differentiation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    ch12q13肥胖位点是全基因组关联研究中确定的最重要的儿童肥胖位点之一。该基因座位于FAIM2内的非编码区;因此,潜在的因果变异可能通过影响基因组区域内的顺式调节来影响疾病易感性。我们利用我们内部3D基因组数据的组合,将rs7132908暗示为该位点的推定因果变异,公共领域数据集,和几种计算方法。在人原代星形胶质细胞中使用荧光素酶报告基因测定法,我们观察到包含rs7132908的直接区域的等位基因特异性顺式调节活性。受这一发现的激励,我们继续产生具有CRISPR-Cas9同源定向修复的rs7132908等位基因纯合的等基因人类胚胎干细胞系,以评估由于基因型和染色质可及性在整个分化为下丘脑神经元的基因表达变化。一种已知调节摄食行为的关键细胞类型。我们观察到rs7132908肥胖风险等位基因与其他基因一起影响FAIM2的表达,减少分化过程中产生的神经元的比例,上调的细胞死亡基因集,和相反下调的神经元分化基因集。因此,我们在功能上验证了rs7132908是一种因果肥胖变体,它在时间上调节ch12q13基因座附近的效应基因并影响神经发育和存活。
    The ch12q13 obesity locus is among the most significant childhood obesity loci identified in genome-wide association studies. This locus resides in a non-coding region within FAIM2; thus, the underlying causal variant(s) presumably influence disease susceptibility via an influence on cis-regulation within the genomic region. We implicated rs7132908 as a putative causal variant at this locus leveraging a combination of our inhouse 3D genomic data, public domain datasets, and several computational approaches. Using a luciferase reporter assay in human primary astrocytes, we observed allele-specific cis-regulatory activity of the immediate region harboring rs7132908. Motivated by this finding, we went on to generate isogenic human embryonic stem cell lines homozygous for either rs7132908 allele with CRISPR-Cas9 homology-directed repair to assess changes in gene expression due to genotype and chromatin accessibility throughout a differentiation to hypothalamic neurons, a key cell type known to regulate feeding behavior. We observed that the rs7132908 obesity risk allele influenced the expression of FAIM2 along with other genes, decreased the proportion of neurons produced during differentiation, up-regulated cell death gene sets, and conversely down-regulated neuron differentiation gene sets. We have therefore functionally validated rs7132908 as a causal obesity variant which temporally regulates nearby effector genes at the ch12q13 locus and influences neurodevelopment and survival.
    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:对神经再生分子机制的研究导致发现了几种在成功的神经再生过程中诱导的蛋白质。RICH蛋白被鉴定为硬骨鱼视神经再生过程中诱导的蛋白。这些蛋白质是2',3\'-环核苷酸,可通过羧基末端膜定位域与细胞膜结合的3'-磷酸二酯酶。它们与微管蛋白细胞骨架相互作用,并能够通过促进神经突分支的形成来增强神经元结构可塑性。
    结果:产生了表达融合蛋白的PC12稳定转染子细胞,该融合蛋白结合了红色荧光蛋白和无催化活性的斑马鱼RICH蛋白突变体。这些细胞用作模型以分析蛋白质对神经突发生的影响。分化实验表明,次生神经突的形成增加了2.9倍,分支点增加了2.4倍。在神经突再生测定中观察到次级神经突的形成增加2.2倍。
    结论:使用荧光融合蛋白促进了表达水平的检测。两种计算机辅助形态分析方法表明,无催化活性的RICH蛋白在分化和神经突再生过程中均诱导分支点和次级神经突的形成。基于随机场图像分析的程序提供了与经典神经突追踪方法相当的结果。
    Studies of the molecular mechanisms of nerve regeneration have led to the discovery of several proteins that are induced during successful nerve regeneration. RICH proteins were identified as proteins induced during the regeneration of the optic nerve of teleost fish. These proteins are 2\',3\'-cyclic nucleotide, 3\'-phosphodiesterases that can bind to cellular membranes through a carboxy-terminal membrane localization domain. They interact with the tubulin cytoskeleton and are able to enhance neuronal structural plasticity by promoting the formation of neurite branches.
    PC12 stable transfectant cells expressing a fusion protein combining a red fluorescent protein with a catalytically inactive mutant version of zebrafish RICH protein were generated. These cells were used as a model to analyze effects of the protein on neuritogenesis. Differentiation experiments showed a 2.9 fold increase in formation of secondary neurites and a 2.4 fold increase in branching points. A 2.2 fold increase in formation of secondary neurites was observed in neurite regeneration assays.
    The use of a fluorescent fusion protein facilitated detection of expression levels. Two computer-assisted morphometric analysis methods indicated that the catalytically inactive RICH protein induced the formation of branching points and secondary neurites both during differentiation and neurite regeneration. A procedure based on analysis of random field images provided comparable results to classic neurite tracing methods.
    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
    胎儿酒精综合症(FAS)影响很大一部分,超过90%,受苦的孩子,导致严重的眼像差,如小眼症和视神经发育不全。在怀孕的早期阶段,神经视网膜神经发生的开始代表了人眼发育的关键时期,由于缺乏意识,同时使发育中的视网膜结构暴露于最高的产前乙醇暴露风险。尽管这段时期至关重要,短期乙醇暴露对人类神经视网膜发育过程的确切影响和潜在机制在很大程度上仍然难以捉摸。在这项研究中,我们利用人类胚胎干细胞衍生的视网膜类器官(hRO)来概述最初的视网膜神经发生,并发现1%(v/v)乙醇通过诱导强烈的细胞死亡和视网膜神经节细胞分化缺陷来减缓hRO的生长。BulkRNA-seq分析和双光子显微镜活体钙成像揭示了由乙醇诱导的RYR1和CACNA1S下调引起的钙信号动力学改变。此外,钙结合蛋白RET,钙信号通路的下游效应基因之一,协同整合乙醇和钙信号以中止神经元分化并导致细胞死亡。总而言之,我们的研究说明了乙醇对人类胚胎神经视网膜初始神经发生的影响和分子机制,为FAS的眼部表型提供了新的解释,并为易感人群提供了潜在的预防措施。
    Fetal Alcohol Syndrome (FAS) affects a significant proportion, exceeding 90%, of afflicted children, leading to severe ocular aberrations such as microphthalmia and optic nerve hypoplasia. During the early stages of pregnancy, the commencement of neural retina neurogenesis represents a critical period for human eye development, concurrently exposing the developing retinal structures to the highest risk of prenatal ethanol exposure due to a lack of awareness. Despite the paramount importance of this period, the precise influence and underlying mechanisms of short-term ethanol exposure on the developmental process of the human neural retina have remained largely elusive. In this study, we utilize the human embryonic stem cells derived retinal organoids (hROs) to recapitulate the initial retinal neurogenesis and find that 1% (v/v) ethanol slows the growth of hROs by inducing robust cell death and retinal ganglion cell differentiation defect. Bulk RNA-seq analysis and two-photon microscope live calcium imaging reveal altered calcium signaling dynamics derived from ethanol-induced down-regulation of RYR1 and CACNA1S. Moreover, the calcium-binding protein RET, one of the downstream effector genes of the calcium signaling pathway, synergistically integrates ethanol and calcium signals to abort neuron differentiation and cause cell death. To sum up, our study illustrates the effect and molecular mechanism of ethanol on the initial neurogenesis of the human embryonic neural retina, providing a novel interpretation of the ocular phenotype of FAS and potentially informing preventative measures for susceptible populations.
    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: Preprint
    从亲本配子遗传的染色质状态的差异影响后代中母本和父本等位基因的调节。这种现象,被称为基因组印记,结果基因优先从一个亲本等位基因转录。虽然已知DNA甲基化等局部表观遗传因素对于建立印迹基因表达很重要,关于差异甲基化区域(DMRs)导致跨广泛染色质的等位基因表达差异的机制知之甚少。在多个印迹基因座上观察到等位基因特异性的高阶染色质结构,与在多个DMRs上观察到染色质组织因子CTCF的等位基因结合一致。然而,等位基因染色质结构是否影响等位基因基因的表达对于大多数印迹位点是未知的。在这里,我们描述了Peg13-Kcnk9基因座的脑特异性印迹表达的潜在机制,与智力残疾相关的印记区域。我们从相互杂交杂交中对小鼠大脑进行了区域捕获Hi-C,并发现了由CTCF与Peg13DMR的等位基因结合引起的印迹高阶染色质结构。使用体外神经元分化系统,我们表明,在发育早期形成的母体等位基因增强子-启动子接触使大脑特异性钾泄漏通道Kcnk9在神经发生之前用于母体表达。相比之下,这些增强子-启动子接触被父系等位基因上的CTCF阻断,防止父系Kcnk9激活。这项工作提供了印迹染色质结构的高分辨率图谱,并证明了在早期发育中建立的染色质状态可以促进分化后的印迹表达。
    Differences in chromatin state inherited from the parental gametes influence the regulation of maternal and paternal alleles in offspring. This phenomenon, known as genomic imprinting, results in genes preferentially transcribed from one parental allele. While local epigenetic factors such as DNA methylation are known to be important for the establishment of imprinted gene expression, less is known about the mechanisms by which differentially methylated regions (DMRs) lead to differences in allelic expression across broad stretches of chromatin. Allele-specific higher-order chromatin structure has been observed at multiple imprinted loci, consistent with the observation of allelic binding of the chromatin-organizing factor CTCF at multiple DMRs. However, whether allelic chromatin structure impacts allelic gene expression is not known for most imprinted loci. Here we characterize the mechanisms underlying brain-specific imprinted expression of the Peg13-Kcnk9 locus, an imprinted region associated with intellectual disability. We performed region capture Hi-C on mouse brain from reciprocal hybrid crosses and found imprinted higher-order chromatin structure caused by the allelic binding of CTCF to the Peg13 DMR. Using an in vitro neuron differentiation system, we show that on the maternal allele enhancer-promoter contacts formed early in development prime the brain-specific potassium leak channel Kcnk9 for maternal expression prior to neurogenesis. In contrast, these enhancer-promoter contacts are blocked by CTCF on the paternal allele, preventing paternal Kcnk9 activation. This work provides a high-resolution map of imprinted chromatin structure and demonstrates that chromatin state established in early development can promote imprinted expression upon differentiation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号